In today’s data-driven landscape, the efficient transfer of large datasets to and from Amazon Simple Storage Service (Amazon S3) is a critical piece of an enterprise’s cloud strategy Common business use cases that need frequent transferring of large data sets include cloud-based data lakes that depend on receiving data from various sources Amazon S3 can also serve as the starting point for your Generative AI journey Generative AI applications need large data sets and by transferring this data into Amazon S3 organizations can use the full suite of Amazon Web Services (AWS) artificial intelligence/machine learning (AI/ML) tools Once a model is trained in AWS using this data the model artifacts can also be stored in Amazon S3 Other use cases include backup and restore There are three general patterns when transferring data to and from Amazon S3: This post details three network architectures for setting up connectivity for the pattern using AWS Direct Connect These architectures differ in terms of the services used Understanding these network design options and their tradeoffs is crucial for organizations to optimize their cloud storage operations The following services are included in the architectures that are covered The following are brief descriptions of each one You can select the links to learn more about each service All three of the architectures that are covered use Direct Connect then you can configure a new VIF on the existing connection If you have a hosted connection that only supports one VIF then you must order an additional hosted connection to support an additional VIF If your plan is to have a landing zone with many VPCs and to provide access to AWS services and applications inside and outside of those VPCs, then a dedicated connection is recommended because it provides more flexibility with the network design. It is also recommended to have at least two connections for resiliency as covered in the AWS Direct Connect Resiliency Toolkit The Direct Connect charges include a port hours charge that is based on the type of connection and the capacity of the connection. There is also a charge based on how much data is transferred outbound from AWS to on-premises. Data that is transferred inbound from on-premises to AWS is free. For more details on pricing, refer to the Direct Connect pricing page Each architecture description includes a pricing estimate based on the following example scenario You have two 10 Gbps dedicated Direct Connect connections and you want to setup connectivity to Amazon S3 for data transfer from on-premises You transfer an estimated 4 TB of data per month into Amazon S3 You estimate that you must retrieve 2 TB of data per month from Amazon S3 and transfer it back to on-premises The following calculations are based on the AWS Pricing Calculator that can be used to run your own calculations based on your architecture and specific use case While all pricing shown in the following architecture examples is based on AWS Regions in the United States The AWS Pricing Calculator can be used to show pricing information for these other AWS Regions The following figure shows this architecture using two Direct Connect connections each with a public VIF connecting to the AWS network This architecture is a good choice if you want to minimize the data transfer costs related to transferring data into Amazon S3 it needs additional configuration for the public VIF because it exposes the on-premises network to the AWS public network thus you need to take this into consideration the costs associated with this architecture include the Direct Connect charges as shown in the following table The total estimated charges would be $6,610.96 USD a month the traffic is sent to Amazon S3 over the AWS network each with a Private VIF connecting through the Direct Connect Gateway to a VPC with an interface endpoint Figure 2: Private VIF through VPC interface endpoint This architecture is a good choice if you do not want to create a public VIF You may not want to add the additional configuration needed to filter the advertised Amazon prefixes You may have security concerns with connecting to the AWS public network especially if appropriate security services are not in-line with the Direct Connect connection You may already have a hosted connection with a Private VIF that you prefer to use rather than ordering another hosted connection for another VIF The costs associated with this architecture include the Direct Connect charges covered in Architecture 1 in addition to the interface endpoint charges shown in the following table The total estimated charges would be $6,610.96 USD + $90.64 USD = $6,701.60 USD per month This architecture uses a Transit VIF and the Transit Gateway A single Transit VIF can be used to connect to all VPCs through the Transit Gateway This avoids having to configure a separate Private VIF for each VPC which was common practice before the Transit Gateway service became available In this architecture a VPC with an interface endpoint is attached to the Transit Gateway which is connected back to on-premises using a Transit VIF Access to Amazon S3 through the interface endpoint would work as described in Architecture 2 with the exception being that traffic would flow over the Transit VIF to the interface endpoint each with a Transit VIF connecting through the Direct Connect Gateway to the same Transit Gateway that connects to a VPC with an interface endpoint Figure 3: Transit VIF through Transit Gateway and VPC interface endpoint If you already have a Transit Gateway in place then this architecture is the least complex from an implementation and operational standpoint and the most scalable It is a good choice if you do not want to create additional VIFs or already have a hosted connection with a VIF and do not want to order another hosted connection for another VIF It allows you to use a single Transit VIF to send data to VPCs and to Amazon S3 without configuring additional VIFs It requires configuring the Transit Gateway and all of the attachments along with Transit Gateway routing tables The costs associated with this architecture include the Direct Connect charges and interface endpoint charges covered in Architectures 1 and 2 in addition to the Transit Gateway charges as shown in the following table The total estimated charges would be $6,610.98 USD + $90.64 USD + $318.76 USD= $7,020.38 USD per month This post discussed different network architectures for transferring large datasets between on-premises environments and Amazon S3 using AWS Direct Connect By understanding these architecture options and Transit VIFs through the Transit Gateway in addition to the implications of using each service from a cost and configuration complexity standpoint you can choose the right design for your organization The choice is influenced by a number of things If you are a new user that is building a landing zone for the first time, then you can reference the Hybrid Network Connectivity Whitepaper to learn more about the considerations for choosing the right connectivity type and connectivity design 2024: An earlier version of this post incorrectly stated that AWS IP address ranges from ip-ranges.json can be used for BGP prefix filtering Architecture 3 has been updated to include multiple interface endpoints for a more resilient architecture The amount of outbound traffic has been adjusted to reflect a more realistic example.  Chuck is a Senior Solutions Architect at AWS with a background in Network Engineering he works with customers to design and build innovative resilient and cost-effective solutions in the AWS Cloud ensuring success in their cloud adoption journeys Metrics details HIV-1 has well-established mechanisms to disrupt essential pathways in people with HIV diversity of the amino acid sequences in fundamental HIV-1 proteins including Tat and Vif have been linked to dysregulating these pathways and subsequently influencing clinical outcomes in people with HIV the relationship between Tat and Vif amino acid sequence variation and specific immune markers and metabolites of the tryptophan-kynurenine (Trp-Kyn) pathway remains unclear this study aimed to investigate the relationship between Tat/Vif amino acid sequence diversity and Trp-Kyn metabolites (quinolinic acid (QUIN) as well as specific immune markers (sCD163 NGAL and hsCRP) in n = 67 South African cART-naïve people with HIV Sanger sequencing was used to determine blood-derived Tat/Vif amino acid sequence diversity a LC–MS/MS metabolomics platform was employed using a targeted approach Enzyme-linked immunosorbent assays and the Particle-enhanced turbidimetric assay was used sCD163 (p = 0.042) and KA (p = 0.031) were higher in participants with Tat signatures N24 and R57 and amino acid variation at position 24 (adj R2 = 0.048 p = 0.031) of Tat were associated with sCD163 and KA These preliminary findings suggest that amino acid variation in Tat may have an influence on underlying pathogenic HIV-1 mechanisms and therefore this line of work merits further investigation previous studies highlight that these specific amino acid signatures may be important in the pathogenesis of HIV-1 there is a need to profile the influence of viral protein amino acid sequence diversity in subtype C-specific cohorts we aimed to use an exploratory approach to investigate the potential associations between specific Tat and Vif amino acid sequence variants with soluble urokinase plasminogen activator receptor (suPAR) high-sensitivity C-reactive protein (hsCRP) soluble CD163 (sCD163) and neutrophil gelatinase-associated lipocalin (NGAL) as well as Trp-Kyn pathway metabolites in a cART treatment-naive South African cohort all participants received counselling from a trained counsellor The initial step in determining their HIV status involved using the First Response rapid HIV card test (Premier Medical Corporation Limited following the protocol outlined by the South African Department of Health the SD BIOLINE HIV 1/2 3.0 card test (Standard Diagnostics they then received post-counselling and were referred to the nearest clinical/hospital for further assessment CD4 + counts were analysed using the flow cytometric method (Beckman COULTER EPICS XL™ machine whole blood samples were collected in EDTA tubes samples underwent centrifugation at 2000 × g for 15 min at 10 °C within a 2-h timeframe the samples were transferred into microcentrifuge tubes and subsequently stored at -80 °C until further analysis the same rapid freezing process was employed but they were maintained at -18 °C for a maximum of five days before transportation to the laboratory these samples were again stored at -80 °C until they underwent subsequent analysis it is reasonable to hypothesize that Tat and Vif amino acid variations may influence other immune markers Plasma hsCRP were analysed using a particle-enhanced turbidimetric assay (Cobas Integra 400 plus while IL-6 levels were determined via the electrochemiluminescence immunoassay method (Elecsys 2010 ELISA assays (R&D Systems DuoSet) were employed for plasma sCD163 and NGAL measurements following the instructions from the manufacturer The coefficients of variation for both intra- and inter-assay tests fell within acceptable ranges Key mutations within the Tat/Vif regions were identified and specifically highlighted for further analysis All analyses were carried out utilizing SPSS software (IBM P-values below 0.05 were deemed statistically significant for all analyses Normality of variables was evaluated by visually inspecting QQ plots alongside descriptive statistics It was observed that the data distribution of immune markers IL-6 the data for skewed variables were log-transformed prior to statistical analyses Data presented acceptable skewness and kurtosis values within the range of -2 and 2 residual plots indicated homoscedasticity and linearity The Durbin-Watson statistic was within acceptable range residuals of the regression models were normally distributed χ2 tests were utilized to assess group disparities across amino acid variants for sex Independent sample T-tests were utilized to detect differences in study characteristics (such as age as well as levels of immune markers/metabolites For the χ2 tests and independent sample t-tests p values of < 0.05 we deemed significant To correct for the number of immune markers or metabolites tested a Bonferroni correction was implemented (α/n = 0.05/5 = 0.01) in all relevant analyses Pearson correlation analysis was utilized to identify covariates by exploring correlations between sociodemographic and lifestyle variables (age and locality) and specific immune markers/metabolites An Analysis of Covariance (ANCOVA) was used to adjust for the influence of covariates which helps in isolating the effect of the independent variable on the dependent variable with immune marker/metabolite levels as the dependent variables to compare their levels among Tat or Vif amino acid variants and locality in the investigation of immune markers and for alcohol use and BMI in the examination of metabolites to prevent model overfitting Multiple regression analysis using the enter method was employed to determine associations between Tat/Vif amino acid variants and immune marker/metabolite levels after adjusting for covariates For the Pearson correlation and multiple regression analyses Participants were stratified based on Tat amino acid variants at position 24 (K: 14 vs as well as Vif amino acid variants at position 17 (K: 25 vs Using independent sample t-tests as well as χ2 tests no significant differences were found in study characteristics (sex and smoking) amongst the investigated groups Participants with the Tat N24 variant had higher levels of sCD163 compared to participants with the K24 variant (p = 0.04) (supplementary Fig sCD163 levels were significantly higher in participants with Tat S31 compared to participants with C31 (p < 0.001) (supplementary Fig following the application of a Bonferroni correction (p = 0.05/5 = 0.01) only higher levels of sCD163 remained statistically significant in participants with the S31 variant (p < 0.001) None of the remaining Tat amino acid variants (positions 24 and 68) displayed significant differences for any of the immune markers or metabolites investigated (supplementary Figs None of the immune markers or metabolites were significantly different between the Vif amino acid variants (supplementary Figs Significant findings for Tat position 24 and 57 (A) sCD163 levels were significantly higher in participants with the N24 amino acid variant in contrast to participants with the K24 amino acid variant (p = 0.042) KA levels were significantly higher in the participants with the R57 amino acid variant compared to participants with the S57 variant (p = 0.031) The bars depict the average protein concentrations across the diverse study groups and are articulated as mean values with standard error of the mean (SEM) Volcano plot demonstrating the association between viral protein amino acid variations and immune marker/metabolite levels. The plot includes Tat amino acid position 24 (blue circles), position 31 (brown circles), position 57 (green circles), and position 68 (black circles). It also includes Vif amino acid positions 17 (orange triangles) and 31 (yellow triangles). Significant values, those with > -log10 1.3, are in red text Heatmap representing the associations (normalized effect size β) between Tat/Vif amino acid positions and peripheral immune and metabolic markers Significant associations with p-value < 0.05 are indicated by white asterisks Several key findings emerged: (1) After adjusting for covariates sCD163 and KA were higher in participants with Tat signatures N24 and R57 respectively and (2) amino acid variation at position 24 and 57 of Tat were associated with sCD163 and KA this previous study also investigated a small cohort; therefore these findings warrant further validation in larger cohorts further investigation is necessary to elucidate the complete functional significance of variation at this position Here our results also showed that participants with the R57 signature had higher levels of KA and amino acid variation at this position (between R57 and S57) was associated with KA a consistent trend for the impact of the R57 variant is observed in the dysregulation of metabolism indicating the necessity for larger cohort studies to further investigate this amino acid signature This implies that the Tat C31S status might not serve as an adequate biomarker for adverse clinical outcomes as the effects of mutations at this position could be concealed by other unexplored clinical factors no significant findings were reported for the influence of these amino acid signatures The precise roles of these Tat and Vif variants are yet to be fully explored it is evident from this study and many others that amino acid sequence variations of viral proteins may influence the structure–function relationships of these proteins ultimately impacting the underlying mechanisms of HIV-1 pathogenesis the extent to which these variations contribute to overall clinical outcomes in people with HIV requires further investigation and cannot be fully ascertained from this study alone We acknowledge that certain signatures investigated in cell culture (without confounders) similarly reflect characteristics in clinical sample types This discrepancy may stem from the fact that in people with HIV several other proteins may influence the levels of these markers or there may be other confounding factors at play in clinical investigations it is worth considering that certain amino acids may not directly influence outcomes their positioning in critical functional regions of key viral proteins may lead to changes in underlying mechanisms there may be additional potential covariates that could have influenced our findings in the clinical sample we have adjusted our analysis based on demographics and study characteristics that we have determined to have an influence Another limitation of our study is related to the number of statistical tests performed and the multiple testing correction strategy employed To identify covariates associated with our outcomes of interest This approach allowed us to adjust for only those covariates that were significantly associated thereby reducing the risk of overfitting the model While this conservative approach helps control the family-wise error rate it also increases the likelihood of Type II errors which means some true associations may not have been detected The stringent nature of the Bonferroni correction can lead to an overly cautious interpretation of results potentially overlooking meaningful findings we only investigated specific immune markers and metabolites and there may be other markers more directly involved in pathways related to the amino acid changes of the proteins we investigated the exploratory nature of this study should be taken into consideration when interpreting the findings presented here In a treatment-naïve cohort from South Africa subtype C we investigated the associations between changes in amino acid sequences in Tat and Vif and specific immune and metabolic markers respectively and amino acid variation at position 24 and 57 of Tat were associated with sCD163 and KA Findings from this study highlight the potential influence of amino acid sequence variation of the Tat protein on inflammatory and metabolic pathways in people with HIV The data supporting the findings of this study are available in the supplementary material of this article The sequences are accessible in GenBank under the accession numbers OR621303-OR621349 for Tat and OR194556-OR194606 for Vif UNAIDS. In danger: UNAIDS global AIDS update 2022. Joint United Nations Programme on HIV/AIDS; 2022. Available from: https://www.unaids.org/en/resources/documents/2022/in-danger-global-aids-update The epidemic of HIV/AIDS in developing countries; the current scenario in Pakistan The influence of viral protein R amino acid substitutions on clinical outcomes in people living with HIV: A systematic review Variation in HIV-1 Nef function within and among viral subtypes reveals genetically separable antagonism of SERINC3 and SERINC5 Structural similarity-based predictions of protein interactions between HIV-1 and Homo sapiens Molecular Modeling of Subtype-Specific Tat Protein Signatures to Predict Tat-TAR Interactions That May Be Involved in HIV-Associated Neurocognitive Disorders Signatures of HIV-1 subtype B and C Tat proteins and their effects in the neuropathogenesis of HIV-associated neurocognitive impairments HIV-1 integrase variants retarget viral integration and are associated with disease progression in a chronic infection cohort HIV-1 Subtype C Vpr Amino Acid Residue 45Y and Specific Conserved Fragments Are Associated with Neurocognitive Impairment and Markers of Viral Load New insights into pathogenesis point to HIV-1 Tat as a key vaccine target Viral infectivity factor: a novel therapeutic strategy to block HIV-1 replication Mutations in the highly conserved SLQYLA motif of Vif in a simian–human immunodeficiency virus result in a less pathogenic virus and are associated with G-to-A mutations in the viral genome Targeting Tat–TAR RNA Interaction for HIV-1 Inhibition The structural basis for HIV-1 Vif antagonism of human APOBEC3G Antiretroviral therapy does not block the secretion of the human immunodeficiency virus tat protein Partially active HIV-1 Vif alleles facilitate viral escape from specific antiretrovirals An investigation of the HIV Tat C31S and R57S mutation on peripheral immune marker levels in South African participants: A pilot study HIV-1 subtype C Tat exon-1 amino acid residue 24K is a signature for neurocognitive impairment Differential effects of HIV type 1 clade B and clade C Tat protein on expression of proinflammatory and antiinflammatory cytokines by primary monocytes Human immunodeficiency virus type 1 subtype C Tat fails to induce intracellular calcium flux and induces reduced tumor necrosis factor production from monocytes Differential induction of interleukin-10 in monocytes by HIV-1 clade B and clade C Tat proteins Impact of subtype C-specific amino acid variants on HIV-1 Tat-TAR interaction: insights from molecular modelling and dynamics HIV-1 Tat amino acid residues that influence Tat-TAR binding affinity: a scoping review A Naturally Occurring Polymorphism in the HIV-1 Tat Basic Domain Inhibits Uptake by Bystander Cells and Leads to Reduced Neuroinflammation Differential anti-APOBEC3G activity of HIV-1 Vif proteins derived from different subtypes Antagonism of PP2A is an independent and conserved function of HIV-1 Vif and causes cell cycle arrest HIV-1 Vif Triggers Cell Cycle Arrest by Degrading Cellular PPP2R5 Phospho-regulators A Pilot Investigation of the Association Between Vpr Amino Acid Substitutions and Peripheral Immune Marker Levels in People With Human Immunodeficiency Virus: Implications for Neurocognitive Impairment A pilot investigation of the association between HIV-1 Vpr amino acid sequence diversity and the tryptophan-kynurenine pathway as a potential mechanism for neurocognitive impairment HIV-1 Vif protein sequence variations in South African people living with HIV and their influence on Vif-APOBEC3G interaction Understanding the mechanisms driving the spread of subtype C HIV-1 Genetic and functional characterization of HIV-1 Vif on APOBEC3G degradation: First report of emergence of B/C recombinants from North India HIV-Related Immune Activation and Inflammation: Current Understanding and Strategies Persistent metabolic changes in HIV-infected patients during the first year of combination antiretroviral therapy Clinical Relevance of Kynurenine Pathway in HIV/AIDS: An Immune Checkpoint at the Crossroads of Metabolism and Inflammation Serum kynurenine-to-tryptophan ratio increases with progressive disease in HIV-infected patients Associations among peripheral and central kynurenine pathway metabolites and inflammation in depression The Prospective Urban Rural Epidemiology (PURE) study: Examining the impact of societal influences on chronic noncommunicable diseases in low- The Plasma [Kynurenine]/[Tryptophan] Ratio and Indoleamine 2,3-Dioxygenase: Time for Appraisal High-sensitivity C-reactive protein among people living with HIV on highly active antiretroviral therapy: a systemic review and meta-analysis Plasma Soluble CD163 Level Independently Predicts All-Cause Mortality in HIV-1–Infected Individuals Factors Associated With Plasma IL-6 Levels During HIV Infection Soluble Urokinase Plasminogen Activator Receptor Is Predictive of Non-AIDS Events During Antiretroviral Therapy-mediated Viral Suppression The Association of Immune Markers with Cognitive Performance in South African HIV-Positive Patients The association of peripheral immune markers with brain cortical thickness and surface area in South African people living with HIV ExPASy: The proteomics server for in-depth protein knowledge and analysis Tryptophan metabolism and its relationship with central nervous system toxicity in people living with HIV switching from efavirenz to dolutegravir The effects of highly active antiretroviral therapy on the serum levels of pro-inflammatory and anti-inflammatory cytokines in HIV infected subjects sCD163 and sCD14 Levels Have Distinct Associations with Antiretroviral Treatment and Cardiovascular Disease Risk Factors High soluble CD163 levels correlate with disease progression and inflammation in Kenyan children with perinatal HIV-infection Interleukin 6 Is a Stronger Predictor of Clinical Events Than High-Sensitivity C-Reactive Protein or D-Dimer During HIV Infection Kynurenic Acid: The Janus-Faced Role of an Immunomodulatory Tryptophan Metabolite and Its Link to Pathological Conditions Tat protein of human immunodeficiency virus type 1 subtype C strains is a defective chemokine Clade C HIV-1 isolates circulating in Southern Africa exhibit a greater frequency of dicysteine motif-containing Tat variants than those in Southeast Asia and cause increased neurovirulence NMDA receptor activation by HIV-Tat protein is clade dependent Impact of the HIV Tat C30C31S dicysteine substitution on neuropsychological function in patients with clade C disease Neuroimaging abnormalities in clade C HIV are independent of Tat genetic diversity Download references Open access funding provided by North-West University MEW received funding from the NRF Thuthuka grant (TTK22031652) and the Poliomyelitis Research Foundation (PRF) grant (23/84) LKA was supported by the NRF Postgraduate Scholarship (140524) and the PRF grant (23/24) Hypertension in Africa Research Team (HART) South African Medical Research Council Unit for Hypertension and Cardiovascular Disease EJVV and ZL: Consulted on statistical analysis The study protocol was approved by the Health Research Ethics Committee of North-West University in compliance with the Declaration of Helsinki All participants provided informed consent Participants were provided with detailed information about the study objectives They were informed that participation was voluntary and they could withdraw from the study at any time without any negative consequences Confidentiality and anonymity of the participants' data were assured throughout the study Written informed consent was obtained from each participant and copies of the consent forms are securely stored at the North-West University The authors declare no competing interests Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations Download citation DOI: https://doi.org/10.1186/s12879-024-09874-0 Anyone you share the following link with will be able to read this content: Sorry, a shareable link is not currently available for this article. Your Ads Privacy ChoicesIMDb Metrics details HIV-1 Vif recruits host cullin-RING-E3 ubiquitin ligase and CBFβ to degrade the cellular APOBEC3 antiviral proteins through diverse interactions Recent evidence has shown that Vif also degrades the regulatory subunits PPP2R5(A–E) of cellular protein phosphatase 2A to induce G2/M cell cycle arrest As PPP2R5 proteins bear no functional or structural resemblance to A3s it is unclear how Vif can recognize different sets of proteins Here we report the cryogenic-electron microscopy structure of PPP2R5A in complex with HIV-1 Vif–CBFβ–elongin B–elongin C at 3.58 Å resolution The structure shows PPP2R5A binds across the Vif molecule with biochemical and cellular studies confirming a distinct Vif–PPP2R5A interface that partially overlaps with those for A3s Vif also blocks a canonical PPP2R5A substrate-binding site indicating that it suppresses the phosphatase activities through both degradation-dependent and degradation-independent mechanisms Our work identifies critical Vif motifs regulating the recognition of diverse A3 and PPP2R5A substrates whereby disruption of these host–virus protein interactions could serve as potential targets for HIV-1 therapeutics Prices may be subject to local taxes which are calculated during checkout Multiple APOBEC3 restriction factors for HIV-1 and one Vif to rule them all Multifaceted HIV-1 Vif interactions with human E3 ubiquitin ligase and APOBEC3s Demystifying cell cycle arrest by HIV-1 Vif Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein APOBEC3G restricts HIV-1 to a greater extent than APOBEC3F and APOBEC3DE in human primary CD4+ T cells and macrophages APOBEC3 proteins can copackage and comutate HIV-1 genomes and APOBEC3H demonstrate a conserved capacity to restrict Vif-deficient HIV-1 Endogenous origins of HIV-1 G-to-A hypermutation and restriction in the nonpermissive T cell line CEM2n The AID/APOBEC family of nucleic acid mutators DNA deamination mediates innate immunity to retroviral infection Hypermutation of HIV-1 DNA in the absence of the Vif protein Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts Retroviral restriction factor APOBEC3G delays the initiation of DNA synthesis by HIV-1 reverse transcriptase APOBEC3G inhibits elongation of HIV-1 reverse transcripts APOBEC3F can inhibit the accumulation of HIV-1 reverse transcription products in the absence of hypermutation Deaminase-independent inhibition of HIV-1 reverse transcription by APOBEC3G APOBEC3G inhibits DNA strand transfer during HIV-1 reverse transcription Cytidine deaminases APOBEC3G and APOBEC3F interact with human immunodeficiency virus type 1 integrase and inhibit proviral DNA formation Human immunodeficiency virus type 1 cDNAs produced in the presence of APOBEC3G exhibit defects in plus-strand DNA transfer and integration APOBEC3F and APOBEC3G inhibit HIV-1 DNA integration by different mechanisms Antiviral function of APOBEC3G can be dissociated from cytidine deaminase activity Deep sequencing of HIV-1 reverse transcripts reveals the multifaceted antiviral functions of APOBEC3G Identification of APOBEC3DE as another antiretroviral factor from the human APOBEC family HIV-1 Vif protein binds the editing enzyme APOBEC3G and induces its degradation Vif overcomes the innate antiviral activity of APOBEC3G by promoting its degradation in the ubiquitin-proteasome pathway Adaptive evolution and antiviral activity of the conserved mammalian cytidine deaminase APOBEC3H The antiretroviral enzyme APOBEC3G is degraded by the proteasome in response to HIV-1 Vif Ubiquitination of APOBEC3 proteins by the Vif-Cullin5-ElonginB-ElonginC complex HIV-1 Vif blocks the antiviral activity of APOBEC3G by impairing both its translation and intracellular stability is suppressed by the HIV-1 and HIV-2 Vif proteins Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex Human APOBEC3F is another host factor that blocks human immunodeficiency virus type 1 replication Vif hijacks CBF-beta to degrade APOBEC3G and promote HIV-1 infection T-cell differentiation factor CBF-beta regulates HIV-1 Vif-mediated evasion of host restriction The Vif and Vpr accessory proteins independently cause HIV-1-induced T cell cytopathicity and cell cycle arrest The Vif accessory protein alters the cell cycle of human immunodeficiency virus type 1 infected cells Greenwood, E. J. et al. Temporal proteomic analysis of HIV infection reveals remodelling of the host phosphoproteome by lentiviral Vif variants. eLife https://doi.org/10.7554/eLife.18296 (2016) Naamati, A. et al. Functional proteomic atlas of HIV infection in primary human CD4+ T cells. eLife https://doi.org/10.7554/eLife.41431 (2019) Human immunodeficiency virus type 1 Vif induces cell cycle delay via recruitment of the same E3 ubiquitin ligase complex that targets APOBEC3 proteins for degradation Vif-CBFbeta interaction is essential for Vif-induced cell cycle arrest The Ebola virus nucleoprotein recruits the host PP2A-B56 phosphatase to activate transcriptional support activity of VP30 Structural basis of host protein hijacking in human T-cell leukemia virus integration B’-protein phosphatase 2A is a functional binding partner of delta-retroviral integrase Moura, M. & Conde, C. Phosphatases in mitosis: roles and regulation. Biomolecules https://doi.org/10.3390/biom9020055 (2019) Protein phosphatases in the regulation of mitosis The B56 family of protein phosphatase 2A (PP2A) regulatory subunits encodes differentiation-induced phosphoproteins that target PP2A to both nucleus and cytoplasm Crystal structure of a PP2A B56-BubR1 complex and its implications for PP2A substrate recruitment and localization Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling Marelli, S. et al. Antagonism of PP2A is an independent and conserved function of HIV-1 Vif and causes cell cycle arrest. eLife https://doi.org/10.7554/eLife.53036 (2020) Critical role of PP2A-B56 family protein degradation in HIV-1 Vif mediated G2 cell cycle arrest HIV-1 Vif triggers cell cycle arrest by degrading cellular PPP2R5 phospho-regulators Salamango, D. J. et al. Functional and structural insights into a Vif/PPP2R5 complex elucidated using patient HIV-1 isolates and computational modeling. J. Virol. https://doi.org/10.1128/JVI.00631-20 (2020) HIV-1 viral infectivity factor interacts with TP53 to induce G2 cell cycle arrest and positively regulate viral replication A conserved motif provides binding specificity to the PP2A-B56 phosphatase Formation of stable attachments between kinetochores and microtubules depends on the B56-PP2A phosphatase The PP2A(B56) phosphatase promotes the association of Cdc20 with APC/C in mitosis Structural basis of antagonism of human APOBEC3F by HIV-1 Vif Structural basis for HIV-1 antagonism of host APOBEC3G via Cullin E3 ligase Distinct domains within APOBEC3G and APOBEC3F interact with separate regions of human immunodeficiency virus type 1 Vif Identification of two distinct human immunodeficiency virus type 1 Vif determinants critical for interactions with human APOBEC3G and APOBEC3F Identification of the HIV-1 Vif and human APOBEC3G protein interface Structural basis for hijacking CBF-beta and CUL5 E3 ligase complex by HIV-1 Vif Expanding the PP2A interactome by defining a B56-specific SLiM Selective PP2A enhancement through biased heterotrimer stabilization Crystal structure of a protein phosphatase 2A heterotrimeric holoenzyme Highly accurate protein structure prediction with AlphaFold Critical role for protein phosphatase 2A heterotrimers in mammalian cell survival Nuclear export and centrosome targeting of the protein phosphatase 2A subunit B56alpha: role of B56alpha in nuclear export of the catalytic subunit Evolutionarily conserved pressure for the existence of distinct G2/M cell cycle arrest and A3H inactivation functions in HIV-1 Vif DeepEMhancer: a deep learning solution for cryo-EM volume post-processing Crystal structures of APOBEC3G N-domain alone and its complex with DNA Automated electron microscope tomography using robust prediction of specimen movements cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination UCSF Chimera–a visualization system for exploratory research and analysis New tools for the analysis and validation of cryo-EM maps and atomic models PHENIX: a comprehensive Python-based system for macromolecular structure solution The PyMOL Molecular Graphics System (DeLano Scientific UCSF ChimeraX: structure visualization for researchers Download references Wang at the Brookhaven Laboratory Cryo-EM facility for assistance with data collection We thank other Xiong laboratory members for discussions This work was supported by National Institutes of Health (NIH) grant no This work was supported in part by the Intramural Research Program of the NIH Center for Cancer Research and by the Innovation Award These authors contributed equally: Yingxia Hu Department of Molecular Biophysics and Biochemistry performed the biophysical and biochemical experiments contributed to experiments and discussions Nature Structural & Molecular Biology thanks Christopher Hill and the other reviewer(s) for their contribution to the peer review of this work Primary Handling Editor: Katarzyna Ciazynska in collaboration with the Nature Structural & Molecular Biology team Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations Size exclusion chromatography (SEC) profiles and SDS-PAGE analysis of individual PPP2R5A Vif/CBFβ/Cul5 E3 and their complexes (repeated twice independently) The cryo-EM reconstructions (surface with fitted models in cartoon representation) of the fully assembled Vif/CBFβ/Cul5 E3/PPP2R5A showed no presence of PPP2R5A The SEC profile and SDS-PAGE analysis of PPP2R5A/VCBC complex crosslinked by Bissulfosuccinimidyl suberate (BS3) The large scale crosslinking of the complex has been repeated 7 times independently The OD280 and OD260 are shown in blue and red The protein bands were detected by Coomassie blue stain Source data The Vif flexible loop that shows the largest local conformational changes upon A3F or A3G binding is highlighted by dashed circles and with details illustrated in insets PDBs used for the superpositions: A3FCTD alone: 3WUS; Vif/CBFβ: 4N9F; Vif/CBFβ/A3FCTD: 6NIL; VCBC/A3G: 8CX1 The individual Vif/CBFβ structures are shown in gray the A3F or A3G bound Vif/CBFβ are shown in magenta/cyan Right: structural model of the HIV-1 Vif recruitment of PPP2R5C-containing PP2A onto the Cul5 E3 ligase complex The in vitro binding assay was performed using MBP-tagged Vif/CBFβ/EloB/EloC variants to pull down SUMO-PPP2R5A variants The SUMO-PPP2R5A bands were recognized by anti-SUMO antibody and the His-CBFβ bands were detected by Anti-His antibody (upper panel) The ratio of band intensities (SUMO-PPP2R5A/His-CBFβ) was quantified by mean ± sem; n = 2 biologically independent samples for R127E:WT n = 3 biologically independent samples for WT:Y294A n = 4 biologically independent samples for K22E:WT n = 7 biologically independent samples for WT:WT with individual data points shown as dots (lower panel) Source data The observed (upper panel) and computational predicted (lower panel) structures are shown in three different views to demonstrate the differences between the observed and predicted binding modes of PPP2R5A Federation of European Biochemical Societies An example of the raw image of the complex (left) and the particle orientation distribution of the untilted dataset (right) Top 2D class averages of the combined untilted and tilted datasets From a total of 516,465 particles in the classes showing preferred orientations (boxed in red) 456,465 particles (~88%) were removed and excluded randomly from the subsequent analysis The final particle set showed a more balanced orientation distribution The Fourier shell correlation (FSC) curves of the cryo-EM reconstruction Local resolution estimate of the cryo-EM map a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law Download citation DOI: https://doi.org/10.1038/s41594-024-01314-6 Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily. a VIF of three or below is not a cause for concern the less reliable your regression results are going to be A VIF equal to one means variables are not correlated and multicollinearity does not exist in the regression model VIF measures the strength of the correlation between the independent variables in regression analysis This correlation is known as multicollinearity which can cause problems for regression models CFI. "Variance Inflation Factor." Isixsigma. "Variance Inflation Factor (VIF)." Welcome to IPE Real Assets. This site uses cookies. Read our policy By 2024-12-04T11:08:00+00:00 Vauban Infra Fibre (VIF) has acquired stakes in broadband operators in low-density areas of France from its co-shareholder Caisse des Dépôts et Consignations (CDC) The French digital infrastructure platform has invested an undisclosed to acquire Project Chrysalide which covers 19 fibre-to-the-home (FttH) networks and will be phased over 2024 and 2025 In September, Singapore’s sovereign wealth fund GIC acquired a minority stake in VIF through a capital increase alongside its existing shareholders which include funds managed by Vauban Infrastructure Partners and Crédit Agricole Assurances Vauban-managed investment funds remain the majority stakeholders in VIF VIF said GIC’s investment provides additional capital to fund future digital infrastructure investments and support long-term growth VIF said it also expects to announce the closing of a new digital infrastructure transaction that will strengthen its position in the French market and diversify its digital infrastructure base founding partners of Vauban Infrastructure Partners, said: “Since 2009 we have significantly expanded our digital investments by building on the recognised expertise of our teams and our close relationships with all the players in the sector which consolidates VIF’s position as the first independent FttH platform in France is fully in line with Vauban’s strategic commitment to continue its involvement as a long-term partner with all stakeholders to develop critical infrastructure within the territories.” To read the latest IPE Real Assets magazine click here Copyright © 1997–2025 IPE International Publishers Limited Site powered by Webvision Cloud and Fernando Galves Gen AI Solutions Architect AWS Outposts is a fully managed service that extends AWS infrastructure, services, APIs, and tools to customer premises. Outposts servers launched in 2022, a 1U or 2U rack-mountable host, with the ability to run Amazon Elastic Compute Cloud (Amazon EC2) and Amazon Elastic Container Service (Amazon ECS), as well as other appropriate smaller scale edge services such as AWS IoT Greengrass This version of Outposts is primarily focused on bringing lower latency AWS compute capabilities to the edge at many user locations During Outposts provisioning, you or AWS creates a service link connection that connects your Outposts server to your chosen AWS Region or home Region Outposts depends on regional connectivity “to reach out to home,” needing very little in terms of networking and providing authentication details through a command line the Outpost servers reach out to the regional endpoint Your Outpost status will show as Active when the process has completed it could take a few hours depending on service link bandwidth Although this has been suitable for the vast majority of use cases there are some locations that can’t provide internet connectivity in their environments This has mostly been in use cases where there is a strong security reason for not having an internet connection (such as financial services kiosks so as to avoid risks such as DDoS attacks and potential hack attempts or to meet requirements for receiving an authority to operate (ATO) These locations either have some form of direct connect or more commonly have a centralized direct connect link to AWS and an MPLS network linking all their remote sites to a central one the requirement is to allow the Outpost servers to resolve and reach the public endpoint for setup and subsequently the public anchor endpoint for management This is done without needing to leave the AWS ecosystem without needing to expose themselves unnecessarily to potential internet threats and without adding more systems to manage themselves we identified several key things that need to be provided if the user does not have internet connectivity at the remote location There are three different types of Virtual Interfaces (VIF) possible to configure on an AWS Direct Connect link: A transit VIF can be used to solve both of these issues a transit VIF deploys an ENI within a VPC (known as an attachment) so that traffic coming from the transit VIF into a VPC can be routed the traffic has to either be sourced or targeted for an ENI in the VPC instead of the transit gateway routing multiple VPCs to the internet Using a transit gateway to forward traffic to an NAT gateway allows you to provide internet connectivity for the Outposts servers without managing virtual appliances because NAT gateway provides this as a service NAT gateways also only allow outbound access so they provide security against any attempted external access by a bad actor from the internet This works for Outposts servers since they only need outbound access Outposts always initiate communication to an anchor or service endpoint and they never receive communication except as a response Architectural diagram showing the use of a Transit VIF and NAT gateway in a Region reaching regional endpoints Although the preceding architecture solves the challenge of how we provide a path for IP packets to transit between the Outposts servers and the public endpoints needed it doesn’t solve the issue of resolving DNS names If the remote site is isolated from the internet Amazon Route53 resolver endpoints allow you to deploy an IP address within a VPC subnet There are two types of resolver endpoints: outbound and inbound Outbound resolver endpoints are used by AWS to send DNS queries to your on-premises DNS servers Inbound resolver endpoints are used by your DNS servers (and hosts) to resolve addresses within Route 53 Route 53 can resolve public DNS names, so the Outposts service endpoint outposts.<region-name>.amazonaws.com becomes resolvable by an inbound resolver endpoint Using a public VIF allows you to provide an internet connection directly to the on-premises site this means you need to implement firewalls and security functions on this connection adding more layers of operational overhead A public VIF also means that the on-premises end of the VIF can be accessed by any public IP on the AWS public network regardless of the instance to which IP is mapped A public VIF is a public IP endpoint on the AWS public network You should treat public VIF traffic as internet-based traffic This can become cumbersome for firewalls teams if they have to allow-list known AWS IP ranges and manage the stateful firewall for a long range of AWS IPs even if the user is happy to implement and manage a firewall on the end of that public VIF there is still a question of how the Outpost would resolve DNS in this setup Unless the private network already has DNS resolution to a public DNS then there are no DNS servers that DHCP can point to in order to allow the Outposts servers to get name resolution This is because there is no public DNS endpoint within the AWS public network Traffic from a user’s public VIF can access the AWS public network but it can’t exit it to other public networks if the you had configured DHCP to point to one of the well-known DNS servers (such as 8.8.8.8) since this DNS servers lives outside of the AWS public network requests originating from the on-premises side of a public VIF would be dropped as it hit the border of the AWS autonomous system The only way for a DNS request to be resolved would be to build a bind forwarding service within a VPC and point the DHCP DNS values at this IP address This network configuration introduces complexity and won’t be possible for those with highly regulated workloads You would need to manage a firewall on-premises allow a public network to reach the on-premises location and manage a bind servers setup within a VPC a public VIF is generally not an option unless the user is already running one and is familiar with the steps to secure it Architectural diagram showing traffic flow using a public VIF and AWS Outposts Virtual private gateways do not have an ENI associated with them but are pointed to as a next hop within a subnet routing table If we take this example and look at what the Outposts servers would be trying to pass as traffic then it would send a packet with a source address of the Outposts servers and a destination address of the Outposts service public endpoint (assuming that it could resolve it) then neither the source nor destination address would belong to an ENI within the VPC Even if there was a routing rule on the subnet pointing the next hop for all traffic to a NAT gateway (ideal for internet egress) This is because the packet from the Outposts servers doesn’t have a destination of the NAT gateway but instead a destination of the setup endpoint in the internet It’s possible to use a combination of ingress routing and transparent proxies to ingest the traffic and pass it to an instance running a proxy service to forward to the internet this adds complexity having to manage and maintain proxy servers a private VIF is generally not recommended Architectural diagram showing VGW and packet drops because of transitive routing not being supported In this post, we discussed architecture patterns you can use to provision your Outposts when public internet connectivity is unavailable. To get started with Outpost servers please visit our Server User Guide. For more information, contact us to learn more The human immune deficiency virus (HIV) first entered public consciousness in the early 1980s after cases of unfamiliar and deadly illnesses began to overwhelm medical centres across North America “An estimated 42 million people have died from HIV/AIDS to date and while people can now live full lives with access to treatments it is still a chronic condition that people would like a cure for,” said Dr Immunology and Biochemistry in the College of Medicine at the University of Saskatchewan (USask).   Chelico’s lab studies immune responses to HIV particularly a family of proteins called APOBEC3 which cause mutations in the HIV virus Her research and others’ show that the virus uses a counterattack that breaks down APOBEC3’s defences helping HIV win and establish an infection.  “There is a battle going on in the cell,” said Chelico “HIV also has a protein known as Vif that it uses against APOBEC3 so it’s this fight against each other.”  have tried to create drugs that block Vif from interacting with APOBEC3 giving proteins the added “boost” it needs to fully inactivate the HIV virus and prevent an infection but these blockers have been mostly ineffective.  Now, four decades after HIV was first identified, Chelico and her colleagues, including USask post-doctoral fellow Dr. Amit Gaba (PhD), have discovered previously unknown interactions between APOBEC3 and Vif proteins which offer a better map for drug design. Their findings were recently published in the American Microbiology Association’s Journal of Virology.  previous inhibitors weren’t successful because we weren’t looking at the right interface between the protein interactions,” said Chelico “We think that with a better map of this interface we can design a better inhibitor.”  Previous studies used one Vif protein and a single APOBEC3 protein to simulate conditions during an infection but this experimental design isn’t quite measuring up to what is actually happening inside our cells Chelico and her team found that there are differences in the Vif proteins among HIV strains and that APOBEC3s are not working individually against HIV.  “My lab established that APOBEC3s are working together during an infection and they form a new structure,” said Chelico “APOBEC proteins are part of a natural defence system against viruses and if we can stop that interaction with Vif They could be a kind of natural cure.”  this groundbreaking discovery could pave the way for new treatments The potential for APOBEC proteins to serve as a natural defence against HIV brings a new wave of optimism in the fight against the virus.  “HIV still remains a global public health issue,” said Chelico “Current treatments only suppress the virus so we’re really hopeful that our research offers a new avenue where we can help the body’s natural defences stopping HIV infections from taking hold.”  Want to learn more about Chelico’s revolutionizing work? Check out her 2024 TEDxUniversityofSaskatchewan talk.   Together, we are addressing the world's greatest challenges. Join our ambitious vision for the future The University of Saskatchewan's main campus is situated on Treaty 6 Territory and the Homeland of the Métis. © University of SaskatchewanDisclaimer | Privacy | Accessibility Volume 4 - 2013 | https://doi.org/10.3389/fmicb.2013.00034 This article is part of the Research TopicRetroviruses, retroelements and their restrictionsView all 13 articles The research on virion infectivity factor (Vif) protein had started in late 1980s right after HIV-1 was cloned and the function of Vif had been a mystery for a long time the research on Vif has finally lead to the identification of APOBEC3G which opens up a new era in the research field of host restriction factors in HIV-1 infection followed by TRIM5α This suggests that continuation of basic research on fundamental questions is quite important We still have many questions on Vif and APOBEC3 and should continue to work on these proteins in the future in order to better regulate HIV-1 We will discuss not only the history but also recent advances in Vif research The underlying mechanism of Vif function had been unsolved and a mystery for a long time In addition to the above described main function, early studies also revealed several important Vif functions including dimerization (Yang et al., 2001), virion incorporation (Camaur and Trono, 1996; Simon et al., 1997), and phosphorylation (Yang et al., 1996; Yang and Gabuzda, 1998); however the significances of these functions are not discussed much recently a novel Vif function on cell cycle has been reported Schematic figure of the virion infectivity factor (Vif) protein and amino acid motifs for binding to Vif-interacting proteins Pink indicates binding motifs for A3G; light blue indicates binding motifs for A3F; light green indicates binding motifs for Cul5; yellow indicates binding motifs for EloC; light pink indicates motifs for dimerization Vif binds to p53 and CBFβ in its N-terminal regions but binding motifs were not elucidated yet It is quite important to reveal the interaction sites between Vif and APOBEC3 proteins because the regulation of this interaction may lead to the development of novel therapeutic strategies for HIV-1 infection their structural information is not fully elucidated yet because it is quite difficult to produce these proteins as soluble forms the information described below is mainly obtained by many studies using site-directed mutagenesis we have to wait a little longer until we will get the structural information of these complexes the mechanisms by which CBFβ regulates the E3 ligase complex are still under investigation since CBFβ is an important T cell transcription factor it would be very interesting to determine whether Vif affects T cell differentiation The identification of the E3 ligase has lead to elucidation of the mechanisms of Vif-induced G2 cell cycle arrest described below The mechanisms how Vif is ubiquitinated and degraded and how Vif induces G2 cell cycle arrest Vif inhibits ubiquitination of p53 by MDM2 to induce activation and nuclear import of p53 Activated p53 induces transcription of several genes including MDM2 and p21 Enhanced expression of MDM2 may lead to more Vif ubiquitination and degradation which forms the autoregulatory circuit of Vif expression activation of p21 leads to G2 cell cycle arrest HIV-1 needs to have G2 cell cycle arrest to efficiently replicate so that it possesses two accessory genes such as vif and vpr Vif induces G2 arrest in a p53-dependent manner while Vpr accomplishes the same goal in a p53-independent manner not only because it opens up a new era in the research field of host restriction factors but also because it has a variety of functions for the viral life cycle by interacting several cellular proteins It suggests that it might be a good target for control of HIV-1 infection The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest This work was partly supported by grants-in-aid from the Ministry of Education and Technology and from the Ministry of Health This work was also partly supported by grants from SENSHIN Medical Research Foundation High level expression of human immunodeficiency virus type-1 Vif inhibits viral infectivity by modulating proteolytic processing of the Gag precursor at the p2/nucleocapsid processing site Pubmed Abstract | Pubmed Full Text | CrossRef Full Text A single amino acid difference in the host APOBEC3G protein controls the primate species specificity of HIV type 1 virion infectivity factor Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Characterization of human immunodeficiency virus type 1 Vif particle incorporation Pubmed Abstract | Pubmed Full Text A patch of positively charged amino acids surrounding the human immunodeficiency virus type 1 Vif SLVx4Yx9Y motif influences its interaction with APOBEC3G Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Identification of a novel WxSLVK motif in the N terminus of human immunodeficiency virus and simian immunodeficiency virus Vif that is critical for APOBEC3G and APOBEC3F neutralization Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Identification of highly attenuated mutants of simian immunodeficiency virus Pubmed Abstract | Pubmed Full Text The Vif protein of human immunodeficiency virus type 1 is posttranslationally modified by ubiquitin Pubmed Abstract | Pubmed Full Text | CrossRef Full Text The sor gene of HIV-1 is required for efficient virus transmission in vitro Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Expression of HIV-1 accessory protein Vif is controlled uniquely to be low and optimal by proteasome degradation Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Role of vif in replication of human immunodeficiency virus type 1 in CD4+ T lymphocytes Pubmed Abstract | Pubmed Full Text Insights into the dual activity of SIVmac239 Vif against human and African green monkey APOBEC3G Pubmed Abstract | Pubmed Full Text | CrossRef Full Text HIV-1 Vpr increases viral expression by manipulation of the cell cycle: a mechanism for selection of Vpr in vivo Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Human immunodeficiency virus type 1 viral protein R (Vpr) arrests cells in the G2 phase of the cell cycle by inhibiting p34cdc2 activity Pubmed Abstract | Pubmed Full Text Characterization of conserved motifs in HIV-1 Vif required for APOBEC3G and APOBEC3F interaction Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Identification of amino acid residues in APOBEC3G required for regulation by human immunodeficiency virus type 1 Vif and Virion encapsidation Pubmed Abstract | Pubmed Full Text | CrossRef Full Text HIV-1 Vif-mediated ubiquitination/degradation of APOBEC3G involves four critical lysine residues in its C-terminal domain Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Structural features of antiviral APOBEC3 proteins are linked to their functional activities Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Ubiquitination of APOBEC3G by an HIV-1 Vif–Cullin5–Elongin B–Elongin C complex is essential for Vif function Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Primate lentiviral virion infectivity factors are substrate receptors that assemble with cullin 5-E3 ligase through a HCCH motif to suppress APOBEC3G Pubmed Abstract | Pubmed Full Text | CrossRef Full Text An endogenous inhibitor of human immunodeficiency virus in human lymphocytes is overcome by the viral Vif protein Pubmed Abstract | Pubmed Full Text A single amino acid determinant governs the species-specific sensitivity of APOBEC3G to Vif action Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Species-specific exclusion of APOBEC3G from HIV-1 virions by Vif Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Phosphorylation of a novel SOCS-box regulates assembly of the HIV-1 Vif–Cul5 complex that promotes APOBEC3G degradation Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Vif Overcomes the innate antiviral activity of APOBEC3G by promoting its degradation in the ubiquitin–proteasome pathway Pubmed Abstract | Pubmed Full Text | CrossRef Full Text A zinc-binding region in Vif binds Cul5 and determines cullin selection Pubmed Abstract | Pubmed Full Text | CrossRef Full Text HIV-1 Vpr induces ATM-dependent cellular signal with enhanced homologous recombination Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Human immunodeficiency virus type 1 Vif inhibits packaging and antiviral activity of a degradation-resistant APOBEC3G variant Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Regulation of APOBEC3 proteins by a novel YXXL motif in human immunodeficiency virus type 1 Vif and simian immunodeficiency virus SIVagm Vif Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Human immunodeficiency virus type 1 Vpr arrests the cell cycle in G2 by inhibiting the activation of p34cdc2-cyclin B Pubmed Abstract | Pubmed Full Text Activation of the ATR-mediated DNA Damage Response by the HIV-1 viral protein R Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text Cell-dependent requirement of human immunodeficiency virus type 1 Vif protein for maturation of virus particles Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text A single amino acid of APOBEC3G controls its species-specific interaction with virion infectivity factor (Vif) Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Polyubiquitination of APOBEC3G is essential for its degradation by HIV-1 Vif Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Ubiquitination of APOBEC3 proteins by the Vif–Cullin5–ElonginB–ElonginC complex Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Phosphorylation of APOBEC3G by protein kinase A regulates its interaction with HIV-1 Vif Pubmed Abstract | Pubmed Full Text | CrossRef Full Text The Vif and Gag proteins of human immunodeficiency virus type 1 colocalize in infected human T cells Pubmed Abstract | Pubmed Full Text Evidence for a newly discovered cellular anti-HIV-1 phenotype Pubmed Abstract | Pubmed Full Text | CrossRef Full Text The regulation of primate immunodeficiency virus infectivity by Vif is cell species restricted: a role for Vif in determining virus host range and cross-species transmission Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text The HIV ‘A’ (sor) gene product is essential for virus infectivity Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text | CrossRef Full Text HIV-1 Vif versus the APOBEC3 cytidine deaminases: an intracellular duel between pathogen and host restriction factors Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Zinc chelation inhibits HIV Vif activity and liberates antiviral function of the cytidine deaminase APOBEC3G Pubmed Abstract | Pubmed Full Text | CrossRef Full Text A single amino acid substitution in human APOBEC3G antiretroviral enzyme confers resistance to HIV-1 virion infectivity factor-induced depletion Pubmed Abstract | Pubmed Full Text | CrossRef Full Text The multimerization of human immunodeficiency virus type I Vif protein: a requirement for Vif function in the viral life cycle Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Mitogen-activated protein kinase phosphorylates and regulates the HIV-1 Vif protein Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Phosphorylation of Vif and its role in HIV-1 replication Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif–Cul5–SCF complex Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Selective assembly of HIV-1 Vif–Cul5–ElonginB–ElonginC E3 ubiquitin ligase complex through a novel SOCS box and upstream cysteines Pubmed Abstract | Pubmed Full Text | CrossRef Full Text Pubmed Abstract | Pubmed Full Text Citation: Takaori-Kondo A and Shindo K (2013) HIV-1 Vif: a guardian of the virus that opens up a new era in the research field of restriction factors. Front. Microbio. 4:34. doi: 10.3389/fmicb.2013.00034 Copyright: © 2013 Takaori-Kondo and Shindo. This is an open-access article distributed under the terms of the Creative Commons Attribution License distribution and reproduction in other forums provided the original authors and source are credited and subject to any copyright notices concerning any third-party graphics etc *Correspondence: Akifumi Takaori-Kondo, Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Shogoin-Kawaracho 54, Sakyo-ku, Kyoto 606-8507, Japan. e-mail:YXRha2FvcmlAa3VocC5reW90by11LmFjLmpw Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher 94% of researchers rate our articles as excellent or goodLearn more about the work of our research integrity team to safeguard the quality of each article we publish Metrics details that antagonizes A3 family members by targeting them for degradation Diversification of A3 allows host escape from Vif whereas adaptations in Vif enable cross-species transmission of primate lentiviruses How this ‘molecular arms race’ plays out at the structural level is unknown we report the cryogenic electron microscopy structure of human APOBEC3G (A3G) bound to HIV-1 Vif and the hijacked cellular proteins that promote ubiquitin-mediated proteolysis A small surface explains the molecular arms race including a cross-species transmission event that led to the birth of HIV-1 we find that RNA is a molecular glue for the Vif–A3G interaction enabling Vif to repress A3G by ubiquitin-dependent and -independent mechanisms Our results suggest a model in which Vif antagonizes A3G by intercepting it in its most dangerous form for the virus—when bound to RNA and on the pathway to packaging—to prevent viral restriction By engaging essential surfaces required for restriction suggesting a general mechanism by which RNA binding helps to position key residues necessary for viral antagonism of a host antiviral gene Although it is commonly assumed that sites of molecular arms races report on direct protein interactions physical evidence of this interaction site to explain the mechanisms of how Vif promotes processive ubiquitination on A3G and how mutations in Vif or A3G promote host escape and viral adaptation leaving substantial gaps in our knowledge of molecular mechanisms of Vif antagonism of A3 proteins and molecular arms races between them logo plot of amino acids found in the consensus of all HIV-1 clades as well as SIVcpz (black bar) and all other SIV strains with equal distribution of each SIV (white bar) Both this result and our structure suggest that A3G bound to purine-rich RNA is the substrate of the Vif E3 ligase which validates their importance in our structure Ribbon diagram showing position of molecular arms race interface (spheres) relative to the RNA interface (sticks) Close-up of molecular arms race interface (top) and residues that contribute to Vif–A3G binding and in contact with RNA (bottom) Residues D128 and D130 of A3G are under diversifying selection; residue Q83 is an adaptation that allowed SIVrcm Vif to neutralize hominid primate A3G and thus enable cross-species transmission Logo plots of natural sequence variation in Vif residues that line the molecular arms race (top) and Vif–A3G–RNA interface (bottom) HIV-1 and SIVcpz sequences (black bars) are the consensus of all HIV-1 clades as well as SIVcpz and SIV sequences (white bars) are all other SIV strains using equal distribution of each SIV We suggest that structural plasticity in Vif enabled amino acid substitutions to neutralize A3G and enable cross-species transmission of SIV from red-capped mangabeys to chimpanzees We conclude that Vif binds A3G/RNA in a manner that limits A3G escape over long evolutionary timescales by engaging an essential surface required for antiviral function explaining why genetic signatures of diversifying selection and adaptation are constrained to the direct protein interactions observed at the molecular arms race interface Bottom panels show close-up of interactions within A3G CDA domains This observation indicates that VCBC binds A3G in a manner that inhibits its self-association We suggest that these surfaces may be bridged by cellular cofactors as described for A3G possibility is that interactions with Vif are stabilized by bipartite interactions with tandem CDA domains of A3 proteins Structural studies of Vif–A3 complexes purified after coexpression or native purification from eukaryotic cells will allow this question to be addressed in future studies We propose that the substrate of the Vif E3 ligase is not A3G but rather a complex of A3G bound to purine-rich RNA including purine-rich sequences found in the viral genome This finding suggests that Vif binding to A3G has the capacity to block its packaging independent of ubiquitination activity a mechanism that may potentiate repression of restriction a,b, Packaging of A3G into HIV-1 virus requires A3G dimerization and its interaction with viral RNA (a); Vif neutralizes A3G early during its biosynthesis by binding RNA-bound A3G, inhibition of A3G dimerization and promotion of ubiquitin-mediated proteolysis (b). Created with BioRender.com A defocus range of −0.8 to −2.0 μm was applied The weak density in this region precluded precise atomic modelling and thus the A3G–Vif dimeric interface for state 2 is interpretable on one side only We generated a library of variants at positions 22 26 and 40 using degenerate oligonucleotide mutagenesis in the HIV-1 LAI vif gene Individual colonies were sequenced and ligated into a lentiviral vector flanked by a C-terminal 3XFLAG epitope tag in the pcDNA4/TO vector backbone (Thermo Fisher was transfected into HEK293T cells (ATCC CRL-3216 regularly tested for mycoplasma contamination) plated in six-well dishes at a density of 1.5 × 105 cells ml–1 The amount of A3G packaged into virions was assayed by cotransfection of 1,000 ng of Vif vector 200 ng of A3G-3XFlag and 500 ng of psPAX2 for gag/pol production with TransIT-LT1 transfection reagent (Mirus MIR2304) at a reagent to plasmid DNA ratio of 3:1 1 ml of the supernatant was filtered through a 0.2 μm syringe filter and virions were pelleted in an Eppindorf 5415R tabletop microcentrifuge for 1 h at 4 °C and maximum speed and 25 μl of NuPAGE 4× loading dye (Invitrogen no Samples were boiled for 10 min at 95 °C and loaded on an SDS–PAGE gel 3537) antibodies were used for immunoblotting at a dilution of 1:5,000 Mouse IgG HRP-conjugated antibody (R&D systems HAF007) was used to detect primary antibodies at a dilution of 1:5,000 Chemiluminescent signals from all immunoblots were imaged using the ChemiDocMP imaging system (Bio-Rad) and images were processed with ImageJ software to quantify the densitometry for each detected antibody band Normalized A3G in virions was calculated by dividing the amount of A3G by that of p24gag and setting that number to 1.0 for the ‘No Vif’ control Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article Structural insights into APOBEC3-mediated lentiviral restriction The battle between retroviruses and APOBEC3 genes: its past and present HIV-1 uncoats in the nucleus near sites of integration Reconstitution and visualization of HIV-1 capsid-dependent replication and integration in vitro Nuclear pore blockade reveals that HIV-1 completes reverse transcription and uncoating in the nucleus RNA-dependent oligomerization of APOBEC3G is required for restriction of HIV-1 Identification of amino acid residues in APOBEC3G required for regulation by human immunodeficiency virus type 1 Vif and virion encapsidation Transcriptional regulation of APOBEC3 antiviral immunity through the CBF-beta/RUNX axis Fab-based inhibitors reveal ubiquitin independent functions for HIV Vif neutralization of APOBEC3 restriction factors Human immunodeficiency virus type 1 Vif protein is packaged into the nucleoprotein complex through an interaction with viral genomic RNA Ancient adaptive evolution of the primate antiviral DNA-editing enzyme APOBEC3G The host restriction factor APOBEC3G and retroviral Vif protein coevolve due to ongoing genetic conflict Rules of engagement: molecular insights from host-virus arms races Gene loss and adaptation to hominids underlie the ancient origin of HIV-1 Structure of the Vif-binding domain of the antiviral enzyme APOBEC3G Crystal structure of a soluble APOBEC3G variant suggests ssDNA to bind in a channel that extends between the two domains Understanding the structural basis of HIV-1 restriction by the full length double-domain APOBEC3G Structural determinants of HIV-1 Vif susceptibility and DNA binding in APOBEC3F Conformational dynamics of the HIV-Vif protein complex Host gene evolution traces the evolutionary history of ancient primate lentiviruses Function analysis of sequences in human APOBEC3G involved in Vif-mediated degradation The Vif protein of HIV triggers degradation of the human antiretroviral DNA deaminase APOBEC3G Functional analysis and structural modeling of human APOBEC3G reveal the role of evolutionarily conserved elements in the inhibition of human immunodeficiency virus type 1 infection and Alu transposition Antiviral protein APOBEC3G localizes to ribonucleoprotein complexes found in P bodies and stress granules Two regions within the amino-terminal half of APOBEC3G cooperate to determine cytoplasmic localization The anti-HIV-1 editing enzyme APOBEC3G binds HIV-1 RNA and messenger RNAs that shuttle between polysomes and stress granules Identification of amino acid residues in HIV-1 Vif critical for binding and exclusion of APOBEC3G/F Vif proteins from diverse primate lentiviral lineages use the same binding site in APOBEC3G The RNA binding specificity of human APOBEC3 proteins resembles that of HIV-1 nucleocapsid Mechanism of auxin perception by the TIR1 ubiquitin ligase Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide Examination of the APOBEC3 barrier to cross species transmission of primate lentiviruses Structural basis for a species-specific determinant of an SIV Vif protein toward hominid APOBEC3G antagonism Ubiquitin ligation to F-box protein targets by SCF-RBR E3-E3 super-assembly Dispersed sites of HIV Vif-dependent polyubiquitination in the DNA deaminase APOBEC3F ARIH2 is a Vif-dependent regulator of CUL5-mediated APOBEC3G degradation in HIV infection The biased nucleotide composition of the HIV genome: a constant factor in a highly variable virus Newly synthesized APOBEC3G is incorporated into HIV virions Differential sensitivity of “old” versus “new” APOBEC3G to human immunodeficiency virus type 1 vif MacroBac: new technologies for robust and efficient large-scale production of recombinant multiprotein complexes SerialEM: a program for automated tilt series acquisition on Tecnai microscopes using prediction of specimen position Scipion: a software framework toward integration reproducibility and validation in 3D electron microscopy MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy Sampling the conformational space of the catalytic subunit of human gamma-secretase Visualizing density maps with UCSF Chimera RELION: implementation of a Bayesian approach to cryo-EM structure determination 3D variability analysis: resolving continuous flexibility and discrete heterogeneity from single particle cryo-EM Optimal determination of particle orientation and contrast loss in single-particle electron cryomicroscopy Improvement of cryo-EM maps by density modification Quantifying the local resolution of cryo-EM density maps Addressing preferred specimen orientation in single-particle cryo-EM through tilting Asarnow, D., Palovcak, E. & Cheng, Y. asarnow/pyem:UCSF pyem v0.5. Zenodo https://zenodo.org/record/3576630#.Y-Tme3bP3IU (2019) Comparative protein modelling by satisfaction of spatial restraints Comparative protein structure modeling using MODELLER Coot: model-building tools for molecular graphics ISOLDE: a physically realistic environment for model building into low-resolution electron-density maps Real-space refinement in PHENIX for cryo-EM and crystallography Haruspex: a neural network for the automatic identification of oligonucleotides and protein secondary structure in cryo-electron microscopy maps Correcting pervasive errors in RNA crystallography through enumerative structure prediction MolProbity: all-atom structure validation for macromolecular crystallography Measurement of atom resolvability in cryo-EM maps with Q-scores BINANA 2: characterizing receptor/ligand interactions in Python and JavaScript PLIP: fully automated protein-ligand interaction profiler LigPlot+: multiple ligand-protein interaction diagrams for drug discovery Schrödinger, L. & DeLano, W. L. PyMOL. http://www.pymol.org/pymol (2020) UCSF ChimeraX: meeting modern challenges in visualization and analysis Statistical potential for assessment and prediction of protein structures CUL5-ARIH2 E3-E3 ubiquitin ligase structure reveals cullin-specific NEDD8 activation Deciphering key features in protein structures with the new ENDscript server Download references Cheng and Gross laboratories for helpful discussions on this project; D Bulkley of UCSF Cryo-EM facility for assistance with data collection; S Gradia of Macrolab for insect cell expression vectors; and A Manglik of UCSF for advice on model building and refinement was supported by the University of Washington STD/AIDS Research Training Fellowship (no NIH/NIAID T32-AI07140) and by a 2021 New Investigator Award from the University of Washington/Fred Hutch Center for AIDS Research (NIH-funded programme under award no was supported by a University of Washington Cellular and Molecular Biology Training Grant (no This work was supported by funding from NIH P50AI150476 and U54AI170792 to M.E. Divisions of Human Biology and Basic Sciences Molecular and Cellular Biology Graduate Program Department of Cellular and Molecular Pharmacology Department of Bioengineering and Therapeutic Sciences designed and expressed purified protein complexes collected and processed cryo-EM data and performed model building and refinement developed and performed functional experiments and evolutionary analysis in microscope operation and cryo-EM data acquisition helped with EM data analysis and interpretation checked all published mutants of A3G and Vif All authors contributed to data interpretation Similar class averages were obtained from three independent preparations imaged on Quantifoil Gold grids Shown at the bottom are expanded views of the fourth and fifth 2D classes with two copies of A3G-VCBC labeled in white and yellow Shown on the right are the A3G and VCBC structures fit in the consensus map that is colored by subunits Masks were used to determine different regions of volume for focused refinement and 3D variability analysis in cryoSPARC Green and orange boxes indicate the final reconstruction for monomer and dimers respectively; their corresponding Gold-Standard Fourier Shell Correlation (GSFSC) curves are shown at the bottom The nominal resolution of the final map for monomer Right of each panel: Logo plot of amino acids found in the consensus of all HIV-1 clades as well as SIVcpz (black bar) and all other SIV strains using equal distribution of each SIV (white bar) Residue F46 and W74 of SIVrcm Vif previously reported to be critical for rcmA3G neutralization engage in extensive hydrophobic interactions with rcmA3G in the model (bottom) Note amino acids 16 and 86 of SIVrcm Vif correspond to amino acids 15 and 83 Sequence alignment of A3G residues that contact RNA or Vif from Old World Monkeys and hominids Fully conserved residues are highlighted with white text on black background Buried solvent accessible surface area for A3G–RNA–VCBC monomer structure Supplementary discussion: this file discusses the structure of A3G-RNA-VCBC dimeric complexes 1 contains uncropped source images for Fig 2–4 show the model–map fit for A3G-RNA-VCBC monomeric and dimeric complexes Supplementary Table 1 summarizes Vif residues reportedly involved in RNA binding based on mutational analyses Supplementary Table 2 summarizes A3G residues reportedly involved in RNA binding based on mutational analyses Comparison of dimeric complex structure in states 1 and 2 States 1 and 2 are related by rigid-body motion of A3G-RNA-VCBC protomers Download citation DOI: https://doi.org/10.1038/s41586-023-05779-1 a shareable link is not currently available for this article Nature Reviews Molecular Cell Biology (2024) Sign up for the Nature Briefing newsletter — what matters in science By 2024-09-20T14:22:00 Sovereign wealth fund injects capital into French digital infrastructure platform via an affiliate Already a registered user or subscriber? Sign in here Register today PropertyEU has now merged with IPE Real AssetsRegistration will give you access to the PropertyEU archive.If you have a PropertyEU membership find out how to get access Metrics details Human APOBEC3 (A3) cytidine deaminases are antiviral factors that are particularly potent against retroviruses HIV-1 uses a viral infectivity factor (Vif) to target specific human A3s for proteasomal degradation Vif recruits cellular transcription cofactor CBF-β and Cullin-5 (CUL5) RING E3 ubiquitin ligase to bind different A3s distinctively but how this is accomplished remains unclear in the absence of the atomic structure of the complex we present the cryo-EM structures of HIV-1 Vif in complex with human A3H CBF-β and components of CUL5 ubiquitin ligase (CUL5 Vif nucleates the entire complex by directly binding four human proteins The structures reveal a large interface area between A3H and Vif primarily mediated by an α-helical side of A3H and a five-stranded β-sheet of Vif This A3H-Vif interface unveils the basis for sensitivity-modulating polymorphism of both proteins including a previously reported gain-of-function mutation in Vif isolated from HIV/AIDS patients Our structural and functional results provide insights into the remarkable interplay between HIV and humans and would inform development efforts for anti-HIV therapeutics how these multiple interacting interfaces contribute to the remarkable interplay between HIV and host remains incomplete due to the lack of high-resolution structures of Vif-host factor complexes We report here the cryo-EM structures of human A3H bound to HIV-1 Vif in complex with CBF-β and multiple components of CUL5 E3 ligase The structures reveal the architecture of the multicomponent HIV-human protein complex and illuminate the molecular details of the crucial interface between A3H and Vif that enables stable polyubiquitin chain formation and degradation of A3H Our study further provides insights into the A3H ubiquitination sites targeted by the Vif-CUL5 E3 ligase complex a Alignment of Vif sequences from various HIV-1/SIV strains At amino acid position 48 (marked by a triangle) At amino acid position 97 (marked by a triangle) whereas chimpanzee A3H has glutamine (Q97) Conserved zinc-coordinating cysteines are marked by asterisks c Vif-mediated degradation assay of A3H in HEK293T cells and steady-state levels of A3H at post-48 h transfection were analyzed by Western blots Quantified A3H levels are shown in the right-side chart Assays were performed independently in triplicate (mean ± s.d.; n = 3) The statistical significance of A3H degradation was assessed by two-tailed t-test assuming equal variance; *P < 0.05; **P < 0.005; ***P < 0.0005; NS The addition of N48H mutation in Vif lowered the levels of both WT and K97Q A3H K97Q mutation in A3H lowered the A3H levels when transfected with either WT or N48H Vif An anti-α-tubulin antibody was used as a loading control d Binding analysis of A3H and VCBCCR complex by SEC The biggest peak shift was observed when K97Q A3H was combined with N48H NL4-3 Vif The fractions corresponding to peak 1 were used for the cryo-EM study SDS-PAGE gels show the protein components in the indicated SEC fractions Vif-mediated ubiquitin chain initiation and elongation on the purified A3H were tested with or without ubiquitin-conjugating enzymes ARIH2/UBE2L3 and UBE2R1 Mono-ubiquitination and poly-ubiquitination were the most efficient when the K97Q A3H was combined with N48H NL4-3 Vif Source data are provided as a Source Data file In vitro ubiquitination assay showed essentially the same trend, as the addition of K97Q mutation enhanced the rate of mono- and poly-ubiquitination of A3H, and the most efficient ubiquitination pattern was observed when K97Q A3H was combined with N48H Vif (Fig. 1e) these results highlight the determinants of molecular interaction between A3H and Vif and demonstrate that the resulting protein complex is functional in vitro a Domain organization and construct design of A3H and VCBCC complex Gray lines are not included in the construct the invisible regions in the cryo-EM map are colored in gray The key point-mutations for the stable complex assembly are marked with stars b 3.2 Å cryo-EM reconstruction (top) and resultant atomic model (bottom) of the A3H-VCBCC complex and RNA are shown in semi-transparent density superposed with corresponding atomic models (sticks) a Structure of the A3H-Vif subregion in the A3H-VCBCC complex b Close-up view of the interface between A3H α3 and Vif c Close-up view of the interface between A3H α4 and Vif d Interface between A3H loops 4 and 6 and Vif e Schematic of the amino acid residues involved in the A3H-Vif interactions g Vif-mediated degradation assay of A3H mutants Mutations were introduced in the Vif-interface residues around α3 and α4 (f) and around loops 4 and 6 (g) on top of K97Q Vif-sensitivity enhancing mutation The A3H levels in the presence and absence of Vif (+N48H) were probed by Western blots h Vif-mediated degradation assay using Vif mutants Mutations were introduced in the A3H-interface residues on top of N48H A3H levels in the presence of Vif mutants were probed by Western blots Quantified A3H levels are shown in the right chart These results indicate that while the α3 and α4 of A3H are major structural components of Vif-interface the two electrostatic interactions outside the α3-α4 patch are also essential to the A3H-Vif interaction The results also indicate that the A3H E70–Vif R93 pair plays a more dominant role than A3H D100–Vif K92 pair a Spatial relationship of A3H and C/R module for ubiquitin transfer based on the fitted atomic models of A3H-VCBCCR complex b Clustered surface-exposed lysine residues around loops 1 and 3 of A3H c Sequence alignment of primate A3H highlighting the conserved lysine residues in loops 1 and 3 that contain a lysine-rich patch Numbers above the sequence correspond to residue numbering in hA3H d Vif-mediated degradation assay of A3H lysine mutants Lysines in loops 1 and 3 were replaced with arginines which would maintain the positively charged side chains but block ubiquitination The combination of four lysine-to-arginine mutations rendered A3H largely resistant to Vif-mediated degradation The mutations were introduced on top of the K97Q Vif-sensitivity enhancing mutation Quantified A3H levels are shown next to the gel image a Comparison of surface areas responsible for Vif binding and ubiquitination in A3H (left) and A3G (right) b Vif surface residues responsible for A3H binding (sticks in cyan) and A3G-RNA binding (sticks in blue) Y30 is the only residue involved in both complexes (stick in pink) c Atomic model showing key amino-acid contacts modulating the A3H-Vif interaction the glutamine forms a hydrogen bond with Vif K63 in a gain-of-function HIV strain that has adapted to A3H-mediated HIV restriction Vif E63 can form an electrostatic interaction with K97 indicating that no major conformational change in RNA is induced upon the binding to Vif Two complex structures enabled us to map Vif amino acid residues for both A3G-RNA and A3H binding (Fig. 5b) A3G-RNA-binding residues largely reside around the α1 and loop 3 while A3H-binding residues reside around the 5-stranded β-sheets and loop 3 Y30 at the end of α1 is the only residue directly involved in both complex formations Vif loop 3 is also central to both A3G-RNA and A3H binding while no Vif loop 3 residues are directly involved in both A3G and A3H binding both demonstrated that A3H residues on α3 and α4 contribute to the sensitivity to Vif in cell-based degradation assays further generated a structural model by using anchor points revealed by the Vif adaptation experiments they demonstrated that Vif mutation E45Q can restore the degradation of Vif-resistant A3H mutant S86E The cryo-EM structure we obtained here largely agrees with the predicted model we did observe a polar interaction between A3H S86 and Vif E45 in the structure these studies showed that HIV clones with K63E in Vif resulted in dramatically elevated infectivity and enhanced replication kinetics in the presence of A3H expression suggesting more effective inactivation of A3H antiviral activity this gain-of-function mutation may represent a possible evolutionary route for the current HIV strains the critical α4 residue W90 is either glycine or arginine in Old world monkey A3Hs Notwithstanding the molecular details uncovered here for the interface between human A3H and HIV Vif further work is desirable to unveil how Vif-mediated antagonism of A3H is accomplished in other primates and their cognate SIV strains The atomic and chemical details uncovered in this study would be helpful for future HIV therapeutic efforts targeting this fundamental virus-host interface FLAG-A3H-pcDNA or HA-A3H-pcDNA variants were co-transfected with either Vif-pcDNA or pcDNA3.1(+) empty vector into HEK293T cells (ATCC) by using X-tremeGENE 9 DNA Transfection Reagent (Roche) the cells were washed once with PBS and lysed in RIPA buffer with 1× complete protease inhibitors (Roche) The lysates were then subjected to Western blot with anti-FLAG M2 mAb from mouse (Sigma-Aldrich 1:5000) and anti-Vif mAb from mouse (NIH AIDS Reagent Program #319 Cy3-labeled goat-anti-mouse mAb (GE Healthcare 1:3000 dilution) was used as a secondary antibody to detect the signal The fluorescent signal was detected and visualized with Typhoon RGB Biomolecular Imager (GE Healthcare) and His6-RBX2/GB1-CUL5-pETDuet-1 expression vectors were transformed into the E and A3H-pMAL-c5X expression vector was transformed into C43 (DE3) pLysS coli cells harboring the expression vectors were grown in an LB medium at 37 °C until the OD600 reached 0.6 The recombinant proteins were induced by 0.3 mM isopropyl β-D-1-thiogalactopyranoside (IPTG) at 16 °C for 18 h the cell pellets were resuspended with buffer A (20 mM Tris-HCl (pH 8.0) and 0.5 mM TCEP) containing RNase A (0.1 mg/ml and cellular debris was removed by centrifugation The supernatant containing the His6-VCBC complex was loaded onto the Ni-NTA agarose column (Qiagen) The nickel column was extensively washed with wash buffer (20 mM Tris-HCl (pH 8.0) and the protein was eluted with elution buffer (20 mM Tris-HCl (pH 8.0) The His6-tag was cleaved by incubating with PreScission protease overnight VCBC complex was subjected to HiLoad 16/600 Superdex 200 pg column (Cytiva) equilibrated with buffer A The peak fractions were collected and concentrated for mixing with either CUL5-NTD or CUL5/RBX2 complex the cell pellets were resuspended with buffer A The supernatant containing GST-CUL5-NTD was loaded onto glutathione sepharose column (GE Healthcare) The glutathione column was extensively washed with buffer A and the GST tag was cleaved with PreScission protease on the column overnight CUL5-NTD was eluted from the glutathione column and subjected to HiLoad 16/600 Superdex 75 pg column (GE Healthcare) equilibrated with buffer A The peak fractions were collected and concentrated for mixing with the VCBC complex The supernatant containing His6-RBX2/GB1-CUL5 was loaded onto the Ni-NTA agarose column The His6-tag and GB1-tag were cleaved by incubating with PreScission protease overnight CUL5/RBX2 complex was subjected to Superdex 200 Increase 10/300 GL column (Cytiva) equilibrated with buffer A the cell pellets were resuspended with the buffer B (25 mM HEPES-NaOH (pH 7.5) and 0.5 mM TCEP) containing RNase A (0.1 mg/ml) The supernatant containing MBP-A3H was loaded onto amylose column (New England Biolabs) The amylose column was extensively washed with wash buffer (25 mM HEPES-NaOH (pH 7.5) and the protein was eluted with the elution buffer (25 mM HEPES-NaOH (pH 7.5) Eluted fractions were concentrated and subjected to HiLoad 16/600 Superdex 200 pg column (Cytiva) equilibrated with buffer B Peak fractions of dimeric MBP-A3H were separated and concentrated VCBCC complex and VCBCCR complexes were formed by mixing VCBC complex and CUL5-NTD or CUL5/RBX2 by 1:1 molar ratio and incubating on ice for 30 min The protein complex was purified by Superdex 200 Increase 10/300 GL column MBP-tag was cleaved from A3H with PreScission protease A3H-VCBCC and A3H-VCBCCR complexes were formed by mixing them at a 1:1 molar ratio and incubating on ice for 30 min The protein mix was further purified by Superdex 200 Increase 10/300 GL column The peak fraction was isolated and concentrated for cryo-EM work ARIH2, UBE2L3 and UBE2R1 proteins for in vitro ubiquitination assay were purified as previously described37 Protein purity and stoichiometry of the protein complexes were assessed by SDS-PAGE at each purification step Purified MBP-A3H and VCBCCR complex were mixed by 1:1 molar ratio in binding buffer (25 mM HEPES-NaOH (pH 7.5) and 0.5 mM TCEP) and incubated at 4 °C for 30 min The mixture was then subjected to Superdex 200 Increase 10/300 GL column equilibrated with the binding buffer Fractions were concentrated and visualized by SDS-PAGE NEDD8 was conjugated to CUL5 in the VCBCCR complex with either WT or N48H Vif to activate the E3 ubiquitin ligase for recruiting E2 ubiquitin-conjugating enzymes The NEDD8 conjugation reaction was performed by mixing 10 μM VCBCCR complex 0.8 μM E1 NEDD8 activating enzyme (Enzo Life Sciences) 15 U/ml inorganic pyrophosphatase from baker’s yeast (Sigma-Aldrich) 1 mM DTT and incubating for 2.5 h at room temperature NEDD8-conjugated VCBCCR (N8~VCBCCR) complex was purified by Superdex 200 Increase 10/300 GL column to remove excess amount of NEDD8 and other enzymes in the reaction MBP-tag was cleaved from A3H with PreScission protease before the in vitro ubiquitination assay The mono- and poly-ubiquitination of A3H were performed by mixing 1 μM HA-A3H (and its variants) 0.4 μM E1 ubiquitin-activating enzyme (Enzo Life Sciences) 3.6 μM UBE2R1 (for poly-ubiquitination only) 1 mM DTT in 20 μl reaction volume and incubating for 2 h at room temperature The reaction was stopped by adding 2x sample buffer The ubiquitinated products were then subjected to western blot with anti-HA mAb from mouse (Sigma-Aldrich 5 µl of 0.02 mg/ml A3H-VCBCC complex sample was applied onto glow-discharged ultrathin formvar/carbon supported copper 400-mesh grids (EMS) blotted and stained with 2.0% uranyl acetate Negative-stained grids were imaged on a Tecnai F20 transmission electron microscope (FEI) operated at 200 kV The freshly reconstituted protein complex was lightly crosslinked with 1 mM bis-sulfosuccinimidyl suberate (BS3 Thermo Fisher Scientific) on ice for 30 min The reaction was quenched by 50 mM Tris (pH 8.0) for an additional 10 min 4 μl aliquots of 0.15 mg/ml purified A3H-VCBCC complex or A3H-VCBCCR complex were applied to graphene oxide-coated Quantifoil R1.2/1.3 gold 400-mesh grids (Electron Microscopy Sciences) Grids were then blotted and vitrified in liquid ethane using Vitrobot Mark IV (Thermo Fisher Scientific) Vitrified EM grids were screened in Talos F200C (Thermo Fisher Scientific) or Tecnai F20 (FEI) transmission electron microscopes to optimize the freezing conditions Cryo-EM data of the A3H-VCBCC complex were collected in Titan Krios (Thermo Fisher Scientific) equipped with K3 direct electron detector and post-BioQuantum GIF energy filter (Gatan) operated at 300 kV in electron counting mode Movies were collected at a nominal magnification of 165,000× and a pixel size of 0.51 Å A total dose of 50 e−/Å2 per movie was used with a total exposure time of approximately 3.5 s A total of 14,725 movies were recorded by automated data acquisition with EPU (Thermo Fisher Scientific) Cryo-EM data of the A3H-VCBCCR complex were collected in Glacios (Thermo Fisher Scientific) equipped with Falcon-4 direct electron detector operated at 200 kV in electron counting mode Movies were collected at a nominal magnification of 150,000× and a pixel size of 0.92 Å in EER format A total dose of 40 e-/Å2 per movie was used with a dose rate of 5-6 e−/Å2/s A total of 12,546 movies were recorded by automated data acquisition with EPU an extra round of ab initio reconstruction and heterogenous refinement was performed to yield three classes Two good classes from the heterogenous refinement were selected and the particles were re-extracted with full resolution Non-uniform refinement was then performed to yield the final 3.2 Å resolution map Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article Structure-guided analysis of the human APOBEC3-HIV restrictome APOBEC3F properties and hypermutation preferences indicate activity against HIV-1 in vivo The activity spectrum of Vif from multiple HIV-1 subtypes against APOBEC3G Human cytidine deaminase APOBEC3H restricts HIV-1 replication HIV-1 Vif adaptation to human APOBEC3H haplotypes Family-wide comparative analysis of cytidine and methylcytidine deamination by eleven human APOBEC proteins APOBEC3 multimerization correlates with HIV-1 packaging and restriction activity in living cells subcellular localization and mC selectivity of a genomic mutator and anti-HIV factor APOBEC3H The Antiviral and Cancer Genomic DNA Deaminase APOBEC3H Is Regulated by an RNA-Mediated Dimerization Mechanism Structural basis of chimpanzee APOBEC3H dimerization stabilized by double-stranded RNA APOBEC3H structure reveals an unusual mechanism of interaction with duplex RNA The resistance of human APOBEC3H to HIV-1 NL4-3 molecular clone is determined by a single amino acid in Vif The range of human APOBEC3H sensitivity to lentiviral Vif proteins Natural polymorphisms in human APOBEC3H and HIV-1 Vif combine in primary T lymphocytes to affect viral G-to-A mutation levels and infectivity Mapping region of human restriction factor APOBEC3H critical for interaction with HIV-1 Vif Stably expressed APOBEC3H forms a barrier for cross-species transmission of simian immunodeficiency virus of chimpanzee to humans Structural basis of intersubunit recognition in elongin BC-cullin 5-SOCS box ubiquitin-protein ligase complexes The molecular basis of JAK/STAT inhibition by SOCS1 Polymorphisms and splice variants influence the antiretroviral activity of human APOBEC3H A single amino acid difference in human APOBEC3H variants determines HIV-1 Vif sensitivity Determinants of efficient degradation of APOBEC3 restriction factors by HIV-1 Vif Two distinct types of E3 ligases work in unison to regulate substrate ubiquitylation Structural insights into RNA bridging between HIV-1 Vif and antiviral factor APOBEC3G Guidelines for naming nonprimate APOBEC3 genes and proteins Genetic and mechanistic basis for APOBEC3H alternative splicing Antiretroelement activity of APOBEC3H was lost twice in recent human evolution Biochemical characterization of APOBEC3H variants: implications for their HIV-1 restriction activity and mC modification The structural interface between HIV-1 Vif and human APOBEC3H Lentiviral Vif degrades the APOBEC3Z3/APOBEC3H protein of its mammalian host and is capable of cross-species activity Recombinant protein complex expression in E Positive-unlabeled convolutional neural networks for particle picking in cryo-electron micrographs Non-uniform refinement: adaptive regularization improves single-particle cryo-EM reconstruction High-resolution noise substitution to measure overfitting and validate resolution in 3D structure determination by single particle electron cryomicroscopy UCSF Chimera-a visualization system for exploratory research and analysis Towards automated crystallographic structure refinement with phenix.refine Download references Electron microscopy data were collected at the Core Center of Excellence in Nano Imaging (CNI) at USC and California NanoSystems Institute (CNSI) at UCLA which is supported in part by grants from the National Science Foundation (DBI-1338135 and DMR-1548924) Cryo-EM data was computed at the Center for Advanced Research Computing (CARC) at USC and John Curulli for assisting with the operation and maintenance of transmission electron microscopes at CNI Tomek Osinski for assisting with computing work at CARC and Cornelius Gati for advice on cryo-EM sample preparation and data processing This work was funded by NIH grant R01AI150524 to X.S.C was a former fellowship awardee from Nakajima Foundation conceived the project and designed the experiments purified the proteins and reconstituted the protein complex performed the functional biochemical analyses Nature Communications thanks Yasumasa Iwatani and the other anonymous reviewer(s) for their contribution to the peer review of this work Download citation DOI: https://doi.org/10.1038/s41467-023-40955-x Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research Metrics details Great effort has been devoted to discovering the basis of A3G-Vif interaction the key event of HIV’s counteraction mechanism to evade antiviral innate immune response Here we show reconstitution of the A3G-Vif complex and subsequent A3G ubiquitination in vitro and report the cryo-EM structure of the A3G-Vif complex at 2.8 Å resolution using solubility-enhanced variants of A3G and Vif We present an atomic model of the A3G-Vif interface which assembles via known amino acid determinants This assembly is not achieved by protein-protein interaction alone The cryo-EM structure and in vitro ubiquitination assays identify an adenine/guanine base preference for the interaction and a unique Vif-ribose contact This establishes the biological significance of an RNA ligand Further assessment of interactions between A3G and RNA ligands show that the A3G-Vif assembly and subsequent ubiquitination can be controlled by amino acid mutations at the interface or by polynucleotide modification suggesting that a specific chemical moiety would be a promising pharmacophore to inhibit the A3G-Vif interaction Source data are provided as a Source data file f Cryo-EM reconstruction of the sA3G-VC-RNA20 complex at 2.8 Å resolution The complex is a C2-symmetric dimer (right) The asymmetric unit (left) is colored in the same manner as in (b) g Ribbon presentation of the atomic model of sA3G-VC-RNA20 complex and dissection of its components The orientation and coloring scheme follow the right panel in (f) Our cryo-EM structure shows interactions between A3G-Vif and in vitro ubiquitination assays identify key adenine and guanine bases for the Vif-induced ubiquitination of A3G Selected side chains and nucleotides are presented as stick models and labeled Predicted hydrogen bonds between the ribose 2’-hydroxyl group and side chains H42/H43 of Vifred are represented by dashed lines (d) f Stereo views of the atomic model showing nucleotide rA7 (e) and rA18 (f) accommodations in protein surface pockets of sA3G and Vifblue Polypeptides and nucleotides are shown as stick models or in ribbon representation Predicted hydrogen bonds between protein and nucleotides are depicted by cyan-colored dashed lines It is noteworthy that Vifblue H43 and W70 contact sA3G Y19 whereas the same Vif residues face sA3G W127 and D128 in the sA3G-Vifred interface These intermolecular interactions would compensate for an entropic penalty upon the complex assembly to confer base-specificity of the RNA ligand The molecular arrangement of this pocket likely excludes the O6 atom of guanine Adenine satisfies the geometry and interactions with chemical moieties of surrounding amino acids nucleotide rC17 is exposed to the protein exterior The atomic model shows that phosphate groups of rC17 and rA18 are within hydrogen bonding distance of side chains of T20 and R17 the impact of T32 phosphorylation on degradation is also likely caused by exclusion of the ligand RNA Although these amino acids are located away from the sA3G-Vif interface they mediate complex formation indirectly through interactions with RNA Our cryo-EM map and biochemical data demonstrate that RNA mediates both sA3G-Vifred and sA3G-Vifblue assemblies The RNA20 ligand increases the interface areas by up to ~1500 A2 and ~950 A2 for sA3G-Vifred-RNA20 and sA3G-Vifblue-RNA20 thus maximizing domain interactions and stabilizing the complex It explains how RNA20 captures the sA3G-VC complex and enables structure determination by cryo-EM participate in base-specific interactions with their protein binding partners sA3G-VC assembly can be promoted by various RNA sequences We further assessed base specificity and impact on sA3G ubiquitination The RNA20 model is drawn in cartoon representation binding sites on sA3G with Vifred and RNA20 are colored red and yellow whereas interaction sites on Vifred with sA3G and RNA20 are colored green and yellow sA3G interacts with Vifblue and RNA20 via regions colored blue and yellow while Vifblue binds to sA3G and RNA20 through regions colored green and yellow f–i Electrostatic potential surface representations of sA3G without (f) and with (g) RNA20 Calculated surface potentials are colored with a gradient from red (negative) to blue (positive) The electrostatic potential scale is indicated in (i) a In vitro ubiquitination assay using sA3G and its mutants Substrates were specifically detected using a C-terminally-tagged c-myc sequence (lanes 1 and 2) Ubiquitination of sA3G required ARIH2 and ubiquitin with a C-terminal G76 (lanes 3 and 4) The reaction was enhanced in the presence of RNA20 (compare lanes 5 and 8) D128K (lane 6) and K4Rs (K297R/K301R/K303R/K334R) (lane 7) appeared to reduce polyubiquitination beyond diubiquitin Most of sA3G was ubiquitinated within 1 h of reaction (lane 9) The band of unreacted sA3G is indicated by an arrow sA3G was depleted within 30 min (lanes 1–4) whereas the ubiquitination was attenuated by sA3G mutation D128K (lanes 5–8) DNA20 showed almost no sA3G ubiquitination (lanes 9–12) c Quantification of unreacted sA3G during the reaction presented in (b) Assays were performed independently in triplicate (n = 3) e Impact of dinucleotide rG6rA7 on sA3G ubiquitination Reactants were analyzed by western blotting (d) The amount of unreacted sA3G was quantified (e) A significant (p < 0.05 by two-sided t test no adjustments) decrease in unreacted sA3G is indicated by an asterisk compared with that in presence of U20; p values are 0.0095 sA3G was depleted in the presence of U20-rGrA (d whereas U20 showed a much slower reaction (d U20-rUrA retained some ability to enhance sA3G ubiquitination (d lanes 9–12) and U20-rGrU (e) significantly lost the ability to enhance sA3G ubiquitination f Close-up view of dinucleotide rG6rA7 at the sA3G-Vifred interface The solvent-accessible surface of sA3G is shown in green and side chains of Vifred H42/H43 are drawn in stick representation and labeled Predicted hydrogen bonds are depicted as dashed yellow lines Source data are provided as a Source data file (a–e) The molecular models show that U9-rGrA and U7-rGrA are too short to provide a large negatively charged surface for Vif interaction our in vitro ubiquitination and in cell degradation assays showed that the dinucleotide rG6rA7 serves an essential biological function in conferring local base preference upon the RNA ligand to promote A3G-Vif interaction The length of RNA ligand is likely important for the enhancement of the A3G-Vif interaction it is plausible that A3G-Vif and A3G-Gag interactions may compete with each other and reduce encapsidation of A3G a, b Comparison of Vif-binding interfaces from sA3G (a) (this study) and A3F (b)40 The molecular orientation of A3 proteins is indicated using a cartoon representation (bottom right inset) whereas the interface on A3F with a single Vif is colored red (b) c Predicted model of the entire ubiquitin ligase complex The model was built using alignments of atomic coordinates obtained from our cryo-EM structure and PDB IDs 4N9F d Solvent-accessible surface representations of the predicted ubiquitin ligase complex linked through sA3G-Vifred-CUL5 Amino acids sA3G K297/K301/K303/K334 and ubiquitin G75 are drawn as space-filled models and labeled e Unprocessed TEM micrograph of a negatively stained sample from a mixture of in vitro ubiquitination assay reactant The image reflects many states of the ongoing reaction Arrowheads indicate possible U-shaped ubiquitin ligase complex particles f 2D class average images of putative U-shaped ubiquitin ligase complex particles The 6 classes were composed of 1095 particles harvested from 179 micrographs g Superimposition of the predicted ubiquitin ligase complex model (d) on a class average image from (f) our high-resolution cryo-EM structure of sA3G-VC-RNA20 revealed that Vif binds sA3G through interactions with ssRNA Our in vitro and in cellulo experiments showed that the sA3G-Vif-RNA trimolecular interface involving the dinucleotide rG6rA7 is most critical for ubiquitination of sA3G; therefore this trimolecular interface is likely used to form a ubiquitin E3-ligase complex The good fit of the predicted ubiquitin ligase complex model with 2D class average images holds promise for determining the structure of the fully assembled ubiquitin ligase complex which may provide further understanding of structure-activity relationships of A3G ubiquitination Preparation procedures were partially modified and are briefly described as follows Expression plasmids for glutathione S-transferase (GST)-fused sNTD or sA3G and its mutants were constructed using pCold-GST vectors (a gift from Dr coli strain BL21 (DE3) was transformed using the plasmid and cultivated in LB media at 37 °C expression was induced at 20 °C by adding 0.2 mM isopropyl-β-thiogalactopyranoside (IPTG) cells were harvested and resuspended in buffer-1 [50 mM sodium phosphate and 0.5-mM Tris(2-carboxyethyl)phosphine hydrochloride (TCEP)] and the lysate was centrifuged at 20,000 × g for 30 min The supernatant was applied to glutathione-immobilized resin (Genscript) equilibrated in buffer-1 The protein-bound resin was washed with buffer-2 (50 mM Tris-HCl An aliquot of HRV 3 C protease solution (Fujifilm Wako Pure Chemical Japan) was added to the resin resuspension and the mixture was incubated at 4 °C overnight The supernatant was applied to a Superdex 200 column (Cytiva) equilibrated with buffer-3 (25 mM Tris-HCl Fractionated protein was concentrated and kept in a freezer at −80 °C until further use Protein purity was verified on 12% or 15% polyacrylamide gels followed by Coomassie brilliant blue staining or on 4–12% gradient Bis-Tris gels (Invitrogen) stained with SimplyBlue SafeStain (Invitrogen) sA3G mutants and C-terminally c-myc-tagged constructs were prepared in the same manner Vif and CBFβ constructs shown in Fig. 1b were inserted into BamHI/HindIII and NdeI/XhoI sites coli strain BL21 (DE3) was transformed using this plasmid and cultivated in LB media at 37 °C cells were harvested and resuspended in buffer-2 supplemented with 20 mM imidazole and 10% dimethyl sulfoxide (DMSO) The supernatant was applied to buffer-2-equilibrated Ni-NTA resin (Fujifilm Wako Pure Chemical) bound proteins were eluted with buffer-2 supplemented with 200 mM imidazole and 500 mM dimethylethylammonium propane sulfonate Eluent was loaded onto a Superdex 200 column equilibrated in buffer 2 and the mixture was concentrated and kept in a freezer at −80 °C until further use ubiquitin-conjugating enzyme E2L3 (M1 to D154) NEDD8-conjugating enzyme UBE2F (M1 to R185) were codon-optimized for bacterial expression and synthesized (Genscript) Their mutants were constructed using a PCR-based technique and UBE2F were individually inserted into NdeI/XhoI sites on pCold-GST vectors Protein expression and purification followed the procedure for sA3G preparation described above DNAs encoding CUL5 and Rbx2 constructs were inserted into the BamHI/XhoI site on a pGEX-6P-1 vector (Cytiva) and into BamHI/HindIII sites on a pRSFDuet vector (Novagen) Proteins CUL5 and Rbx2 were co-expressed and co-purified in a manner similar to sA3G preparation DNAs encoding NAE1 and UBA3 were inserted into BamHI/HindIII and NdeI/XhoI sites NAE1/UBA3 preparation followed the method for VC purification described above Obtained protein solutions were concentrated and then frozen at −80 °C until further use Residue numbering follows wild-type constructs The fraction of sNTD-F126 in complex with VCBC was prepared for RNA extraction with TRIzol and precipitated with ethanol The pellet was used for a serial enzymatic reaction and for the cloning: 3′-end of RNA was extended by poly(A) polymerase (New England BioLabs) 5′-adapter (5′-rGrUrUrCrArGrArGrUrUrCrUrArCrArGrUrCrCrGrArCrGrArUrC-3′) was ligated with T4 RNA ligase (New England BioLabs) followed by reverse transcription with a poly-thymidine DNA oligo and amplification of the cDNA by polymerase chain reaction (PCR) using Taq DNA polymerase (Takara Bio Resulting DNAs were inserted into pMD20-T vectors (Takara Bio and clones were subjected to DNA sequencing Purified sA3G, VC and a synthesized RNA oligomer (see Table 1; IDT technologies Japan) were mixed at a ratio of 1:1.5:1.5 in buffer-3 supplemented with 200 mM NDSB-195 The mixture was applied to a Superdex 200 column equilibrated in buffer-3 and purified by size-exclusion chromatography The largest fraction of the complex was used for negative staining EM and cryo-EM grid preparations immediately after collection The relative amount of harvested complex was monitored by UV absorbance at 280 nm (A280) and RNA binding was roughly estimated by the A260/A280 ratio The prepared sA3G-VC-RNA complex typically had a ratio of 0.98–1.02 and VCBC were mixed at a final concentration of 5 μM each with 10 μM NEDD8 and 1 mM of freshly prepared ATP in an assay buffer The mixture was incubated at 20 °C for at least 1 h NEDDylation of CUL5 was verified by PAGE analysis A 10-μL aliquot of the resulting mixture was mixed with 1 μM sA3G or its mutant and the total volume was adjusted to 50 μL with assay buffer and an aliquot of the reactant was harvested for further analysis after 0 The concentration of freshly prepared sA3G-VC-RNA complex solution was adjusted so that the UV absorbance at 280 nm was in the range of 0.30–0.33 A holey EM grid (Quantifoil R1.2/1.3) was plasma-cleaned (Solarus II Gatan Inc.) and treated with graphene oxide film flakes (Sigma Aldrich) 3 μL of sample solution was deposited onto the grid at 4 °C and 100% humidity The sample grid was blotted with filter paper for 3 s and vitrified by plunging it into ethane/propane at liquid nitrogen temperature on a Vitrobot Mark IV (Thermo Fisher Scientific) Vitrified sample grids were stored in liquid nitrogen until further use All cryo-EM data were collected semi-automatically using EPU software (Thermo Fisher Scientific) Electrostatic potentials were calculated with the program statistical tests were conducted to evaluate the statistical significance between two datasets: F-test for evaluation of standard deviation equivalency and t-test for assessment of significant difference between two data sets The significant difference was defined as less than 0.05 of the p value Source data and uncropped unprocessed scans are provided as a Source data file Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article Structural perspectives on HIV-1 Vif and APOBEC3 restriction factor interactions Suppression of APOBEC3-mediated restriction of HIV-1 by Vif APOBEC3G DNA deaminase acts processively 3’ -> 5’ on single-stranded DNA The cytidine deaminase CEM15 induces hypermutation in newly synthesized HIV-1 DNA Cone-shaped HIV-1 capsids are transported through intact nuclear pores Vif Hijacks CBF-β to degrade APOBEC3G and promote HIV-1 infection Structural basis for hijacking CBF-β and CUL5 E3 ligase complex by HIV-1Vif Defining APOBEC3 expression patterns in human tissues and hematopoietic cell subsets Structure of the DNA deaminase domain of the HIV-1 restriction factor APOBEC3G Definition of the interacting interfaces of Apobec3G and HIV-1 Vif using MAPPIT mutagenesis analysis Amino-terminal region of the human immunodeficiency virus type 1 nucleocapsid is required for human APOBEC3G packaging Specific packaging of APOBEC3G into HIV-1 virions is mediated by the nucleocapsid domain of the gag polyprotein precursor Human apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like 3G (APOBEC3G) is incorporated into HIV-1 virions through interactions with viral and nonviral RNAs APOBEC3G incorporation into human immunodeficiency virus type 1 particles Complementary function of the two catalytic domains of APOBEC3G catalytically active human APOBEC3G: correlation with antiviral effect Crystal structure of the catalytic domain of HIV-1 restriction factor APOBEC3G in complex with ssDNA Computational model and dynamics of monomeric full-length APOBEC3G High-speed atomic force microscopy directly visualizes conformational dynamics of the HIV Vif protein in complex with three host proteins The HIV-1 Vif PPLP motif is necessary for human APOBEC3G binding and degradation Crystal structure of APOBEC3A bound to single-stranded DNA reveals structural basis for cytidine deamination and specificity Insights into DNA substrate selection by APOBEC3G from structural Single-stranded RNA facilitates nucleocapsid: APOBEC3G complex formation Promiscuous RNA binding ensures effective encapsidation of APOBEC3 proteins by HIV-1 NIH Image to ImageJ: 25 years of image analysis CTFFIND4: fast and accurate defocus estimation from electron micrographs Macromolecular structure determination using X-rays neutrons and electrons: recent developments in Phenix MolProbity: more and better reference data for improved all-atom structure validation Scalable molecular dynamics on CPU and GPU architectures with NAMD Download references This research was supported by the Platform Project for Supporting Drug Discovery and Life Science Research (BINDS) from AMED under grant number JP18am0101076 and by direct funding from OIST (to M.W.); the Toyobo Biotechnology Foundation (to T.K.) and T.K.‘s personal funds was supported in part by a grant from the U.S National Institutes of Health R01GM118474/R01AI150478 and federal funds from the National Cancer Institute Chojiro Kojima at Osaka University for providing pCold-GST vectors Keiko Kono at OIST for advice on human cell experiments and western blotting analysis Melissa Matthews for critical reading of the manuscript Aird for technical editing of the manuscript We thank the OIST Imaging and Analysis Section (IMG) for use of the EM facility and the OIST Scientific Data Analysis Section (SCDA) for use of the Deigo and Saion high performance computing clusters Adrian Koh for collecting cryo-EM test data of sA3G-VC-RNA-IV-20 on the Titan Krios G4 Selectris-X at Thermo Fisher Scientific Eindhoven Present address: Department of Efficacy Evaluation Okinawa Institute of Science and Technology Graduate University Frederick National Laboratory for Cancer Research designed sA3G and all wild-type A3G/sA3G mutants designed all DNA/RNA/hybrid oligomer sequences cloned all protein constructs used in this study prepared grapheneoxide-coated cryo-EM grids processed cryo-EM data with support of S.S. conducted atomic model building and refinement analyzed molecular structures and recapitulated the entire project All authors read and contributed to writing the manuscript supervised and provided guidance on all cryo-EM experiments has filed patents on preparation of the solubility-enhanced human A3G construct its application for A3G-Vif complex analysis [PCT-JP2019-019938 (2019) and JP patent application number 2021-519919 (2021)]; on DNA/RNA/hybrid-based antagonism against A3G-Vif interaction [PCT-JP2021-42773 (2021)] Nature Communications thanks the anonymous reviewers for their contribution to the peer review of this work Download citation DOI: https://doi.org/10.1038/s41467-023-39796-5 Metrics details HIV-1 virion infectivity factor (Vif) promotes degradation of the antiviral APOBEC3 (A3) proteins through the host ubiquitin-proteasome pathway to enable viral immune evasion Disrupting Vif-A3 interactions to reinstate the A3-catalyzed suppression of human immunodeficiency virus type 1 (HIV-1) replication is a potential approach for antiviral therapeutics the molecular mechanisms by which Vif recognizes A3 proteins remain elusive Here we report a cryo-EM structure of the Vif-targeted C-terminal domain of human A3F in complex with HIV-1 Vif and the cellular cofactor core-binding factor beta (CBFβ) at 3.9-Å resolution The structure shows that Vif and CBFβ form a platform to recruit A3F revealing a direct A3F-recruiting role of CBFβ beyond Vif stabilization and captures multiple independent A3F-Vif interfaces Together with our biochemical and cellular studies our structural findings establish the molecular determinants that are critical for Vif-mediated neutralization of A3F and provide a comprehensive framework of how HIV-1 Vif hijacks the host protein degradation machinery to counteract viral restriction by A3F Other data are available from corresponding authors upon reasonable request T-cell differentiation factor CBF-β regulates HIV-1 Vif-mediated evasion of host restriction Evolution of the AID/APOBEC family of polynucleotide (deoxy)cytidine deaminases Identification of specific determinants of human APOBEC3F and APOBEC3DE and African green monkey APOBEC3F that interact with HIV-1 Vif The retroviral hypermutation specificity of APOBEC3F and APOBEC3G is governed by the C-terminal DNA cytosine deaminase domain Core binding factor beta plays a critical role by facilitating the assembly of the Vif-cullin 5 E3 ubiquitin ligase CBFβ stabilizes HIV Vif to counteract APOBEC3 at the expense of RUNX1 target gene expression Structural basis for hijacking CBF-β and CUL5 E3 ligase complex by HIV-1 Vif Crystal structure of the DNA cytosine deaminase APOBEC3F: the catalytically active and HIV-1 Vif-binding domain Structural insights into HIV-1 Vif-APOBEC3F interaction Identification of 81LGxGxxIxW89 and 171EDRW174 domains from human immunodeficiency virus type 1 Vif that regulate APOBEC3G and APOBEC3F neutralizing activity The APOBEC3C crystal structure and the interface for HIV-1 Vif binding A single amino acid in human APOBEC3F alters susceptibility to HIV-1 Vif Transcriptional regulation of APOBEC3 antiviral immunity through the CBF-β/RUNX axis The assembly of Vif ubiquitin E3 ligase for APOBEC3 degradation The binding interface between human APOBEC3F and HIV-1 Vif elucidated by genetic and computational approaches Multifaceted counter-APOBEC3G mechanisms employed by HIV-1 Vif HIV-1 viral infectivity factor (Vif) alters processive single-stranded DNA scanning of the retroviral restriction factor APOBEC3G Structural insights into NEDD8 activation of cullin-RING ligases: conformational control of conjugation Phosphorylation of a novel SOCS-box regulates assembly of the HIV-1 Vif-Cul5 complex that promotes APOBEC3G degradation Evolutionary paradigms from ancient and ongoing conflicts between the lentiviral Vif protein and mammalian APOBEC3 enzymes Evolution of HIV-1 isolates that use a novel Vif-independent mechanism to resist restriction by human APOBEC3G Long-term restriction by APOBEC3F selects human immunodeficiency virus type 1 variants with restored Vif function HIV-1 and HIV-2 Vif interact with human APOBEC3 proteins using completely different determinants Codon optimization of the HIV-1 vpu and vif genes stabilizes their mRNA and allows for highly efficient Rev-independent expression Generation of high-titer pseudotyped retroviral vectors with very broad host range Cytokine signals are sufficient for HIV-1 infection of resting human T lymphocytes Identification of a tripartite interaction between the N-terminus of HIV-1 Vif and CBFβ that is critical for Vif function Gctf: real-time CTF determination and correction Prevention of overfitting in cryo-EM structure determination UCSF Chimera—a visualization system for exploratory research and analysis REFMAC5 for the refinement of macromolecular crystal structures Download references Devarkar and other Xiong lab members for discussions This work was supported by National Institutes of Health grant AI116313 (Y.X.) and by an Intramural AIDS Targeted Antiviral Program grant and the Innovation Fund Present address: Department of Biochemistry and Biophysics The authors declare no competing financial interests Peer review information Inês Chen was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team The Vif–CBFβ–A3FCTDm fusion complex with or without the Vif α-domain and corresponding interacting CBFβ C terminus stays as a tetramer in low-salt solution The unfused Vif–CBFβ–A3FCTDm without these regions switches from monomer to tetramer at high protein concentration (146 μΜ loading concentration) No obvious shift for the elution peak was observed upon incubation of CBFβ and A3FCTDm compared to the CBFβ alone or A3FCTDm alone The SDS-PAGE analysis of the peak fractions of CBFβ alone A3FCTDm alone and CBFβ/A3FCTDm mixture is indicated Co-immunoprecipitation (Co-IP) analysis of the interaction between A3F and CBFβ in the presence or absence of Vif in cells Flag-A3F and CBFβ-myc were cotransfected with or without Vif-HA and co-immunoprecipitated using an anti-Flag antibody no binary A3F and CBFβ binding was observed A representative blot from two independent experiments was shown The 5-Å cryo-EM reconstruction of Vif–CBFβ–A3FCTDm with (right) or without (left) the docked-in model (ribbon) The density corresponding to the flexible Vif α-domain and the corresponding interacting CBFβ C terminus (circled) is not visible the 3.9-Å cryo-EM reconstruction of the truncated Vif–CBFβ–A3FCTDm overlay of the cryo-EM models of Vif–CBFβ–A3FCTDm ternary complexes with (magenta) and without (yellow) the Vif α-domain and the corresponding interacting CBFβ C terminus shows that the removal of these regions does not affect the architecture of the ternary complex Central slices of the top 3D classes of the Vif–CBFβ–A3FCTDm complex with (upper) or without (lower) the Vif α-domain and the corresponding interacting CBFβ C terminus indicate that removing these flexible regions reduces the tetramer flexibility The location of the Vif α-domain and the corresponding interacting CBFβ C terminus is marked by yellow arrows in the first class average The color spectrum and the coil thickness represent the deviation of the aligned Cα atoms in the structures, which varies from 0 Å (blue) to ~10 Å (orange). The Vif C-terminal residues 173–176 missing in the Vif–E3 ligase structure are colored in red. The Vif–CBFβ structure without A3FCTDm binding used for superposition is extracted from the Vif–E3 ligase structure (PDB 4N9F) The effect of D347R mutation on A3F sensitivity to Vif-mediated degradation indicated by western blot (left) quantified A3F levels relative to no Vif (mean ± s.d.; n = 4 biologically independent experiments; middle) and relative infectivity (mean ± s.d.; n = 4 biologically independent experiments; right) A3FD347R retained wild-type A3F-like sensitivity to Vif-mediated degradation which resulted in rescue of viral infectivity The effect of D260A/D261A or D260R/D261R double mutants on A3F sensitivity to Vif-mediated degradation indicated by western blot (left) quantified A3F levels relative to no Vif (mean ± s.d.; n = 3 biologically independent experiments; middle) and relative infectivity (mean ± s.d.; n = 3 biologically independent experiments; right) Both alanine and arginine mutants did not confer resistance indicating that the side chains of the residues are not involved in the Vif interaction Western blot (a) and quantified A3 levels relative to ‘no Vif’ (b) show that the Vif K50E mutant could not induce A3F degradation in the presence of either CBFβ wild-type or CBFβ E54K but could induce A3G degradation (mean ± s.d.; n = 3 biologically independent experiments) the charge-swapped Vif R15E/A3F E289K double mutation did not restore the Vif-mediated A3F degradation or viral infectivity in cells The blot was cut as indicated (gray arrow) where one half was used to detect Flag-A3F and HSP90 and the other half was used to detect Vif-HA left) blocks the catalytic site of A3FCTDm (red and marked by an arrow) One Vif–CBFβ–A3FCTDm ternary complex with the major interface is circled with an oval FSC curves of the half-maps from gold standard refinements of the Vif–CBFβ–A3FCTDm complex with (cyan) or without (blue) the Vif α-domain and the corresponding interacting CBFβ C terminus The FSC curve of the map and final model of the truncated Vif–CBFβ–A3FCTDm complex is in green Resolution of the maps are determined by the cut-off values at FSC = 0.143 The Euler angle distribution of the classified particles of the truncated Vif–CBFβ–A3FCTDm complex used for the final 3D reconstruction Color coded local resolution estimation of the D2 symmetrized map of the truncated Vif–CBFβ–A3FCTDm complex a−c, Detailed illustrations of the secondary structure elements of Vif176 (a), CBFβ151 (b) and A3FCTD (c). The secondary structures are annotated on primary amino acid sequences (left) and tertiary structures (right). The tertiary structures for illustration are: Vif, extracted from PDB 4N9F; CBFβ151, extracted from our cryo-EM structure; A3FCTD, PDB 3WUS Download citation DOI: https://doi.org/10.1038/s41594-019-0343-6 Metrics details HIV-1 infection-induced cGAS–STING–TBK1–IRF3 signaling activates innate immunity to produce type I interferon (IFN) The HIV-1 nonstructural protein viral infectivity factor (Vif) is essential in HIV-1 replication as it degrades the host restriction factor APOBEC3G whether and how it regulates the host immune response remains to be determined we found that Vif inhibited the production of type I IFN to promote immune evasion HIV-1 infection induced the activation of the host tyrosine kinase FRK which subsequently phosphorylated the immunoreceptor tyrosine-based inhibitory motif (ITIM) of Vif and enhanced the interaction between Vif and the cellular tyrosine phosphatase SHP-1 to inhibit type I IFN the association of Vif with SHP-1 facilitated SHP-1 recruitment to STING and inhibited the K63-linked ubiquitination of STING at Lys337 by dephosphorylating STING at Tyr162 the FRK inhibitor D-65495 counteracted the phosphorylation of Vif to block the immune evasion of HIV-1 and antagonize infection These findings reveal a previously unknown mechanism through which HIV-1 evades antiviral immunity via the ITIM-containing protein to inhibit the posttranslational modification of STING These results provide a molecular basis for the development of new therapeutic strategies to treat HIV-1 infection Carbohydrate-specific signaling through the DC-SIGN signalosome tailors immunity to Mycobacterium tuberculosis HIV-1 exploits innate signaling by TLR8 and DC-SIGN for productive infection of dendritic cells Type I interferon responses by HIV-1 infection: association with disease progression and control Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8 HIV-1 single-stranded RNA induces CXCL13 secretion in human monocytes via TLR7 activation and plasmacytoid dendritic cell-derived type I IFN Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses IFI16 senses DNA forms of the lentiviral replication cycle and controls HIV-1 replication The capsids of HIV-1 and HIV-2 determine immune detection of the viral cDNA by the innate sensor cGAS in dendritic cells IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV Sequence-specific activation of the DNA sensor cGAS by Y-form DNA structures as found in primary HIV-1 cDNA Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA NONO detects the nuclear HIV capsid to promote cGAS-mediated innate immune activation IFI16 targets the transcription factor Sp1 to suppress HIV-1 transcription and latency reactivation Structural basis of STING binding with and phosphorylation by TBK1 Interferon responses in HIV infection: from protection to disease Type I interferon and HIV: subtle balance between antiviral activity The macrophage in HIV-1 infection: from activation to deactivation The cytokine network of acute HIV infection: a promising target for vaccines and therapy to reduce viral set-point Cyclophilin A modulates the sensitivity of HIV-1 to host restriction factors CPSF6 defines a conserved capsid interface that modulates HIV-1 replication HIV-1 evades innate immune recognition through specific cofactor recruitment Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu HIV-1 Vif binds to APOBEC3G mRNA and inhibits its translation Restricting HIV the SAMHD1 way: through nucleotide starvation Restriction by SAMHD1 limits cGAS/STING-dependent innate and adaptive immune responses to HIV-1 HIV-1 blocks the signaling adaptor MAVS to evade antiviral host defense after sensing of abortive HIV-1 RNA by the host helicase DDX3 NLRX1 sequesters STING to negatively regulate the interferon response thereby facilitating the replication of HIV-1 and DNA viruses HIV-1 and interferons: who’s interfering with whom The cytosolic exonuclease TREX1 inhibits the innate immune response to human immunodeficiency virus type 1 Immunoreceptor tyrosine-based inhibition motifs: a quest in the past and future Inhibition of TLR signaling by a bacterial protein containing immunoreceptor tyrosine-based inhibitory motifs The E3 ligases Itch and WWP2 cooperate to limit TH2 differentiation by enhancing signaling through the TCR The ubiquitin ligase TRIM56 regulates innate immune responses to intracellular double-stranded DNA TRIM32 protein modulates type I interferon induction and cellular antiviral response by targeting MITA/STING protein for K63-linked ubiquitination TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity The ubiquitin E3 ligase TRIM31 promotes aggregation and activation of the signaling adaptor MAVS through Lys63-linked polyubiquitination The tyrosine kinase FRK/RAK participates in cytokine-induced islet cell cytotoxicity Innate immune sensing of HIV-1 by dendritic cells Shp1 regulates T cell homeostasis by limiting IL-4 signals Activation of phosphotyrosine phosphatase activity attenuates mitogen-activated protein kinase signaling and inhibits c-FOS and nitric oxide synthase expression in macrophages infected with Leishmania donovani SAMHD1 suppresses innate immune responses to viral infections and inflammatory stimuli by inhibiting the NF-kappaB and interferon pathways Structural insight into the human immunodeficiency virus Vif SOCS box and its role in human E3 ubiquitin ligase assembly The Phyre2 web portal for protein modeling Download references This work was supported by grants from the Program of Shanghai Academic Research Leader (21XD1402900) the Natural Science Foundation of Shanghai (21ZR1481400) the National Natural Science Foundation of China (31972900) the National Youth Talent Support Program (Ten Thousand Talent Program) the National Key Research and Development Program of China (2018YFC1705505) and the National Megaproject on Key Infectious Diseases (2017ZX10202102) Sun (Shanghai Institute of Biochemistry and Cell Biology Germany) for pBR322-HIV-1-M-NL4-3-IRES-eGFP env STOP plasmid (pseudotyping is required for infection) China) for cDNAs encoding SRC-family kinases These authors contributed equally: Yu Wang Shanghai Institute of Infectious Disease and Biosecurity & Shanghai Public Health Clinical Center National Engineering Research Centre of Immunological Products Department of Microbiology and Biochemical Pharmacy National Engineering Laboratory for AIDS Vaccine Shanghai Key Laboratory of Organ Transplantation Zhongshan Hospital & Institutes of Biomedical Sciences and DY conceived the project and designed the experiments and ZZ performed most of the experiments and analyzed the data and TX assisted with the experiments and provided technical help and XZ provided comments and assisted with manuscript preparation Download citation DOI: https://doi.org/10.1038/s41423-021-00802-9 The GroupLimagrain is a seed and agri-food group owned by French farmers based in the heart of central France’s Auvergne region. Why join us ?Discover our job familiesView our vacancies enfrPassionately reveals the best in plants. Founded in 2021 in the Auvergne-Rhône-Alpes region, VIF Systems (Vertical Innovative Farming) is a major French player in vertical farm equipment combining technical, digital and economic innovation for the production of seedlings in a controlled environment. For Limagrain, this partnership will ensure the long-term future of the family farming model specific to the Limagne-Val d'Allier region (Auvergne), while fitting in with its strategy of developing new value chains. Genuine industrial short circuits, these channels provide the Cooperative's farmer-members with high value-added outlets. They contribute to the sustainability of farms, and to the dynamism and attractiveness of the region, making it a center of agricultural excellence. The Group Business Lines The dates displayed for an article provide information on when various publication milestones were reached at the journal that has published the article activities on preceding journals at which the article was previously under consideration are not shown (for instance submission All content on this site: Copyright © 2025 Elsevier B.V. All content on this site: Copyright © 2025 Elsevier B.V., its licensors, and contributors. All rights are reserved, including those for text and data mining, AI training, and similar technologies. For all open access content, the relevant licensing terms apply. Volume 11 - 2020 | https://doi.org/10.3389/fmicb.2020.622012 Accessory proteins are a key feature that distinguishes primate immunodeficiency viruses such as human immunodeficiency virus type I (HIV-1) from other retroviruses A prime example is the virion infectivity factor which hijacks a cellular co-transcription factor (CBF-β) to recruit a ubiquitin ligase complex (CRL5) to bind and degrade antiviral APOBEC3 enzymes including APOBEC3D (A3D) Although APOBEC3 antagonism is essential for viral pathogenesis and a more than sufficient functional justification for Vif’s evolution most viral proteins have evolved multiple functions Vif has long been known to trigger cell cycle arrest and recent studies have shed light on the underlying molecular mechanism Vif accomplishes this function using the same CBF-β/CRL5 ubiquitin ligase complex to degrade a family of PPP2R5 phospho-regulatory proteins These advances have helped usher in a new era of accessory protein research and fresh opportunities for drug development HIV-1 genome organization and counteraction of host-cell restriction factors by viral accessory proteins (A) Depiction of the genomic organization of HIV-1 with yellow indicating structural proteins and viral enzymes green indicating the envelope glycoprotein and red highlighting the viral accessory proteins (B) Simplified schematic of the HIV-1 life-cycle with activities of the viral accessory proteins highlighted in red Nef and Vpu downmodulate the anti-viral factors TETHERIN and SERINC3/5 from the cell surface Vif targets cytoplasmic APOBEC3s for proteasomal degradation prior to viron encapsidation Vpr antagonizes many different cellular processes including DNA-damage response APOBEC3 restriction and counter action by Vif (A) Schematic of the APOBEC3 locus with anti-HIV-1 activity indicated above or below the respective APOBEC3 enzyme (B) Depiction of APOBEC3 C-to-T mutations during conversion of the viral genomic RNA (gRNA) to cDNA Mutations are depicted at either “CC” or “TC” dinucleotide contexts which is reflective of A3G or A3D/F/H target sites Fixation of the mutations is depicted by the final double-stranded DNA product (right) (C) Structure of the E3-ubiquitin ligase complex nucleated by Vif (PDB: 4N9F) Full length CUL5 has been modeled in using the partial CUL5 sequence from PDB: 4N9F RBX2 has been modeled in using RBX1 from PDB: 1LDJ as a template The inset depicts the Vif/CBF-β hetero-dimer as ribbon diagrams from the crystal structure (D) Amino acid sequences and surface models of Vif with substrate binding residues highlighted in the indicated color The shared surface indicated residues that are required for degradation of two or more Vif substrates these biochemical constraints suggest that a single Vif/E3-complex can only degrade one substrate at a time making Vif’s ability to target so many distinct substrates even more impressive While these observations collectively pointed toward Vif degrading a cellular factor to induce arrest the identity of this factor remained elusive for nearly 10 years following the discovery of this activity The PPP2R5 surface recognized by Vif is extremely electronegative and supports a model in which Vif maintains this interaction through a favorable network of electrostatics all of the residues required for Vif recognition are conserved among all five family members which clarifies how Vif can recognize and degrade five new cellular substrates and explains genetic evidence that loss of at least two family members is required for inducing G2/M arrest (A) Amino acid residues on PPP2R5 and APOBEC3 that have been shown to be required for Vif-mediated degradation (top) with substrate surfaces (middle) and electrostatic potential maps (bottom) (B) Depiction of amino acid residues on the surface of PPP2R5 that are recognized by Vif or cellular substrates The LxxIxE substrate peptide is depicted bound to the PPP2R5 surface to highlight that Vif and cellular substrates compete for an overlapping surface these observations support a competitive binding model in which Vif directly interacts with the surface of PPP2R5 proteins and occludes the binding of cellular substrates The discovery of PPP2R5 substrates was a major step forward in understanding Vif-induced cell cycle arrest the regulatory checkpoints altered downstream of PPP2R5 degradation remain unknown PP2A/PPP2R5 complexes have been shown to regulate entrance and exit of the G2-to-M phase transition at multiple different checkpoints we discuss these checkpoints in the context of previous observations regarding Vif-induced G2/M arrest and postulate a model in which antagonism of discrete PP2A/PPP2R5 complexes would lead to simultaneous inhibition of multiple checkpoints Diagram of potential Vif-induced G2/M arrest mechanisms Depiction of normal and aberrant regulation of the G2-to-M phase transition in the absence (top) and presence (bottom) of Vif Key cell cycle checkpoints regulated by PP2A/PPP2R5 complexes are color-coded in the absence or presence of Vif In addition to manipulation of the PP2A/PPP2R5 axis, it has been suggested that Vif can induce G2/M arrest by blocking MDM2-mediated ubiquitination and nuclear export of TP53 (Izumi et al., 2010) This study observed that Vif binding to TP53 could block MDM2 recognition and subsequent turnover of TP53 which is required for proper cell cycle progression it is likely that the Vif/TP53 interaction occurs in the nuclear compartment thus directly shielding TP53 from MDM2 recognition and subsequent nuclear export it is plausible that there are two pools of Vif that act in concert to stall cell cycle progression Nuclear localized Vif protects TP53 from MDM2 and antagonizes nuclear PPP2R5C/D substrates whereas cytoplasmic Vif antagonizes the APOBEC3s and cytoplasmic PPP2R5A/B/E Additional work will be required to determine if these mechanisms are separate or connected through shared components Vif clearly has two distinct sets of cellular substrates APOBEC3 enzymes and PPP2R5 phospho-regulators (10 proteins total) While APOBEC3 counteraction has been shown to be essential for viral infectivity and pathogenesis in vivo the importance of PPP2R5 degradation remains to be established at least three key observations have been made that imply that alteration of the cell cycle may be beneficial for HIV-1 pathogenesis it is possible that Vif antagonism of PP2A holoenzymes leads to increased protein translation during G2/M when host cell translation is normally stalled it is possible that Vif and Vpr act in concert to induce G2/M arrest and boost transcription and translation of HIV-1 genes these observations support a model in which PPP2R5 antagonism and global changes in the cellular phospho-proteome are likely to be advantageous for the pathogenesis of HIV-1 as well as other prominent viruses continuing to unravel the complex molecular mechanisms HIV-1 has evolved to subvert cellular processes and enhance pathogenicity may provide major clues for the development of innovative therapeutics that lead to virus eradication Both authors contributed to the article and approved the submitted version This work was supported by NIAID R37-AI064046 (to RH) DS received salary support from an NIAID K99/R00 transition award (K99-AI147811) RH is the Margaret Harvey Schering Land Grant Chair for Cancer Research a Distinguished University McKnight Professor and an Investigator of the Howard Hughes Medical Institute and consultant of ApoGen Biotechnologies Inc The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest HIV-1 Vpr: mechanisms of G2 arrest and apoptosis Barré-Sinoussi Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS) Human immunodeficiency virus type 1 cell cycle control: Vpr is cytostatic and mediates G2 accumulation by a mechanism which differs from DNA damage checkpoint control The SOCS-box of HIV-1 Vif interacts with ELONGINBC by induced-folding to recruit its Cul5-containing ubiquitin ligase complex Google Scholar Cytidine deamination of retroviral DNA by diverse APOBEC proteins NMR structure of human restriction factor APOBEC3A reveals substrate binding and enzyme specificity Determinants of sequence-specificity within human AID and APOBEC3G The conserved KMN network constitutes the core microtubule-binding site of the kinetochore Genomic structure of an attenuated quasi species of HIV-1 from a blood transfusion donor and recipients Kinetochore microtubule dynamics and attachment stability are regulated by Hec1 Molecular mechanisms of antibody somatic hypermutation doi: 10.1146/annurev.biochem.76.061705.090740 Cell cycle perturbations induced by infection with the coronavirus infectious bronchitis virus and their effect on virus replication Vif-CBFβ interaction is essential for Vif-induced cell cycle arrest Google Scholar HIV-1 regulatory/accessory genes: keys to unraveling viral and host cell biology HIV-1 Vif’s capacity to manipulate the cell cycle is species specific Breaking bad: how viruses subvert the cell cycle PubMed Abstract | CrossRef Full Text | Google Scholar Roles of aurora kinases in mitosis and tumorigenesis Further investigation of simian immunodeficiency virus Vif function in human cells Frequent detection and isolation of cytopathic retroviruses (HTLV-III) from patients with AIDS and at risk for AIDS Phosphatase control of 4E-BP1 phosphorylation state is central for glycolytic regulation of retinal protein synthesis The aurora kinases in cell cycle and leukemia PubMed Abstract | CrossRef Full Text | Google Scholar Temporal proteomic analysis of HIV infection reveals remodelling of the host phosphoproteome by lentiviral Vif variants Promiscuous targeting of cellular proteins by Vpr drives systems-level proteomic remodeling in HIV-1 infection Structural basis for hijacking CBF-β and CUL5 E3 ligase complex by HIV-1 Vif PP2A regulatory subunit PP2A-B′ counteracts S6K phosphorylation Human T-cell leukemia virus-I tax oncoprotein functionally targets a subnuclear complex involved in cellular DNA damage-response Evolutionary paradigms from ancient and ongoing conflicts between the lentiviral Vif Protein and mammalian APOBEC3 enzymes RNA editing enzyme APOBEC1 and some of its homologs can act as DNA mutators CrossRef Full Text | Google Scholar The Vpr protein of human immunodeficiency virus type 1 influences nuclear localization of viral nucleic acids in nondividing host cells Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection Functional interaction of Aurora-A and PP2A during mitosis Lentiviral Vpr usurps Cul4-DDB1[VprBP] E3 ubiquitin ligase to modulate cell cycle HIV-1 and HIV-2 exhibit divergent interactions with HLTF and UNG2 DNA repair proteins subcellular localization and mC selectivity of a genomic mutator and Anti-HIV factor APOBEC3H Vif hijacks CBF-β to degrade APOBEC3G and promote HIV-1 infection Characterization of HIV-1 vpr nuclear import: analysis of signals and pathways PP2A function toward mitotic kinases and substrates during the cell cycle The Elongin BC complex interacts with the conserved SOCS-box motif present in members of the SOCS Selective tropism of lymphadenopathy associated virus (LAV) for helper-inducer T lymphocytes Local sequence targeting in the AID/APOBEC family differentially impacts retroviral restriction and antibody diversification Translational regulation of the cell cycle: when The ebola virus nucleoprotein recruits the host PP2A-B56 phosphatase to activate transcriptional support activity of VP30 Direct binding between BubR1 and B56-PP2A phosphatase complexes regulate mitotic progression Premature activation of the SLX4 complex by Vpr promotes G2/M arrest and escape from innate immune sensing An inhibitor of p34cdc2/cyclin B that regulates the G2/M transition in Xenopus extracts First-in-class small molecule inhibitors of the single-strand DNA cytosine deaminase APOBEC3G Ubiquitin-mediated degradation of aurora kinases PubMed Abstract | CrossRef Full Text | Google Scholar B′-protein phosphatase 2A is a functional binding partner of delta-retroviral integrase HIV-1 accessory proteins–ensuring viral survival in a hostile environment Role for the PP2A/B56delta phosphatase in regulating 14-3-3 release from Cdc25 to control mitosis CD4 counts as predictors of opportunistic pneumonias in human immunodeficiency virus (HIV) infection Phosphatases in mitosis: roles and regulation PubMed Abstract | CrossRef Full Text | Google Scholar Functional proteomic atlas of HIV infection in primary human CD4+ T cells PubMed Abstract | CrossRef Full Text | Google Scholar Mitotic repression of RNA polymerase II transcription is accompanied by release of transcription elongation complexes coli suggesting a DNA deamination mechanism for antibody diversification and continuous production of cytopathic retroviruses (HTLV-III) from patients with AIDS and pre-AIDS The local dinucleotide preference of APOBEC3G can be altered from 5′-CC to 5′-TC by a single amino acid substitution HIV-1 Vpr protein enhances proteasomal degradation of MCM10 DNA replication factor through the Cul4-DDB1[VprBP] E3 Ubiquitin ligase to induce G2/M cell cycle arrest Toxicity of human adenovirus E4orf4 protein in Saccharomyces cerevisiae results from interactions with the Cdc55 regulatory B subunit of PP2A HIV-1 Nef promotes infection by excluding SERINC5 from virion incorporation Human immunodeficiency virus type 1 Vif causes dysfunction of Cdk1 and CyclinB1: implications for cell cycle arrest Functional and structural insights into a Vif/PPP2R5 complex elucidated using patient HIV-1 isolates and computational modeling Google Scholar HIV-1 Vif triggers cell cycle arrest by degrading cellular PPP2R5 Phospho-regulators Nef-mediated suppression of T cell activation was lost in a lentiviral lineage that gave rise to HIV-1 The antiviral and cancer genomic DNA deaminase apobec3h is regulated by an RNA-mediated dimerization mechanism HIV-1 Nef antagonizes SERINC5 restriction by downregulation of SERINC5 via the Endosome/Lysosome system Structural basis for targeted DNA cytosine deamination and mutagenesis by APOBEC3A and APOBEC3B The HIV ‘A’ (sor) gene product is essential for virus infectivity PubMed Abstract | CrossRef Full Text | Google Scholar Regulation of mRNA translation during mitosis PP2A is required for centromeric localization of Sgo1 and proper chromosome segregation Cell cycle arrest in G2 induces human immunodeficiency virus type 1 transcriptional activation through histone acetylation and recruitment of CBP and c-Jun to the long terminal repeat promoter Protein phosphatase 2A in the regulation of wnt signaling Cdc25 phosphatases are required for timely assembly of CDK1-cyclin B at the G2/M transition SERINC3 and SERINC5 restrict HIV-1 infectivity and are counteracted by Nef The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein The role of RNA in HIV-1 Vif-mediated degradation of APOBEC3H Expanding the PP2A interactome by defining a B56-Specific SLiM A dynamic charge-charge interaction modulates PP2A:B56 substrate recruitment Dissection of the HIV Vif interaction with human E3 ubiquitin ligase The DDB1-DCAF1-Vpr-UNG2 crystal structure reveals how HIV-1 Vpr steers human UNG2 toward destruction Structure and function of the PP2A-shugoshin interaction Selective assembly of HIV-1 Vif-Cul5-ElonginB-ElonginC E3 ubiquitin ligase complex through a novel SOCS box and upstream cysteines HIV-1 Vpr induces cell cycle arrest and enhances viral gene expression by depleting CCDC137 T-cell differentiation factor CBF-β regulates HIV-1 Vif-mediated evasion of host restriction Conserved and non-conserved features of HIV-1 and SIVagm Vif mediated suppression of APOBEC3 cytidine deaminases Viral infections and cell cycle G2/M regulation PubMed Abstract | CrossRef Full Text | Google Scholar Reduced APOBEC3H variant anti-viral activities are associated with altered RNA binding activities Characterization of the interaction of full-length HIV-1 Vif protein with its key regulator CBFβ and CRL5 E3 ubiquitin ligase components Dispersed and conserved hydrophobic residues of HIV-1 Vif are essential for CBFβ recruitment and A3G suppression Citation: Salamango DJ and Harris RS (2021) Dual Functionality of HIV-1 Vif in APOBEC3 Counteraction and Cell Cycle Arrest Copyright © 2021 Salamango and Harris. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) distribution or reproduction in other forums is permitted provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited in accordance with accepted academic practice distribution or reproduction is permitted which does not comply with these terms *Correspondence: Reuben S. Harris, cnNoQHVtbi5lZHU=; Daniel J. Salamango, ZGFuaWVsLnNhbGFtYW5nb0BzdG9ueWJyb29rLmVkdQ== †Present address: Daniel J. Salamango, Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher. 94% of researchers rate our articles as excellent or goodLearn more about the work of our research integrity team to safeguard the quality of each article we publish. held discussions with members of Vivekananda International Foundation (VIF) a centre of excellence for innovative ideas aimed at propelling India to the Global Stage The Minister of Foreign Affairs delivered a speech on the topic "Towards the Asian Century of Dhamma," highlighting the roles of Thailand and India in promoting Buddhism and its teachings worldwide to foster peace and sustainability incollaboration with Bodhigaya Vijjalaya 980 Institute will organise the 4th Samvad (Global Hindu-Buddhist Initiatives) international conference in Thailand in early 2025 This website had been designed to be as accessible as possible to all and is certified by the WCAG 2.0 standard (Level AA) ** Best viewed with Chrome Version 76 up ** Metrics details HIV-1 is characterized by high genetic heterogeneity which is a challenge for developing therapeutics it is necessary to understand the extent of genetic variations that HIV is undergoing in North India The objective of this study was to determine the role of genetic and functional role of Vif on APOBEC3G degradation Vif is an accessory protein involved in counteracting APOBEC3/F proteins Genetic analysis of Vif variants revealed that Vif C variants were closely related to South African Vif C whereas Vif B variants and Vif B/C showed distinct geographic locations This is the first report to show the emergence of Vif B/C in our population motifs and phosphorylation sites were well conserved Vif C variants differed in APOBEC3G degradation from Vif B variants Vif B/C revealed similar levels of APOBEC3G degradation to Vif C confirming the presence of genetic determinants in C-terminal region High genetic diversity was observed in Vif variants which may cause the emergence of more complex and divergent strains These results reveal the genetic determinants of Vif in mediating APOBEC3G degradation and highlight the genetic information for the development of anti-viral drugs against HIV Importance: Vif is an accessory HIV-1 protein which plays significant role in the degradation of human DNA-editing factor APOBEC3G thereby impeding the antiretroviral activity of APOBEC3G It is known that certain natural polymorphisms in Vif could degrade APOBEC3G relatively higher rate This is the first report from North India showcasing genetic variations and novel polymorphisms in Vif gene but for the first time we observed putative B/C recombinants with a little high ability to degrade APOBEC3G indicating adaptation and evolving nature of virus in our population Indian Vif C variants were able to degrade APOBEC3G well in comparison to Vif B variants These genetic changes were most likely selected during adaptation of HIV to our population These results elucidate that the genetic determinants of Vif and highlights the potential targets for therapeutics there is no genetic information available for Vif gene which warrant for genetic study of Vif in our population we genetically characterized Vif gene from HIV-1 infected individuals of North India This study revealed the co-circulation of Vif B and Vif C subtypes which might create a condition conducive for the generation of Vif B/C recombinants The possible sequence similarity of our Vif variants with the global Vif variants were analysed by constructing phylogenetic tree Various natural substitutions were observed which may have conferred high pathogenicity advantage to virus Vif C variants resulted in little high APOBEC3G degradation in comparison to Vif B variants whereas Vif B/C recombinants were similar to Vif C in causing APOBEC3G degradation This study discusses the influence of genetic variations of Vif on APOBEC3G degradation (A) Phylogenetic tree of unique Vif variants with M (A to K including A1 (B) Phylogenetic tree of unique Vif variants with global subtype Vif B and Vif C reference sequences Each reference sequence was labelled with subtype followed by the country of isolation and accession number filled circles represent C variants and filled rectangles represent B/C recombinants 1,000 replicates) was indicated with an asterisk (*) at the corresponding nodes of the tree and the scale bar represents the selection distance of 0.01 nucleotides per position in the sequence (A) VifD64 is a representative of Vif B variant. (B) VifS1 is a representative of Vif C variant. (C) VifA6 is a representative of Vif B/C recombinant. Bootscan analyses were performed using consensus B, C and D shown in blue, red and green coloured arrows respectively with variants. Multiple sequence alignment of Vif variants Multiple sequence alignment for the predicted amino acid sequences of unique Vif variants with consensus Vif C and Vif B amino acids identical to Vif C and Vif B are denoted by asterisk (*) Polymorphisms are denoted in alphabetical letters Predicted major functional domains are represented at the top of the sequences within the boxes R19N change was observed in F1 box of about 76%; S23R V98I and D99E changes were observed in RNA binding region of about 76% 43% and 43% respectively; R63K change was observed in FG box of about 76%; HCCH region showed A123T and H127R in 53% of variants; K158Q was observed in multimerization region in 53% of variants; SOCS region showed V166I and T167K in 43% and 36% of variants respectively; K176N and T180I were observed in G box in 36% of variants These regions were conserved but a change in R41G was found in 25% of variants G37D and H48N were observed in 75% of variants which may modulate APOBEC3G/F degradation The amino acid region (from 85 to 99 and 169 to192) is shown to mediate Vif-APOBEC3G binding; we observed S95N D99E and K176N changes in our population which might alter the viral infectivity Amino acid sequence pattern of Vif variants (A) Amino acid signature pattern of Vif variants with consensus Vif B (B) Amino acid signature pattern of Vif variants with consensus Vif C The X-axis represent the amino acid consensus sequence of Vif B and C with the functional domains viz Trp-rich (N-terminal Tryptophan rich region from 1–21aa) F1-box (Interaction with host APOBEC3F from 14–17aa) RNA (RNA binding from 1–71aa and 75–114aa) G-box (Interaction with host from 40–44aa and 170–192aa) FG-box (Interaction with host APOBEC3F and from 54–72aa) F2-box (Interaction with host APOBEC3F from 74–79aa) HCCH motif (zinc-binding domain from 108–139aa) BC motif (Interaction with host elongin BC complex from 144–153aa) SOCS-box (Interaction with host SOCS from 139–176aa) MT (Multimerization from 151–164aa) and the Y-axis represent amino acid frequency observed in variants Motifs and phosphorylation sites in Vif variants Vif variants show protein kinase C (PKC) sites (20–22aa bipartite nuclear localization signal profile (NLS) sites (22–36aa and 167–184aa) N-myristoylation (NM) sites (71–76aa and 82–87aa) cAMP and cGMP dependent protein kinase (AGPK) sites (92–95aa and 167–170aa) amidation (AMD) site (181–184aa) and N-glycosylation (NG) site (186–189aa) The phosphorylation sites (serine and threonine residues) were marked with asterisk (*) at the corresponding amino acid sites on the top of the consensus C sequence despite high genetic variations and B/C recombination (A) APOBEC3G protein expression induced by Vif variants HEK 293T cells were co-transfected with Vif variants and APOBEC3G-HA cells were lysed and run on SDS-PAGE and then APOBEC3G protein was checked by immmunobloting with anti-HA antibody The relative protein intensity of APOBEC3G degradation by Vif variants were measured by subtracting APOBEC3G-HA positive control Densitometry represents the APOBEC3G degradation by Vif variants The error bar represents the standard deviation of APOBEC3G degradation and the p-value < 0.05 represents statistical significance of relative intensity of APOBEC3G with the relative intensity of respective Vif (* denote significant difference) This study shows the genetic architecture of HIV-1 Vif comprised of B and C subtypes This co-circulation of subtypes within a specific population led to the emergence of novel B/C recombinants in the ORF of Vif among North Indians which was observed to be statistically significant (**p < 0.05) i.e the proportion of recombinants have increased marginally (~3 to 5%) in the last few years implying the stable emergences of recombinants indicating the evolving tendency of the virus in our country Viral genetic recombination analysis also confirmed the existence of B/C recombinants with precise breakpoint in Vif gene These recombinants may contribute for generation of more complex viruses in our population; therefore it is essential to understand the genetic makeup of circulating recombinants and their prevalence in North India which will help in designing effective therapeutics against various circulating strains Phylogenetic analysis of Vif C variants showed sequence similarity with South African C variants whereas Vif B variants showed sequence similarity with the subtype B from different geographic regions that have resulted from multiple introductions of HIV-1 strains from China and other countries viz probably due to itinerant travellers from different countries Our data elucidates the importance of phylogenetic and recombination analyses in determining the sequence similarity between our variants with the highly prevalent global strains of HIV-1 and contribute to the better understanding of virus with respect to natural selection and adaption of viral strains Polymorphism analysis showed certain conserved substitutions which may favour virus for its optimal activities in causing pathogenicity in host cells these substitutions may involve in down-regulating or enhancing the functional activities of Vif and might also aid in the generation of more complex and divergent strains targeting the natural substitutions using RNAi mechanism may help in modulating HIV-1 infection It is important to note that this study is an attempt to analyze Vif variants from ART naïve and ART receiving patients of North India in relation to the contribution of this trait to pathogenicity viral evolution and possible implications for ART regimen and vaccine design low genetic diversity and lesser recombinants were observed among HIV-1 infected individuals receiving ART who showed improved CD4 counts and decreased viral loads implying the importance of ART in restoring immunity Domain conservation analysis based on sequence profiles of 105 HIV-1 infected patients from North India revealed the conservation at various functional domains viz ElonginC and SOCS binding domains highlighting the target sites for designing Anti-viral drugs Motif sites analysis showed conservation at various motifs in variants; remarkably identified nine phosphorylation sites were conserved The dN/dS analysis suggests purifying selection implying genetic integrity of Vif to maintain the functional activity of Vif It is necessary to have a matrix of variants in HIV-1 affected population since it will have a predictive value in determining the type of Anti-viral drug targets Based on sequence analysis of Vif variants in a specific population it is possible to map cytotoxic T lymphocyte (CTL) epitopes and enhance immune response of HIV-1 infected individuals against virus To explore the relationship between Vif-mediated APOBEC3G degradation dual transfection with Vif variants and APOBEC3G was carried out in HEK 293T cells This assay clearly showed that Vif B variants (Vif D43 D48 and E48) degrades APOBEC3G whereas Vif B/C recombinants (VT3 and VT4) carrying the N terminal region of Vif B and the C terminal of Vif C resulted in a little high APOBEC3G degradation as compared to Vif B and similar levels to Vif C confirming the vital role of C-terminal region in inducing APOBEC3G degradation This differential potential of Vif variants was due to the genetic variations in Vif B This study on Vif from HIV-1 infected individuals (n = 105) revealed Indian Vif sequences (VifS1 These matrixes of variants can be used in a combination for the understanding of Vif-host relationship and to develop targets against HIV in our population This study deciphers that HIV-1 virus continue to evolve in the local population but current pace of evolution is not so alarming Whether the dynamics of variation in the evolving population of viruses will retain its current pace or will it be altered is difficult to predict Whether host immune response is adequate to contain the variations in the evolving virus and whether other factors (host immune response etc) are likely to influence it remains to be seen Coinfections such as Tuberculosis may have profound impact on the host immune response thereby compelling the virus to evolve new strategies to synergize with Mycobacterium tuberculosis in the genetic and functional characteristics of the virus which may be subtle or gross for finding a better-fit to the situation in the host encountered by the virus periodic analysis of the virus for selection pressure-dictated variants becomes necessary to determine the trend of viral evolution if the variants are likely to pose a public health problem if changes are necessary in the modality of ART if newer viral targets can be obtained for prophylactic or therapeutic intervention and finally if the biology of the virus is underpinning a new element in basic sciences Our data indicates the genetic and functional role of Vif in causing APOBEC3G degradation targeting Vif by utilizing novel RNAi technology will help in perturbing the viral infectivity This study revealed various natural genetic determinants of Vif in mediating the functional activity of Vif in our population thus this study will immensely help in designing the potential targets based on Vif-APOBEC3G relationship against HIV/AIDS our data demonstrates that HIV-1 virus is evolving relevant to adaptation among North Indians through its ability to generate advantageous substitutions and recombinants in Vif gene to enhance its specific functional activities in the host cells HIV-1 infected patients (n = 105) were collected from North India who were registered and monitored at the immunodeficiency clinics of Guru Teg Bahadur (GTB) hospital Delhi and Post Graduate Institute of Medical Education and Research (PGIMER) Chandigarh during the period from 2004 to 2010 Number of males (45%) and females (38%) were chosen for this study along with vertical transmission variants (17%) This study was approved by Research Project Advisory Committee Institutional Biosafety Committee and Institutional Ethical Committee from Human research of University College of Medical Sciences and Guru Teg Bahadur Hospital India and from Post Graduate Institute of Medical Education and Research These institutes are mentored by National AIDS Control Organization (NACO) Government of India that provides free of cost ART to HIV-1 seropositive patients under a structured HIV/AIDS Control Programme These ethics committees approved the written informed consent which obtained from HIV-1 infected patients and from guardians of HIV-1 infected children participants involved in this study The methods were carried out in “accordance” with the approved guidelines Total RNA was extracted from peripheral blood mononuclear cells (PBMCs) of HIV-1 infected individuals using Trizol reagent (Invitrogen) PBMCs were washed with PBS and 1 ml of Trizol was added to 50–100 mg cells 200 μl of chloroform was added and mixed by vortexing This was incubated for 5 minutes at 37 °C and centrifuged at 13,200 rpm for 15 minutes The upper aqueous phase was transferred to the fresh tube and 500 μl of isopropanol was added to precipitate RNA The mix was incubated at 37 °C for 10 minutes and centrifuged at 13,200 rpm for 10 minutes at 4 °C The supernatant was discarded and pellet was washed with 75% ethanol The pellet was air dried and dissolved in nuclease free water by heating at 55°–60 °C for 10 minutes This RNA was reverse transcribed to form complementary DNA (cDNA) using ImProm-IITM Transcription system (Promega) 1 μg of RNA was reverse transcribed into cDNA using 20 pmol/μl of random primers and incubated at 70 °C for 15 minutes and kept at 4 °C Reverse transcription mix containing 1X reaction buffer rRNasin RNase inhibitor and reverse transcriptase was added to it and incubated at 25 °C for 5 minutes 5 μl of cDNA product was used for amplification of Vif using the following primers: Forward primer: 5′-GCGGATCCATGGAAAACAGATGGCAGG-3′ Reverse primer: 5′-GCCTCGAGCTAGTGTCCATTCATTGTATGG-3′ PCR was carried out in a 15  μl reaction volume The reaction mixture contained 500 ng genomic DNA (2.0 μl) 0.25  μl of Takara Taq DNA polymerase and 8.88 μl of DNase/RNase free water PCR conditions for the above primer sets were as follow: Initial denaturation at 94 °C for 5 minutes (1 cycle) 30 cycles of denaturation at 94 °C for 30 seconds annealing at 65 °C for 45 seconds and extension at 72 °C for 40 seconds and a final extension at 72 °C for 5 minutes (1 cycle) PCR amplified products were analyzed on 1.5% agarose gel The gel purified PCR products were cloned in pGEM-T Easy vector system The ligation reaction was incubated at 4 °C for 10 hours and the ligation mix was then plated on LB ampicillin plates with E.coli DH5α strain as host The plates were then incubated overnight at 37 °C The positive clones were selected by picking a single colony and grown in 5 ml LB Broth with ampicillin antibiotic (100 μg/ml) and incubated overnight at 37 °C Plasmid DNA was isolated from the culture by QIAprep Spin Mini Kit The positive clones were screened by restriction digestion of plasmid DNA with EcoRI in a 10 μl reaction volume at 37 °C for 2 hours The digested products were analyzed on 1.5% agarose gel after electrophoresis and the amplified bands were screened for positive clones after restriction digestion Five positive clones from each individual were sequenced from LabIndia and SciGenom laboratories by dideoxy chain termination method The cloning and sequencing were carried out twice for the positive clones to avoid PCR generated errors A known HIV-1 NL4-3 sequence was included for PCR amplification as a control to assess errors generated by Takara Taq polymerase The reliability of node was tested using the bootstrap method with 1000 replicates The phylogenetic trees were constructed for Vif representative variants (12 Vif B variants 3 Vif C variants and 3 Vif B/C recombinants) from North India and reference sequences which were retrieved from HIV Sequence database that includes subtype B and C sequences obtained from different parts of the world include America The basic criterion for choosing these reference sequences is that to cover all regions in worldwide for analysis in order to determine the sequence similarity with global reference sequences The nucleotide sequences of Vif were translated into amino acid sequences by GeneRunner The multiple sequence alignments were made for all variants with consensus B and C using ClustalW 2.1 It is already well known that subtype C is highly prevalent in India followed by subtype B and choosing consensus B and C sequences for analysis purpose is the idea behind the subtype prevalence in India and were retrieved from HIV Sequence database Novel polymorphisms were identified and their allele frequencies were calculated from total variants (n = 105) The amino acid sequence conservation was determined from the multiple sequence alignments of variants with the corresponding consensus B and C sequences using sequence alignment tool in MEGA5 software HEK-293T (Human Embryonic Kidney 293 cells; NIH AIDS Reagent Programme) were maintained in Dulbecco’s modified Eagle’s medium (DMEM) with 10% fetal bovine serum and 100 units penicillin 0.1 mg streptomycin and 0.25 μg amphotericin B per ml at 37 °C in the presence of 5% CO2 All transfections were performed using Lipofectamine 2000 (Invitrogen) reagent anti- HA tag polyclonal antibody (Clontech) anti-GAPDH antibody (Cell Signalling Technology) anti-vif monoclonal antibody (NIH AIDS Reagent Programme) anti-Rabbit IgG conjugated to HRP (Jackson Immunoresearch) anti-Mouse IgG conjugated to HRP (Jackson Immunoresearch) were used in the experiments Evidence for HTLV-III infection in prostitutes in Tamil Nadu (India) Determination of the rate of base-pair substitution and insertion mutations in retrovirus replication High rate of recombination throughout the human immunodeficiency virus type 1 genome Adaptive evolution of human immunodeficiency virus-type 1 during the natural course of infection A new human immunodeficiency virus derived from gorillas HIV Databases. &lt;www.hiv.lanl.gov/content/sequence/HIV/CRFs/CRFs.html&gt; Human immunodeficiency virus type 1 subtype distribution in the worldwide epidemic: pathogenetic and therapeutic implications Dynamic correlation between intrahost HIV-1 quasispecies evolution and disease progression APOBEC3G encapsidation into HIV-1 virions: which RNA is it Natural variation in Vif: differential impact on APOBEC3G/3F and a potential role in HIV-1 diversification Human immunodeficiency virus superinfection and recombination: current state of knowledge and potential clinical consequences Clinical infectious diseases: an official publication of the Infectious Diseases Society of America 34 Recombination increases human immunodeficiency virus fitness The role of recombination in the emergence of a complex and dynamic HIV epidemic Genetic characterization of natural variants of Vpu from HIV-1 infected individuals from Northern India and their impact on virus release and cell death Tat-Vpr interaction and cell apoptosis by natural variants of HIV-1 Tat exon 1 and Vpr from Northern India Genetic architecture of HIV-1 genes circulating in north India & their functional implications The Indian journal of medical research 134 Molecular and genetic characterization of natural HIV-1 Tat Exon-1 variants from North India and their functional implications A web-based genotyping resource for viral sequences Differential requirement for conserved tryptophans in human immunodeficiency virus type 1 Vif for the selective suppression of APOBEC3G and APOBEC3F Human immunodeficiency virus type 1 Vif protein is an integral component of an mRNP complex of viral RNA and could be involved in the viral RNA folding and packaging process Advances in the structural understanding of Vif proteins Nuclear localization of HIV type 1 Vif isolated from a long-term asymptomatic individual and potential role in virus attenuation Assembly of HIV-1 Vif-Cul5 E3 ubiquitin ligase through a novel zinc-binding domain-stabilized hydrophobic interface in Vif The SOCS box of suppressor of cytokine signaling-1 is important for inhibition of cytokine action in vivo Proceedings of the National Academy of Sciences of the United States of America 98 Potent suppression of viral infectivity by the peptides that inhibit multimerization of human immunodeficiency virus type 1 (HIV-1) Vif proteins HIV-1 infection requires a functional integrase NLS C-terminal half of HIV-1 Vif C possesses major determinant for APOBEC3G degradation Ubiquitination of APOBEC3G by an HIV-1 Vif-Cullin5-Elongin B-Elongin C complex is essential for Vif function High replication fitness and transmission efficiency of HIV-1 subtype C from India: Implications for subtype C predominance Proceedings of the National Academy of Sciences of the United States of America 106 CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting position-specific gap penalties and weight matrix choice Prospects for inferring very large phylogenies by using the neighbor-joining method Proceedings of the National Academy of Sciences of the United States of America 101 HIV Database Tools. &lt;http://www.hiv.lanl.gov/content/sequence/SNAP/SNAP.html&gt; Disease relevance of T11TS-induced T-cell signal transduction through the CD2-mediated calcineurin-NFAT pathway: Perspectives in glioma immunotherapy Mechanism of apoptotic induction in human breast cancer cell by an analog of curcumin in comparison with curcumin--an in vitro and in silico approach angiogenesis and metastasis in breast cancer cells via NF-kappaB pathway-A comparative study with curcumin Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 74 Apoptosis induction by an analog of curcumin (BDMC-A) in human laryngeal carcinoma cells through intrinsic and extrinsic pathways Download references India for helping in the collection of HIV-1 infected blood samples Source of Funding: This study was supported by Department of Biotechnology (BT/PR10599/Med/29/76/2008) and Indian Council of Medical Research (HIV/50/142/9/2011-ECD-II) Present address: Division of Infectious Diseases University College of Medical Sciences & Guru Teg Bahadur Hospital Vaishali Panwar & Vishnampettai G Ramachandran Department of Biochemistry and Molecular Biology Kumaravel Mohankumar & Subhashree Sridharan Jawaharlal Institute of Postgraduate Medical Education and Research Conceived and designed the experiments: L.R. Contributed reagents/materials/analysis tools: L.R. Download citation CHAPEL HILL, NC (PRWEB) January 10, 2017 -- VIF International Education, an education partner of K-12 schools and districts, announced today that it is changing its name to Participate The change reflects the organization’s broader commitment to educator development and its expertise in providing continuous learning opportunities that empower teachers and inspire students to be active contributors to their communities and our world our growing national and international work now includes instructional design program management and education technology In May of 2016, VIF International Education acquired Participate Learning a leading online platform for educational resource discovery and collaboration The acquisition positioned the organization to deliver best-in-class technology to enhance and expand professional learning and collaboration opportunities for educators It represented what became the preeminent U.S cultural exchange program for teachers and afforded us the opportunity to pursue other pressing education needs,” said Participate CEO David Young we believe Participate more accurately reflects these broader education pursuits and the enhanced technology capabilities that will help us reach more students and teachers than we ever thought possible - it recognizes our past present and future in support of collaborative experiential and global learning for all.” Visiting International Faculty (VIF) was founded in 1987 as a family business that sought to promote the value of international perspectives in education by providing universities with international faculty recruitment The organization shifted focus to K-12 education in 1989 when state departments of education began pursuing world language learning for K-12 students VIF has placed thousands of international teachers in K-12 classrooms across the United States as a designated U.S Department of State Exchange Visitor Program sponsor The organization continues to be committed to cultural exchange and supports international teachers’ success in the classroom as it firmly believes that exposing students to intercultural experiences at the K-12 level is key to building global competence - a critical skill for success in our increasingly interconnected world the organization also provides schools and districts with leading-edge technology comprehensive frameworks and support services to impact student outcomes by improving teacher practice Many of its partners are distinguished by school-wide commitments to strengthen academic achievement for diverse student populations through immersive language learning opportunities and equitable access to global The new name is effective immediately and will be implemented across the organization's products and services throughout the remainder of 2017 About Participate Participate (http://www.participate.com) partners with schools and districts to provide leading-edge technology comprehensive frameworks and support services These programs and services impact student outcomes by improving teacher practice through collaborative professional learning educators have used Participate’s professional development and curriculum language acquisition and cultural exchange teacher programs to create engaging learning environments that empower teachers and inspire students to create impact on a global scale Participate is a certified B Corp and ‘Best for the World’ honoree headquartered in Chapel Hill TJ Scholl, Participate, https://www.participate.com/, 919-265-5065, [email protected] Do not sell or share my personal information: AWS Direct Connect (DX) recently launched support for virtual interface (VIF) metrics in Amazon CloudWatch CloudWatch can now track metrics at the DX VIF level and provide greater insight into utilization You can set up alarms based on metrics and trigger actions to remediate problems I’ve heard from many customers that they wanted greater visibility into traffic utilization when using multiple VIFs on the same connection – dedicated or hosted I’m excited about this release as there is now a solution I dig into this new functionality and how it compares to the prior capability Before this launch it was possible to see aggregated metrics at the Direct Connect connection it was not possible to view individual VIFs and determine throughput utilization a 10-Gbps DX connection has a transit VIF for AWS Transit Gateway connectivity a public VIF for connectivity to public AWS resources and a private VIF for connectivity to a VMware environment Looking at the screenshot you can see that there was a bit of a spike in throughput there is not an easy way to drill down and determine which VIF is the source of this traffic Now that that there are VIF level metrics we see a new set of metrics in the CloudWatch console I can now dig in further to see which VIF is responsible for the traffic Here I can see that there are metrics for the VIF that resides within my DX account you can see that the peak here is only around 8-Mbps It turns out that the other VIFs are hosted VIFs and actually reside in a different account than the DX connection Fortunately it is quite simple to share CloudWatch metrics across accounts CloudWatch metrics can be viewed cross-region and cross-account I can see a drop-down in the console to view data from the shared account I can select an account and view its metrics I can graph these metrics alongside the VIF that exists within the DX account Note that there is a fee if anomaly detection is enabled, so be sure to check the CloudWatch pricing page I enabled this on a VIF metric for BpsEgress (bytes per second egress) as shown in the following image Once anomaly detection is enabled I can see that traffic spikes are outside the ordinary baseline for traffic I also can see the expected rate of traffic In this post I showed how VIF CloudWatch metrics can be used and shared between accounts. For a full list of CloudWatch metrics supported by DX you can view the documentation With this new capability even greater visibility into DX utilization is now possible Volume 13 - 2022 | https://doi.org/10.3389/fmicb.2022.828430 Human immunodeficiency virus type 1 (HIV-1) has RNA genome and depends on host cellular machinery for most of its activities Host cellular proteins modulate the expression and activity of viral proteins to combat the virus HIV-1 proteins are known to regulate each other for the benefit of virus by exploiting these modulations we report that HIV-1 Vif increases the levels of Tat via AKT signaling pathway We show that HIV-1 Vif activates AKT signaling pathway by inducing phosphorylation of AKT increases the levels of Tat protein in ubiquitin-dependent manner by inducing Ubiquitin Specific Protease 17 (USP17) which is a deubiquitinase and stabilizes Tat protein HIV-1 proteins exploit AKT signaling pathway to promote viral replication so we investigated the effect of Vif on the expression of Tat to find out how HIV-1 is benefited by activating AKT signaling pathway via its two proteins We found that Vif increased the levels of Tat protein Vif was also found to increase the LTR transcription mediated by Tat protein Inhibition of AKT phosphorylation abrogated Vif-mediated increase in levels of Tat protein Mdm2 (target of AKT) was found to increase the levels of Tat via a deubiquitinase AKT signaling pathway was playing an important role in the regulation of HIV-1 Tat by Vif via Mdm2 mediated stabilization of USP17 This study can have significant implications toward better understanding of the several mechanisms of HIV-1–mediated exploitation of host machinery and viral pathogenesis Human embryonic kidney 293T (HEK-293T) and Tzm-Bl cells were maintained in Dulbecco modified eagle medium (Himedia Laboratories India) supplemented with 10% fetal bovine serum (Gibco and 0.25 μg amphotericin B per ml at 37°C in the presence of 5% CO2 in a humidified incubator and U1 cells were maintained in RPMI-1640 supplemented with 10% fetal bovine serum (Gibco and 0.25 μg amphotericin B (Himedia Laboratories India) per milliliter at 37°C in the presence of 5% CO2 in a humidified incubator Transfections were performed using Lipofectamine 2000 (Invitrogen United States) reagents using the manufacturer’s protocol Plasmid Myc Vif was made by cloning pNL4-3–derived gene vif in pCMV-Myc plasmid from Clontech, United States, as described earlier (Arora et al., 2014) pBlue3′LTR-luc was obtained from NIH AIDS Reference and Reagent Program of NIH Glutathione S-transferase (GST) Tat was generated by cloning pNL4-3 derived tat gene in pGEX-4T1 vector from Addgene HA Tat and Flag Tat were purchased from Addgene HA Mdm2 was purchased from Sino Biologicals and HA Myr AKT were kind gifts from Hui Kuan Lin Renilla luciferase plasmid was a kind gift from Vivek Natrajan His Ub plasmid was gifted by Dimitris Xirodimas Human embryonic kidney 293T cells were transfected with gene of interest for 24 h The cells were harvested and lysed in RIPA lysis buffer (1% NP-40 Protein estimation was carried out using BCA Protein Assay Kit (Pierce An equal amount of protein was loaded on sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and was transferred to nitrocellulose membrane The membranes were blocked with 5% non-fat dry milk (Himedia Laboratories anti–phospho-AKT (S473) (Cell Signaling Technology) The secondary antibodies used were anti-rabbit/mouse–horseradish peroxidase–conjugated (Jackson ImmunoResearch) Blots were developed using ECL (enhanced chemiluminescence) reagent To study the degradation kinetics of proteins HEK-293T cells were transfected with gene of interest for 24 h and treated with CHX (100 μg/mL; Sigma) Cell lysates were prepared at indicated time points and subjected to 10% SDS-PAGE followed by Western blot analysis as described above In vivo ubiquitination assay was performed to detect ubiquitylated proteins in transfected mammalian cells HEK-293T cells were cotransfected with plasmid encoding desired gene and His-Ub (6 × histidine–ubiquitin) for 24 h 20 μM MG132 (Sigma-Aldrich) was added and the cells were further incubated for 8 h The cells were lysed in buffer A (6 M guanidinium-HCl Ni-NTA beads were added to the supernatant and the mixture was incubated at room temperature for 6 h while rotating the beads were washed with buffer A and buffer TI (25 mM Tris The ubiquitinated proteins were eluted in buffer containing 200 mM imidazole The eluates were resolved by SDS-PAGE followed by Western blot analysis pGEX-4T1 vector containing the desired gene was transformed in BL21 strain of Escherichia coli for expression and subsequent purification The bacterial culture was induced with 0.5 mM IPTG at 16°C for 16 h The cells were lysed by adding lysozyme (1 mg/mL) at 4°C with gentle shaking DTT was added to the bacterial lysate after lysozyme treatment (100 μL of 1 M DTT) This was followed by sonication and extraction of proteins with Triton X-100 The solution was centrifuged at 12,000 revolutions/min (rpm) for 15 min at 4°C The supernatant was then used for binding to glutathione beads at 4°C for 3 h The beads were centrifuged at 2,500 rpm for 2 min at 4°C The beads were washed until the supernatant stopped giving color with Bradford reagent GST alone and GST-tagged proteins were expressed and purified as described previously HEK-293T cells were transfected with gene of interest for 36 h Ten micrograms of GST-tagged protein was incubated with the cell lysate at 4°C for 4 h and the beads were washed five to six times with chilled 1 × phosphate-buffered saline (PBS) The beads were boiled in Laemmli buffer and subjected to SDS-PAGE followed by immunoblotting with anti-Myc and anti-GST antibodies The protein–protein interaction was studied by coimmunoprecipitation The genes of interest were cotransfected in HEK-293T cells for 24 h For affinity tag-based immunoprecipitation cell lysates were prepared in CelLytic M and cell lysis reagent (Sigma) and were incubated with anti-Myc agarose beads (Sigma) at 4°C overnight The beads were washed with IP buffer (Sigma) The purified protein complex bound to anti-Myc agarose beads was resolved by SDS-PAGE and subjected to Western blot analysis Pierce™ Direct IP kit (Thermo Scientific) was used to pull down a protein without tag HEK-293T cells were transfected with desired plasmids and antibody-conjugated agarose resin was added resin was pelleted and washed with wash buffer The immunoprecipitated proteins were eluted using elution buffer The aqueous solution containing eluted proteins was boiled with SDS–PAGE loading buffer for 5 min and analyzed by Western blotting Luciferase reporter assay was performed using the dual-luciferase reporter assay kit (Promega HEK-293T cells were cotransfected with luciferase reporter plasmid and the plasmids encoding genes of interest Renilla luciferase was used as control to normalize the transfection efficiency Empty pcDNA3.1 vector was used to equalize the amount of DNA transfected in each well cell were harvested and lysed in lysis buffer (Promega Luciferase activity was measured by luminometer (Tecan Switzerland) using two substrates (Promega United States): one for firefly luciferase and another for Renilla luciferase (mixed with Stop and Glo buffer) The readings of firefly luciferase activity were normalized with those of Renilla luciferase activity to get the true luciferase reporter activity All the experiments were repeated three to four times Results obtained are presented as mean ± standard error of the mean (s.e.m) p-values were calculated by a two-tailed t-test Only values with p < 0.05 were considered significant Human immunodeficiency virus type 1 Vif induces phosphorylation of AKT at Ser473 (A) HEK-293T cells were cotransfected with HA AKT Cell lysates were analyzed by Western blotting with anti-HA (B) HEK-293T cells were transfected with increasing amounts of Myc Vif expression plasmid for 24 h Cell lysates were analyzed by Western blotting with anti-AKT (C) HEK-293T cells were transfected with Myc Vif expression plasmid and treated with AKTi (5 μM) as indicated for 24 h (D) HEK-293T cells were transfected with Myc Vif and treated with increasing dose of AKTi (3 and 5 μM) for 24 h (E) HEK-293T cells were treated with AKTi (5 μM) and transfected with increasing amount of Myc Vif (2 and 4 μg) for 24 h (F) HEK-293T cells were transfected with AKT siRNA and Myc Vif Densitometry analysis was performed using ImageJ and shown as bar graph with error bars Data obtained are presented as mean ± s.e.m p-value was calculated by two-tailed t-test [*p < 0.05 suggesting Vif-mediated increase in the levels of Tat protein Human immunodeficiency virus type 1 Vif increases the expression of Tat via AKT signaling pathway (A) HEK-293T cells were cotransfected with HA Tat and Myc Vif plasmids as indicated for 24 h Cell lysates were analyzed by Western blotting using anti-HA (B) HEK-293 T cells were transfected with HA Tat either alone or along with Myc Vif for 24 h Cells were treated with cycloheximide (100 μg/mL) for the indicated time periods Cell lysates were subjected to Western blot analysis using anti-HA Densitometric analysis was done by ImageJ and shown as line graph with error bars (C) HEK-293T cells were cotransfected with HA Tat and Myc Nef plasmids as indicated for 24 h (D) HEK-293T cells were cotransfected with HIV-1 LTR-luc reporter and Myc Vif expression plasmids as indicated for 24 h and dual luciferase reporter assay was performed using luminometer Relative luciferase activity is shown as bar graph (E) GST Tat and GST alone bound with GST beads were incubated with in vitro synthesized Vif for 2 h at 4°C Vif specific antiserum was used to probe the Vif protein in Western blot analysis (F) Tzm-Bl cells were transfected with HA Tat and Myc Vif plasmids for 24 h Cell lysates were subjected to immunoprecipitation using anti-HA agarose beads at 4°C overnight beads were washed with IP buffer and boiled with PAGE loading buffer followed by SDS-PAGE and Western blotting using anti-Myc antibody (G) HEK-293T cells were cotransfected with HA Tat and Myc Vif plasmids as indicated and treated with AKTi (5 μM) for 24 h (H) HIV-1 replication in U1 cells was induced by PMA (100 μg/mL) and cells were treated with AKTi (5 μM) for 24 h Cell lysates were analyzed by Western blotting using anti-p24 These results suggest intracellular interaction of Tat and Vif proteins we concluded that total ubiquitination of Tat is reduced in the presence of Mdm2 despite the fact that Mdm2 induces K63 ubiquitination of Tat This might be the reason that the difference between the ubiquitination levels of Tat in the presence or absence of Mdm2 is less than expected These results indicate that Mdm2 is the candidate protein of AKT signaling pathway which increases the levels of Tat in ubiquitin-dependent manner Mdm2 stabilizes the expression of HIV-1 Tat in ubiquitin-dependent manner (A) HEK-293T cells were transfected with HA Tat either alone or along with HA Mdm2 for 24 h Cell lysates were subjected to Western blot analysis using anti-HA and anti-GAPDH antibodies (B) HEK-293 T cells were transfected with HA Tat either alone or along with HA Mdm2 for 24 h (C) HEK-293T cells were transfected with Mdm2 siRNA and HA Tat for 48 h (D) HEK-293T cells were cotransfected with His Ub Cells were treated with MG132 (20 μM) for 8 h Cell lysates were subjected to immunoprecipitation with Ni-NTA beads followed by Western blotting with anti-Flag antibody p-value was calculated by two-tailed t-test (*p < 0.05 These results indicate that USP17 stabilizes HIV-1 Tat by inducing its deubiquitination Ubiquitin Specific Protease 17 stabilizes the expression of Tat by deubiquitination (A) HEK-293T cells were cotransfected with HA Tat and His USP17 plasmids for 24 h (B) HEK-293T cells were transfected with HA Tat either alone or along with His USP17 for 24 h (C) HEK-293T cells were cotransfected with His Ub Cell lysates were subjected to immunoprecipitation with Ni-NTA beads followed by Western blotting with anti-HA antibody (D) HEK-293T cells were transfected with HA Tat encoding plasmid cell lysates were subjected to immunoprecipitation using anti–USP17 antibody–bound agarose beads followed by Western blotting using anti-HA antibody (E) HEK-293T cells were cotransfected with HA Tat and pNL4.3 for 24 h Cell lysates were subjected to Western blotting with anti-HA Mdm2 stabilizes the expression of USP17 in ubiquitin-dependent manner (A) HEK-293T and Thp-1 cells were treated with AKTi as indicated for 24 h Cell lysates were subjected to Western blotting using anti-USP17 (B) HEK-293T cells were cotransfected with His USP17 and HA Mdm2 plasmids for 24 h Cell lysates were analyzed by Western blotting using anti-His (C) HEK-293T cells were transfected with His USP17 and increasing amounts of HA Mdm2 encoding plasmid cell lysates were subjected to Western blotting using anti-His and anti-GAPDH antibodies (D) HEK-293T cells were transfected with increasing amounts of HA Mdm2 plasmid for 24 h (E) HEK-293T cells were cotransfected with His Ub Cell lysates were subjected to immunoprecipitation with Ni-NTA beads followed by Western blotting with anti-USP17 antibody (F) HEK-293T cell lysates were subjected to immunoprecipitation using anti–Mdm2 antibody–bound agarose beads followed by Western blotting using anti-USP17 antibody (G) HEK-293 T cells were transfected with siRNA specific to Mdm2 cell lysates were subjected to Western blot analysis using anti-USP17 p-value was calculated by two-tailed t-test (**p < 0.01) These results indicate that Mdm2 increases the levels of USP17 in ubiquitin-dependent manner by interacting with it These results indicate that USP17 is induced by HIV-1 infection Human immunodeficiency virus type 1 Vif increases the expression of USP17 (A) HEK-293T cells were cotransfected with His USP17 and Myc Vif expression plasmids for 24 h Cell lysates were subjected to Western blot analysis using anti-His (B) HEK-293T cells were transfected with pNL4.3 plasmid for 24 h Cell lysates were analyzed by Western blotting with anti-USP17 (C) U1 and U937 cells were treated with PMA (100 μg/mL) for different time intervals as shown (D) HEK-293T cells were transfected with pNL4.3 or pNL4.3ΔVif for 24 h These results indicate that HIV-1–induced enhancement in USP17 protein levels is mediated by Vif Human immunodeficiency virus type 1 Vif induces proteasomal degradation of Mdm2 (A) HEK-293T cells were cotransfected with HA Mdm2 and Myc Vif expression plasmids for 24 h HEK-293T cells were transfected with Myc Vif encoding plasmid for 24 h Cell lysates were analyzed by Western blotting with anti-Mdm2 (B) HEK-293T cells were transfected with HA Mdm2 and Myc Vif expression plasmids for 24 h as indicated and treated with CHX (100 μg/mL) for indicated time periods and cell lysates were subjected to SDS-PAGE followed by Western blotting using anti-HA Densitometry analysis was performed using ImageJ and shown as line graph with error bars (C) HEK-293T cells were cotransfected with HA Mdm2 and Myc Vif encoding plasmids as shown for 24 h and cell lysates were subjected to SDS-PAGE followed by Western blotting with anti-HA (D) HEK-293T cells were transfected with Myc Vif expression plasmid and treated with AKTi (5 μM) for 24 h (E) HEK-293T cells were cotransfected with HA Mdm2 and Myc Vif encoding plasmids for 24 h (F) HEK-293T cells were cotransfected with His Ub These results indicate that Vif induces ubiquitin-mediated proteasomal degradation of Mdm2 the effect of Vif on the expression of Tat and the role of AKT signaling pathway in this process were investigated Vif was found to increase the levels of Tat protein and Tat-mediated LTR transactivation was also found to be enhanced in the presence of Vif HIV-1 Vif and Tat were also found to interact with each other The inhibition of AKT phosphorylation interfered with Vif-mediated increase in Tat levels was found to induce an increase in the levels of Tat protein When the mechanism of Mdm2-mediated stabilization of Tat was investigated it was observed that Mdm2 can up-regulate the levels of HIV-1 Tat protein in ubiquitin-dependent manner via USP17 which stabilizes Tat by deubiquitinating it HIV-1 also induced an increase in the levels of USP17 Vif-mediated increase in the levels of Tat protein might be independent of Tat-Vif interaction as it has been found to be mediated by AKT signaling pathway Vif induced proteasomal degradation of Mdm2 protein instead of increasing its expression which was expected from the inducing effect of Vif on phospho-AKT Ser 473 These results indicate the complex role of AKT signaling pathway in regulation of HIV-1 protein expression and viral replication we hypothesize that HIV-1 exploits AKT signaling pathway through its activation by Tat and Vif so as to maintain the sufficient levels of Mdm2 in the host cell to increase Tat levels and activity But when there is more increase in the levels of Mdm2 HIV-1 regulates the expression of Mdm2 via inducing its degradation by Vif so as to prevent Mdm2-mediated proteasomal degradation of Vif It helps the virus to evade the host restriction machinery more effectively and survive in the host cell Mechanistic model of HIV-1 Vif–mediated up-regulation of Tat via AKT signaling pathway HIV-1 Tat induces AKT signaling pathway and increases the phosphorylation of AKT and Mdm2 HIV-1 Vif increases the expression of Tat protein via AKT signaling pathway where Mdm2 acts as the mediator Mdm2 increases the expression of Tat by inducing an increase in the levels of USP17 which stabilizes the levels of Tat by deubiquitination HIV-1 Vif also induces proteasomal degradation of Mdm2 (blue arrows: previously known phenomena The original contributions presented in the study are included in the article/supplementary material further inquiries can be directed to the corresponding authors SL and VS designed and performed the experiments This work was supported by Department of Biotechnology and Department of Science and Technology of Government of India All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher Several reagents were obtained from AIDS Reference and Reagent Program of NIH We thank Hui Kuan Lin (MD Anderson Cancer Center India) for Renilla luciferase plasmid and Dimitris Xirodimas (University of Dundee) for His Ub plasmid USP7 deubiquitinase controls HIV-1 production by stabilizing Tat protein HIV-1 Vpr redirects host ubiquitination pathway Blagoveshchenskaya HIV-1 Nef downregulates MHC-I by a PACS-1- and PI3K-regulated ARF6 endocytic pathway Extracellular HIV-1 Tat protein activates PI3K and Akt kinase in CD4+ T lymphoblstoid jurkat clls A non-proteolytic role for ubiquitin in Tat-mediated transactivation of the HIV-1 promoter Downregulation of CD4 by human immunodeficiency virus type 1 Nef is dependent on clathrin and involves direct interaction of Nef with the AP2 clathrin adaptor The ubiquitin-specific protease 17 is involved in virus-triggered type I IFN signaling Akt inhibitors as an HIV-1 infected macrophage-specific anti-viral therapy Infection of human immunodeficiency virus and intracellular viral Tat protein exert a pro-survival effect in a human microglial cell line The pivotal role of phosphatidylinositol 3-kinase-Akt signal transduction in virus survival HIV-1-Tat protein activates phosphatidylinositol 3-kinase/AKT-dependent survival pathways in Kaposi’s sarcoma cells Vpu directs the degradation of the human immunodeficiency virus restriction factor BST-2/tetherin via a {beta}TrCP-dependent mechanism Stabilization of Mdm2 via decreased ubiquitination is mediated by protein kinase B/Akt-dependent phosphorylation PubMed Abstract | CrossRef Full Text | Google Scholar Ganser-Pornillos Restriction of HIV-1 and other retroviruses by TRIM5 PubMed Abstract | CrossRef Full Text | Google Scholar HIV-1 Rev downregulates Tat expression and viral replication via modulation of NAD(P)H:quinine oxidoreductase 1 (NQO1) Antiviral inhibition of enveloped virus release by tetherin/BST-2: action and counteraction HIV restriction factors and mechanisms of evasion PubMed Abstract | CrossRef Full Text | Google Scholar PubMed Abstract | CrossRef Full Text | Google Scholar HIV-1 Tat potently stabilises Mdm2 and enhances viral replication Google Scholar SAMHD1 restricts HIV-1 infection in dendritic cells (DCs) by dNTP depletion but its expression in DCs and primary CD4+ T-lymphocytes cannot be upregulated by interferons K63-linked ubiquitination in kinase activation and cancer The HIV-1 Vif protein mediates degradation of Vpr and reduces Vpr-induced cell cycle arrest Sood V and Banerjea AC (2022) Human Immunodeficiency Virus Type 1 Vif Up-Regulates the Expression of Tat via AKT Signaling Pathway: Role of Ubiquitin Specific Protease 17 Copyright © 2022 Lata, Sood and Banerjea. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) *Correspondence: Sneh Lata, c25laGxhdGF2aXJvQGdtYWlsLmNvbQ==; Akhil C. Banerjea, YWtoaWxAbmlpLmFjLmlu Metrics details The data from three independent experiments are summarized in the bar graph and represent the mean ± s.e.m (err bar) The value of controls is arbitrarily set as 100 (%) Asterisk (*) indicate P < 0.05 and double asterisks (**) indicate P < 0.01 relative to controls (the GraphPad Prism software) To evaluate the biological activity of the 26 hits, we assessed their effect on hA3G expression in the presence of HIV-1 Vif. 293 T cells were co-transfected with the expression vectors for hA3G-HA and Vif, and then treated with 10 μM of each compound. The results in Fig. 1B showed that four compounds restored hA3G expression in the presence of Vif compared with that in the cells treated with DMSO This suggests that the four compounds are able to inhibit the degradation of hA3G by HIV-1 Vif IMB-301 inhibits the replication of HIV-1 in the presence of hA3G H9 and SupT1 cells were infected with wild-type HIV-1(NL4-3) in the presence of IMB-293 (A) The infectivity of nascent viruses in the supernatant was measured by infecting the TZM-bl indicator cells (E) 293 T cells were co-transfected with pNL4-3Luc(R-E-) followed by the treatment with various concentration of IMB-301 The supernatants were collected and then used to infect the SupT1 cells (F) H9 or SupT1cells were incubated with various concentrations of IMB-301 or DMSO for 48 hours and cytotoxic effects were then measured using the CCK8 assays Equal volumes of DMSO were added into the culture medium which contain different concentrations of the compounds in order to keep constant final DMSO concentration as 1% (v/v) The value of controls is arbitrarily set as 1 (A–D) or 100% (E and F) IMB-301 specifically inhibitshA3G but not hA3F degradation. 293 T cells were co-transfected with Vif and eitherhA3G-HA (A) or hA3F-HA (B), followed by the treatment of IMB-301. Then cell lysates were analyzed by WB. Total amounts of DNA were maintained the same between transfections by supplementation of empty vector DNA. (A) Octet binding of IMB-301 to hA3G was performed as described in Materials and Methods (B) The predicted binding mode of IMB-301 in the binding site for the homology structure of hA3G-NTD (HM-hA3G-NTD) (C) The detailed interactions between IMB-301 and hA3G-NTD we have identified a small molecular inhibitor IMB-301 via virtual screening according to the hA3G model Further biochemical experiments have shown that IMB-301 binds to hA3G restores hA3G expression in the presence of Vif and inhibits the replication of HIV-1 in a hA3G-dependent manner Our results demonstrate the possibility of inhibiting HIV replication by abrogating the Vif-hA3G interaction with small molecules 293 T and TZM-bl cells were cultured in DMEM (GBICO) supplemented with 10% fetal bovine serum (FBS) (GBICO) SupT1 cells were maintained in RPMI-1640 (GBICO) containing 10% FBS H9 cells were maintained in RPMI-1640 (GBICO) containing 10% FBS Transfections of 293 T cells were performed using Lipofectamine 2000 (Invitrogen) according to the manual from the manufacturer the supernatants of virus-producing cells were pelleted through a 20% sucrose cushion at 35000 rpm for 60 min (Beckman) The harvested viral samples were analyzed by western blotting Western blots were probed with monoclonal antibodies against HIV-1 p24 (NIH) Detection of proteins was performed by enhanced chemiluminescence (Millipore) using secondary antibodies anti-mouse (for β-actin) and anti-rabbit (for P24 and HA) both were purchased from Santa Cruz Biotechnology Inc Bands in western blots were quantitated using ChemiDoc TMMP (Bio-Rad) automated digitizing system and Image J software To assess the effect of the four small compounds on HIV-1 infectivity the experiments were carried out in H9 and SupT1 cells H9 and SupT1 cells were infected with wild-type HIV-1(NL4-3) in the presence of the compounds at various concentrations The infectivity of nascent viruses in the supernatant was measured by infecting the TZM-bl indicator cell for 48 hours (hr) To assess the effect on the pseudotyped HIV-1 293 T cells were co-transfected with 300 ng pNL4-3Luc(R-E-) 200 ng VSVG and 200 ng hA3G-HA (or pcDNA3.1) plasmid DNA in 6-wellplates the media was changed and IMB-301 was added the supernatants were collected and filtered through a 0.45 µm filter and then used to infect the SupT1 cells (1 × 105) in 96-well plates SupT1 cells were lysed and firefly luciferase activities were determined using a firefly Luciferase Assay System (Promega) The cytotoxicity of IMB-301 was measured using the CCK8 Assay Kit (Beyotime) The kit provides an assay that distinguishes metabolically active cells from injured cells and dead cells SupT1 or H9 cells were treated with IMB-301 at various concentrations DMSO treated cells were used as the control the samples were subjected to Live/Dead Cell Vitality Assay Kit following the manufacturer manual the cells were co-transfected with 1 µg Vif and 3 µg hA3G-HA the cells were treated with DMSO or 10 µM IMB-301 for 24 hr the cells were collected and lysated in NP-40-containing buffer for 30 min on ice Cell lysates were centrifugated at 10,000x g for 10 min at 4 °C and supernatant was transferred to a fresh 1.5 ml tube on ice The cell lysates were incubated with anti-HA rabbit antibody for 3 hr at 4 °C Thirty microliters of proteinA-Agarose (Santa Cruz) was added and incubated overnight at 4 °C The beads were washed 4 times with lysis buffer (cold) and at last boiled in 40 µl sample buffer for 5 to 10 min The samples were analyzed by Western blotting Sheehy, A. M., Gaddis, N. C., Choi, J. D. & Malim, M. H. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature 418, 646–650, https://doi.org/10.1038/nature00939 (2002) Shindo, K. et al. The enzymatic activity of CEM15/Apobec-3G is essential for the regulation of the infectivity of HIV-1 virion but not a sole determinant of its antiviral activity. The Journal of biological chemistry 278, 44412–44416, https://doi.org/10.1074/jbc.C300376200 (2003) Hache, G., Liddament, M. T. & Harris, R. S. The retroviral hypermutation specificity of APOBEC3F and APOBEC3G is governed by the C-terminal DNA cytosine deaminase domain. The Journal of biological chemistry 280, 10920–10924, https://doi.org/10.1074/jbc.M500382200 (2005) Zhang, H. et al. The cytidine deaminase CEM15 induces hypermutation in newly synthesized HIV-1 DNA. Nature 424, 94–98, https://doi.org/10.1038/nature01707 (2003) Li, X. Y., Guo, F., Zhang, L., Kleiman, L. & Cen, S. APOBEC3G inhibits DNA strand transfer during HIV-1 reverse transcription. The Journal of biological chemistry 282, 32065–32074, https://doi.org/10.1074/jbc.M703423200 (2007) Mbisa, J. L. et al. Human immunodeficiency virus type 1 cDNAs produced in the presence of APOBEC3G exhibit defects in plus-strand DNA transfer and integration. Journal of virology 81, 7099–7110, https://doi.org/10.1128/jvi.00272-07 (2007) Sheehy, A. M., Gaddis, N. C. & Malim, M. H. The antiretroviral enzyme APOBEC3G is degraded by the proteasome in response to HIV-1 Vif. Nature medicine 9, 1404–1407, https://doi.org/10.1038/nm945 (2003) Yu, X. et al. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science (New York, N.Y.) 302, 1056–1060, https://doi.org/10.1126/science.1089591 (2003) Goila-Gaur, R. et al. HIV-1 Vif promotes the formation of high molecular mass APOBEC3G complexes. Virology 372, 136–146, https://doi.org/10.1016/j.virol.2007.10.017 (2008) Lavens, D. et al. Definition of the interacting interfaces of Apobec3G and HIV-1 Vif using MAPPIT mutagenesis analysis. Nucleic acids research 38, 1902–1912, https://doi.org/10.1093/nar/gkp1154 (2010) Schrofelbauer, B., Senger, T., Manning, G. & Landau, N. R. Mutational alteration of human immunodeficiency virus type 1 Vif allows for functional interaction with nonhuman primate APOBEC3G. Journal of virology 80, 5984–5991, https://doi.org/10.1128/jvi.00388-06 (2006) Compton, A. A. & Emerman, M. Convergence and divergence in the evolution of the APOBEC3G-Vif interaction reveal ancient origins of simian immunodeficiency viruses. PLoS pathogens 9, e1003135, https://doi.org/10.1371/journal.ppat.1003135 (2013) Compton, A. A., Hirsch, V. M. & Emerman, M. The host restriction factor APOBEC3G and retroviral Vif protein coevolve due to ongoing genetic conflict. Cell host & microbe 11, 91–98, https://doi.org/10.1016/j.chom.2011.11.010 (2012) Gooch, B. D. & Cullen, B. R. Functional domain organization of human APOBEC3G. Virology 379, 118–124, https://doi.org/10.1016/j.virol.2008.06.013 (2008) Zhai, C. et al. Identification and characterization of loop7 motif and its role in regulating biological function of human APOBEC3G through molecular modeling and biological assay. Acta pharmaceutica Sinica. B 7, 571–582, https://doi.org/10.1016/j.apsb.2017.05.002 (2017) Letko, M., Booiman, T., Kootstra, N., Simon, V. & Ooms, M. Identification of the HIV-1 Vif and Human APOBEC3G ProteinInterface. Cell reports 13, 1789–1799, https://doi.org/10.1016/j.celrep.2015.10.068 (2015) Nathans, R. et al. Small-molecule inhibition of HIV-1 Vif. Nature biotechnology 26, 1187–1192, https://doi.org/10.1038/nbt.1496 (2008) Sinha, C. et al. Molecular docking analysis of RN18 and VEC5 in A3G-Vif inhibition. Bioinformation 10, 611–616, https://doi.org/10.6026/97320630010611 (2014) Mercenne, G. et al. HIV-1 Vif binds to APOBEC3G mRNA and inhibits its translation. Nucleic acids research 38, 633–646, https://doi.org/10.1093/nar/gkp1009 (2010) Matsui, M. et al. Small molecules that inhibit Vif-induced degradation of APOBEC3G. Virology journal 11, 122, https://doi.org/10.1186/1743-422x-11-122 (2014) Zuo, T. et al. Small-molecule inhibition of human immunodeficiency virus type 1 replication by targeting the interaction between Vif and ElonginC. Journal of virology 86, 5497–5507, https://doi.org/10.1128/jvi.06957-11 (2012) Zhang, S. et al. Identification of an HIV-1 replication inhibitor which rescues host restriction factor APOBEC3G in Vif-APOBEC3G complex. Antiviral research 122, 20–27, https://doi.org/10.1016/j.antiviral.2015.07.009 (2015) Zhang, Z. et al. Molecular modeling of human APOBEC3G to predict the binding modes of the inhibitor compounds IMB26 and IMB35. Acta Pharmaceutica Sinica B 3, 239–244, https://doi.org/10.1016/j.apsb.2013.05.002 (2013) Holden, L. G. et al. Crystal structure of the anti-viral APOBEC3G catalytic domain and functional implications. Nature 456, 121–124, https://doi.org/10.1038/nature07357 (2008) Siu, K. K., Sultana, A., Azimi, F. C. & Lee, J. E. Structural determinants of HIV-1 Vif susceptibility and DNA binding in APOBEC3F. Nature communications 4, 2593, https://doi.org/10.1038/ncomms3593 (2013) Bruns, R. F. & Watson, I. A. Rules for Identifying Potentially Reactive or Promiscuous Compounds. J Med Chem 55, 9763–9772, https://doi.org/10.1021/jm301008n (2012) High throughput docking for library design and library prioritization Jones, G., Willett, P., Glen, R. C., Leach, A. R. & Taylor, R. Development and validation of a genetic algorithm for flexible docking. Journal of molecular biology 267, 727–748, https://doi.org/10.1006/jmbi.1996.0897 (1997) Lecossier, D., Bouchonnet, F., Clavel, F. & Hance, A. J. Hypermutation of HIV-1 DNA in the absence of the Vif protein. Science (New York, N.Y.) 300, 1112, https://doi.org/10.1126/science.1083338 (2003) Wang, Z. et al. Heat-stable molecule derived from Streptococcus cristatus induces APOBEC3 expression and inhibits HIV-1 replication. PloS one 9, e106078, https://doi.org/10.1371/journal.pone.0106078 (2014) Cen, S. et al. Small molecular compounds inhibit HIV-1 replication through specifically stabilizing APOBEC3G. The Journal of biological chemistry 285, 16546–16552, https://doi.org/10.1074/jbc.M109.085308 (2010) Wang, X. et al. Human APOBEC3F incorporation into human immunodeficiency virus type 1 particles. Virus research 191, 30–38, https://doi.org/10.1016/j.virusres.2014.07.011 (2014) Pery, E. et al. Identification of a novel HIV-1 inhibitor targeting Vif-dependent degradation of human APOBEC3G protein. The Journal of biological chemistry 290, 10504–10517, https://doi.org/10.1074/jbc.M114.626903 (2015) Kouno, T. et al. Structure of the Vif-binding domain of the antiviral enzyme APOBEC3G. Nature structural & molecular biology 22, 485–491, https://doi.org/10.1038/nsmb.3033 (2015) Guo, Y. et al. Structural basis for hijacking CBF-beta and CUL5 E3 ligase complex by HIV-1 Vif. Nature 505, 229–233, https://doi.org/10.1038/nature12884 (2014) Nakashima, M. et al. Structural Insights into HIV-1 Vif-APOBEC3F Interaction. Journal of virology 90, 1034–1047, https://doi.org/10.1128/jvi.02369-15 (2015) Smith, J. L. & Pathak, V. K. Identification of specific determinants of human APOBEC3F, APOBEC3C, and APOBEC3DE and African green monkey APOBEC3F that interact with HIV-1 Vif. Journal of virology 84, 12599–12608, https://doi.org/10.1128/jvi.01437-10 (2010) Albin, J. S. et al. A single amino acid in human APOBEC3F alters susceptibility to HIV-1 Vif. The Journal of biological chemistry 285, 40785–40792, https://doi.org/10.1074/jbc.M110.173161 (2010) Structural disorder in the HIV-1 Vif protein and interaction-dependent gain of structure O’Boyle, N. M. et al. Open Babel: An open chemical toolbox. Journal of cheminformatics 3, 33, https://doi.org/10.1186/1758-2946-3-33 (2011) Li, M. et al. First-in-class small molecule inhibitors of the single-strand DNA cytosine deaminase APOBEC3G. ACS chemical biology 7, 506–517, https://doi.org/10.1021/cb200440y (2012) Cen, S. et al. The interaction between HIV-1 Gag and APOBEC3G. The Journal of biological chemistry 279, 33177–33184, https://doi.org/10.1074/jbc.M402062200 (2004) Wang, T., Tian, C., Zhang, W., Sarkis, P. T. & Yu, X. F. Interaction with 7SL RNA but not with HIV-1 genomic RNA or P bodies is required for APOBEC3F virion packaging. Journal of molecular biology 375, 1098–1112, https://doi.org/10.1016/j.jmb.2007.11.017 (2008) Yang, Y., Guo, F., Cen, S. & Kleiman, L. Inhibition of initiation of reverse transcription in HIV-1 by human APOBEC3F. Virology 365, 92–100, https://doi.org/10.1016/j.virol.2007.03.022 (2007) Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy Proceedings of the National Academy of Sciences of the United States of America 94 Asami, Y. et al. Protuboxepin A, a marine fungal metabolite, inducing metaphase arrest and chromosomal misalignment in tumor cells. Bioorganic & medicinal chemistry 20, 3799–3806, https://doi.org/10.1016/j.bmc.2012.04.039 (2012) Download references USA) for providing the candidate compounds This work was supported by the National Key Research and Development program of China (2016YFD0500307 CS) The National Natural Science Foundation of China (81271844 ZJM National Mega-project for Innovative Drugs (2012ZX09102101-018 CS) CAMS Innovation Fund for Medical Sciences (CAMS-I2M-1-012 ZJM) National Mega-Project for Significant new drug discovery (2018ZX09711003-002-002 CAMS Innovation Fund for Medical Sciences (2016-I2M-2-002 LX) Beijing Key Laboratory of Emerging Infectious Diseases (to WJ) and Xiehe Scholar to CS Chinese Academy of Medical Sciences & Peking Union Medical College performed the major part of the experimental study All authors analyzed the results and approved the final version of the manuscript Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations Download citation DOI: https://doi.org/10.1038/s41598-018-26318-3 Molecular and Cellular Biochemistry (2019) À vif! is a comedy-drama directed by John Wells, released in 2015. The film stars Bradley Cooper, Sienna Miller and Daniel Brühl in an intense exploration of the world of haute gastronomy known for his roles in Happiness Therapy and American Sniper With Sienna Miller and Daniel Brühl at his side plunges viewers into the ruthless world of Michelin-starred cuisine was crowned with two Michelin stars before his arrogance and excess led to his downfall he spent several years trying to regain his sobriety he surrounded himself with a team of talented young chefs to reopen a restaurant in London with the ultimate goal of obtaining a third Michelin star the ghosts of his past and the immense pressure of his ambition make his quest for redemption harder than expected He now has just one more chance to prove that he can become a true gastronomic legend will appeal to fans of culinary dramas and those fascinated by the demanding world of haute gastronomy The film stands out for its realistic and passionate portrayal of Michelin-starred cuisine with cooking scenes that are as intense as they are captivating Charismatic performances by Bradley Cooper and Sienna Miller add emotional depth to this story of redemption and perseverance offers a fascinating and moving immersion in the world of great chefs is an intense and emotional dive into the world of gastronomy driven by Bradley Cooper's magnetic performance this film promises to captivate viewers with its blend of drama BetweenAdam Jones' personal and professional challenges this quest for culinary perfection and redemption offers a rich and inspiring cinematic experience Prime Video: new Amazon Originals and Exclusive films and series in June 2024In June, explore Amazon Originals and Exclusives on Prime Video. [Read more] Refer your establishment, click herePromote your event, click here we are excited to announce the availability of AWS Outposts private connectivity the service link endpoints in the region for each AWS Outposts deployment were in the public AWS realm of a customer’s chosen region and could be connected to by either the public internet or an AWS Direct Connect (DX) public virtual interface (VIF) using AWS Outposts public service link connectivity meant that customers needed to enable a connection from the AWS Outposts rack out through their edge routers and firewalls to the service link public endpoint IPs The service link is important as it is a group of encrypted tunnels that are used for carrying management traffic and your intra Amazon VPC traffic between your chosen AWS Region and AWS Outposts Service link establishment is required before an Outpost can be used and must be maintained in order for AWS Outposts to continue to operate For an overview of AWS Outposts network connectivity, including public service link access, see the AWS Outposts – Network Reference Architecture The new AWS Outposts service link private connectivity feature uses AWS Direct Connect changing the endpoint for the service link from a public AWS endpoint to a set of private elastic network interfaces (ENIs) within an Amazon VPC deployed in your environment This means that the service link connects from your AWS Outposts privately over a DX private VIF to the private AWS Outposts service link endpoints in the Region Using a private VIF with your Virtual Private Gateway (VGW) and being attached to an Amazon VPC that you manage allows you to connect privately to the Outposts service link endpoints without having to traverse the public internet It additionally gives you the ability to use DX features to troubleshoot connectivity such as Direct Connect Amazon CloudWatch metrics Using the AWS Outposts private connectivity option for the service link also removes the need for using large public allow-lists on your on-premises firewall edge as the service link endpoints are in a VPC that you control using private addresses that you have allocated to your Amazon VPC Figure 1. AWS Outposts private connectivity and the Virtual Private Gateway. Note for simplicity only one DX connection is shown. We recommend reviewing the DX resiliency recommendations to select a DX architecture that meets your availably requirements is an AWS Outposts (A) installed at your premises with a service link connection (B) to the Outposts service endpoints (C) in the AWS region (D) on the far left we’re using the VGW (E) attached to your Amazon VPC (F) that terminates a DX private VIF (G) and serves as a private endpoint for the service link connections in the Region for your AWS Outposts we do not support VPN or AWS Transit Gateway for AWS Outposts private connectivity you may however want to use an Amazon Direct Connect gateway if needed for cross-region DX access The DX private VIF is a private connection to your edge router in your chosen DX location and uses BGP to exchange routes Your private Amazon VPC CIDR range is advertised through this BGP session to your edge router (10.2.0.0/16 in the Figure 1 the /26 IP address range for the Outpost service link (10.5.0.0/26 in the Figure 1 example) is advertised to the region via BGP from your edge router Note: It is required that your service link endpoint VPC not use the range 10.1.0.0/16 for it’s Amazon VPC CIDR range and that the Amazon VPC and subnet used be created in the same AWS account and Availability Zone as your AWS Outposts you will get the option to select private connectivity for the service link Once you select the private connectivity option AWS automatically creates a private connectivity endpoint and assigns private IPs to it from the VPC subnet’s CIDR that you have selected to use for the AWS Outposts private connectivity all service link traffic between your AWS Outposts rack and the AWS Outposts service endpoints in the Region will use your designated private connectivity For the service link private endpoint in your VPC, you can configure network ACLs (Access Control Lists) if needed, for the subnet that hosts the private service link endpoints. Traffic to this subnet can be restricted to TCP/UDP source/destination ports 443 as an example, see service link firewall requirements for more information on ACL configuration if needed Having a dedicated subnet for the private connectivity endpoint offers the advantage of a defining a single ACL to simplifying management and control of traffic related to the Outposts service link and dedicating a VPC for the service link private endpoints can also help minimize any Outposts service interruptions from other configuration actions in the future The following tutorial guides you through deploying a new AWS Outposts with private connectivity must not conflict with 10.1.0.0/16) in the same AWS account and Availability Zone as your Outpost DX transport between the on-premises Outpost location and the AWS region with a Private VIF connection into the VPC Advertise the subnet CIDR to your on-premises network Start by configuring some environment variables The selection for the private connectivity must be made when creating or provisioning your Outpost simply select the private connectivity option Then select the VPC and the Subnet where AWS will automatically create private connectivity endpoints for connecting to the Outpost service Outposts needs permission to create cross-account network interfaces and attach them to service link endpoint instances so you have to allow us to create a new role to this you can see the all the configurations that will be applied your behalf Finally you will have access to the dashboard where you can view the Outposts summary as well as the connectivity type configured A service role will be created in the process Maintaining the Outposts service link connection up is critical for management of the Outposts Ensure you have redundant connections to region in case of failure The private VPCs deployed in your VPC are fully under your control in order to avoid an unintendedly drop in the service link assign a service control policy that limits deletion of the following resources: Note: If you decide on leveraging a TGW for the service link Now when your Outpost connects back to the associated AWS Region it will use the private service link endpoints in a VPC of your choosing when establishing the AWS Outposts service link connection Private connectivity provides an additional option when public connectivity is not desired The public connectivity option for the Outposts service link is still available and supported by AWS Outposts if limiting your on-premises access to public networks and using an DX private VIF for AWS Outposts service link private connectivity is something that your organization needs 2024: An earlier version of this post misstated that AWS Outposts operation modes can be changed between public and private connectivity by opening a support ticket The post has been updated with the incorrect section removed 2025: The section “Best Practices” was added.  AWS Direct Connect now supports connections to AWS Transit Gateway at speeds of 500 megabits per second (Mbps) and lower The architecture described in this post is no longer needed AWS Transit Gateway provides you with the ability to connect multiple VPCs, VPNs and scale up to 5,000 attachments It simplifies management and reduces operational costs of networks within your AWS environments and connectivity from on-premises networks also known as a sub-1 Gbps hosted AWS Direct Connect connection I explain how to integrate a sub-1 Gbps hosted Direct Connect connection with AWS Transit Gateway without a transit virtual interface by using the following methods: Readers of this blog post should be familiar with Border Gateway Protocol (BGP) and the following AWS services: This method is similar to attaching a VPN to AWS Transit Gateway you establish a VPN to AWS Transit Gateway over AWS Direct Connect The following diagram depicts the scenario and the solution Figure 1: Connecting to transit gateway over a public VIF Because this is a hosted connection, you don’t have to create a connection in your AWS Direct Connect console. Your AWS Direct Connect Partner creates and delegates the connection to your account This architecture works well for point-to-point connections between AWS and the customer’s on-premises network it proves suboptimal for scenarios where the customer’s network consists of multiple sites connected over an MPLS network in a fully meshed manner MPLS L3 VPN provides the flexibility of connecting multiple sites privately you must use a Provider Edge (PE) router managed by the MPLS service provider Due to the scale and multi-tenant nature of these PE routers VPN tunnels are generally not configured on PE routers as that increases complexity and poses operational risks to this layer You can use dynamic or static routing for integrating MPLS L3 VPNs to AWS Transit Gateway The following diagram depicts the architecture used to integrate AWS Transit Gateway to a sub-1 Gbps hosted Direct Connect connection using a dynamic routing protocol Figure 2: Connecting to AWS Transit Gateway over a private virtual interface via L3 MPLS using BGP The following steps provide end-to-end connectivity between on-premises networks and VPCs behind an AWS Transit Gateway This architecture also manages the failover dynamically using BGP Multiple sets of BGP peering enable prefix exchanges between VPCs and on-premises networks The following list outlines the BGP peerings: you don’t have to create a connection in your AWS Direct Connect console for this hosted connection  The AWS Direct Connect Partner creates and delegates the connection to your account Figure 4: Connecting floating virtual private gateway to AWS Transit Gateway using BGP via Edge Transit VPC you will have end-to-end connectivity between VPCs and on-premises networks This option uses static routing between the EC2 instance running a router AMI in the edge transit VPC and the transit gateway It does not use VPN tunnels between EC2 instances with router AMI and AWS Transit Gateway allowing you to optimize cost by not incurring cost for the VPN connection This option still uses VPN tunnels between the floating virtual private gateway and EC2 instance running a router AMI The following diagram depicts this scenario and the steps to deploy it Figure 5: Connecting AWS Transit Gateway over a private VIF via L3 MPLS using static routes in transit gateway follow steps 1 through 6 under Option1: Using dynamic routing protocol between transit gateway and MPLS L3 VPN and then follow these additional steps Figure 6: Connecting floating virtual private gateway to AWS Transit Gateway via Edge Transit VPC using static routes or use automation to detect a failover and then switch the paths by modifying the route tables programmatically I described various ways of integrating AWS Transit Gateway with your on-premises networks over sub-1 Gbps hosted AWS Direct Connect using point-to-point and MPLS L3 VPN I hope this blog post helps you use AWS Transit Gateway with sub-1 Gbps hosted AWS Direct Connect connection and simplify the connectivity with your AWS environment The coexistence of two parties with opposing interests – in this case lentiviruses for replication and the host that attempts to evade viral infection – has led to the ongoing battle between the host and virus playing out on a molecular scale over evolutionary time A3G (also known by its longer acronym APOBEC3G) is a protein that prevents HIV from hijacking host cellular machinery to replicate its genetic material the A3G protein gets packaged into HIV virions to block viral replication the viral protein Vif destroys A3G to prevent it from getting packaged into virions in the first place both A3G and Vif have evolved to outsmart each other resulting in an ongoing molecular arms race Scientists have known about A3G and Vif’s molecular arms race for decades, but the structural basis of this interaction remained unknown. In a new study published in Nature a team of scientists from the Fred Hutchinson Cancer Center and the University of California San Francisco reported the first cryogenic electron microscopy structure of human A3G bound to HIV-1 Vif “The Vif/A3G story has been at the forefront of the conversation surrounding HIV evolution for over 20 years at this point,” said Dr. Michael Emerman a professor in the Human Biology and Basic Sciences Divisions at the Fred Hutch and a co-author on the study mutations in these proteins have given us a quasi-roadmap for how this virus family spilled over into hominids and other labs provided crucial insight into the specificities of this protein interface the structure was something many tried and failed to resolve for the last decade.” The team behind this paper, including Dr. Yen-Li Li, a postdoc in Dr. John Gross’s lab at UCSF, and Dr. Caleigh Azumaya, the former associate director of the Electron Microscopy Shared Resource at the Fred Hutch achieved this feat using cryogenic electron microscopy (cryo-EM) This technique utilizes an electron microscope with a beam of electrons as the source of light to image samples that have been cooled to cryogenic temperatures cryo-EM can render molecular structures at near-atomic resolution “The first thing that jumped out to us was that the ‘arms-race’ interface between A3G and Vif that had been predicted from positive selection analysis was indeed the site of interaction between A3G and Vif,” said Emerman Positive selection analyses identify specific sites in the protein that have undergone recurrent changes as a result of selective pressures likely results from its antagonizing interaction with Vif Previous work had identified two such sites in the A3G protein the identity of these sites is known to determine the adaptation of Vif to a new host species The cryo-EM structure of the site of interaction between A3G and Vif confirmed the prior hypothesis that the region of A3G under positive selection is the site of interaction with Vif is the presence of RNA at the Vif-A3G interface,” said  Caroline Langley a PhD candidate in the Emerman lab and a second author on the paper The cryo-EM structure revealed a single-stranded RNA molecule at the interface of the Vif and A3G proteins suggesting that RNA acts as a “molecular glue” that holds the two proteins together the cryo-EM structure continued to delight the team with the wealth of information it provided “It was also a surprise that Vif was bound to an A3G dimer,” said Langley The ability of A3G to form dimers is critical to its role as a viral restriction factor it cannot get packaged into virions to carry out subsequent antiviral activities it appears that Vif has evolved to target A3G when it poses the largest threat to viral replication.” Although cryo-EM provided the structural data the subsequent analyses of the structure provided much anticipated answers about the evolutionary relationship between Vif and A3G “This analysis revealed to us that the amino acids in the ‘arms race interface’ are highly variable and species specific the identities of the amino acids identified as binding RNA in the structure were highly conserved hinting that RNA interaction is evolutionarily important for Vif antagonism of A3G,” said Langley This article was first published by the Fred Hutch Cancer Center. Read the original. Become a member to receive the print edition four times a year and the digital edition monthly candidate in the molecular and cellular biology program at the University of Washington and the Fred Hutchinson Cancer Center and writes comedy in the crepuscular hours and we’ll send you a weekly email with recent articles Scientists find that liver protein inhibits of pertussis toxin offering a potential new treatment for bacterial respiratory disease Read more about this recent study from the Journal of Biological Chemistry Scientists discover that triacylglycerol synthesis enzyme drives lipoproteins secretion rather than lipid droplet storage Researchers analyze protein and RNA data across 13 cancer types to find similarities that could improve cancer staging Read about this recent article published in Molecular & Cellular Proteomics Scientists develop a software tool to categorize microbe species and antibiotic resistance markers to aid clinical and environmental research Scientists develop a bioinformatics program that maps omics data to metabolic pathways Read about this recent article published in Molecular &amp; Cellular Proteomics Learn how the JBC associate editor went from milking cows on a dairy farm to analyzing kinases in the lab Metrics details The human APOBEC3 family of DNA cytosine deaminases serves as a front-line intrinsic immune response to inhibit the replication of diverse retroviruses APOBEC3F and APOBEC3G are the most potent factors against HIV-1 HIV-1 viral infectivity factor (Vif) targets APOBEC3s for proteasomal degradation Here we report the crystal structure of the Vif-binding domain in APOBEC3F and a novel assay to assess Vif-APOBEC3 binding Our results point to an amphipathic surface that is conserved in APOBEC3s as critical for Vif susceptibility in APOBEC3F Electrostatic interactions likely mediate Vif binding structure-guided mutagenesis reveals a straight ssDNA-binding groove distinct from the Vif-binding site and an ‘aromatic switch’ is proposed to explain DNA substrate specificities across the APOBEC3 family This study opens new lines of inquiry that will further our understanding of APOBEC3-mediated retroviral restriction and provides an accurate template for structure-guided development of inhibitors targeting the APOBEC3-Vif axis (a) Schematic of the seven A3 intrinsic immune restriction factors (A3A The three classes of DNA cytosine deaminase domains The Z2-cytosine deaminase domains are further classified into three subgroups based on sequence similarity A3F and A3G are the two most potent A3 proteins and exhibit disparate Vif-binding sites (CD1 for A3G and CD2 for A3F) (b) SDS–PAGE of the purified A3Fc-CD2 after final size exclusion chromatography (c) BLI kinetic analysis of A3Fc-CD2 binding to ssDNA Biotin-labelled ssDNA was coupled to streptavidin-coated biosensors and monitored for binding to purified A3Fc-CD2 at 0 The data was analysed based on a 1:1 binding model using the BLItz Pro software with the fitted curves shown as grey lines The ssDNA sequence used in the assay is shown below the sensorgram with the A3Fc-CD2 deamination site and target DNA cytosine underlined and double underlined Histogram showing the percent mutation on specific rpoB nucleotide sequences for A3Fc-CD2 and A3G-CD2 Results are expressed as the percentage of total mutations from six independent experiments with at least 20 RifR colonies sequenced for both A3Fc-CD2 and A3G-CD2 Inhibition of HIV-1 Vif binding to A3F or A3G would allow the reactivation of effective host innate immune responses to HIV-1 inhibitors targeting the host side of the A3F–Vif or A3G–Vif interface will be less sensitive to viral mutations and will reduce the probability of HIV-1 variants developing resistance to drug treatments Here we report the crystal structure of the Vif-binding domain in A3F-CD2 Given that A3F and A3G are the most potent A3 restriction factors this is a key structure needed for the development of new classes of inhibitors to enhance intrinsic immunity against HIV We performed site-directed mutagenesis to map out the ssDNA-binding site which revealed a straight groove for DNA binding as well as a novel aromatic switch that conferrs nucleotide preferences we developed the first biophysical HIV-1 Vif-APOBEC3 binding assay and this allowed us to identify a negatively charged Vif-binding surface distinct from the ssDNA-binding groove In A3Fc-CD2, the catalytic zinc atom is sequestered by H249, C280 and C283 within the canonical (C/H)-(A/V)-E-(X23–28)-P-C-X2-C cytidine deaminase motif (Fig. 2a) The active site residues superimpose well in all A3 structures a water molecule is coordinated to the catalytic zinc to complete a tetrahedral geometry This water molecule is activated to become a nucleophile for deamination of the target deoxycytidine nucleotide the catalytic water molecule is not observed due to the moderate resolution of the electron density map loop 3 is shorter and no such protein–protein interactions were detected Both A3G-CD2 molecular surfaces are shown in the same orientation as panel (a) (d) Nucleic acid–protein interaction ELISA assay Alanine scanning mutagenesis of selected A3Fc-CD2 ssDNA-binding site residues Results are expressed as the mean relative absorbance (+s.d coli expressed WT A3G-CD2 and A3G-CD2 ‘YYFW’ Histogram showing the percent total mutation on specific rpoB nucleotide sequences Results are expressed as the percentage of total mutations from six independent experiments with at least 20 colonies sequenced mutations in the ssDNA-binding site did not affect the overall structure of A3Fc-CD2 thus we predict that differences here may determine target DNA specificity with a ‘YYFW’ motif (termed the nucleotide specificity box) the ‘YYFW’ motif is replaced by ‘YYFQ’ and ‘YDDQ’ motifs We propose that this ‘aromatic switch’ in the nucleotide specificity box determines substrate specificities in A3s the rpoB hotspots in A3G-CD2 ‘YYFW’ are in excellent agreement with those preferred by A3F Our Vif-binding assay enabled us to map the HIV-1 Vif-binding interface at single-residue resolution and provides a foundation for the development of new high-throughput assays a complementary binding surface on A3F-CD2 would be negatively charged and hydrophobic (a) BLI kinetic analysis of A3Fc-CD2 binding to refolded full-length HIV-1 Vif Biotin-labelled HIV-1 Vif was coupled to streptavidin-coated biosensors and monitored for binding to purified A3Fc-CD2 at 0 The data were analysed based on a 1:1 binding model as only one A3F deamination motif is found on the ssDNA The calculated fitted curves are shown as grey lines A number of aromatic and hydrophobic residues Y269 and F290) are buried at the A3Fc-CD2 core Residues L263 and S264 (shown in orange) were identified in this study to be not important for Vif binding The acquired immunodeficiency syndrome that results from HIV-1 infection remains a global health threat Interactions between host restriction factors and viral antagonists represent intriguing targets for the development of drugs to restrict viral replication and dissemination we present the crystal structure of a chimeric A3F C-terminal domain containing the Vif-interaction interface Our combinatorial approach encompassing structural biochemical and biophysical studies provides insights into the molecular determinants of ssDNA binding While our apo A3Fc-CD2 structure clearly shows a straight ssDNA-binding groove a substrate-induced conformational change may bring these residues into proximity to bind ssDNA it should be emphasized that our current structural understanding of A3 proteins is limited to the analysis of a single domain of A3 proteins It may also be possible that ssDNA binds to residues identified in both the ‘straight’ and ‘kinked’ models of DNA binding in the context of full-length or higher-ordered oligomeric A3 protein structures Our study clearly demonstrates the importance of four residues in the nucleotide specificity box and a switch in hydrophobicities in determining substrate specificities in A3 proteins suggesting no effects on overall structural stability We suggest that the hydrophobic site is involved in maintaining the structural integrity and stability of A3Fc-CD2 it may be possible that the hydrophobic Vif-binding site has a role in both protein stability and Vif binding Full characterization of the role of the hydrophobic site in Vif binding awaits the structural determination of an A3–Vif complex A negatively charged surface conserved with other Z2-cytosine deaminase domains is proposed to be important for Vif-binding Electrostatic potential mapped onto the molecular surface of A3C (PDB: 3VOW) The proposed footprints of the A3 negative and hydrophobic patch involved in Vif binding are shown by the solid and dashed lines A previously characterized ‘DPD’ motif involved in A3G Vif binding is displayed for the A3G-CD1 homology model Red and blue coloured regions denote negative and positive charges Note: the ssDNA-binding site is at the top of the depicted A3 molecules and has no overlap with the hydrophobic or negatively charged Vif-binding site Our crystal structure presented in this manuscript has now identified well-defined sites on A3Fc-CD2 involved in ssDNA and Vif binding We have also identified novel structural determinants that explain the differences in substrate specificities between A3 family members our studies and findings will be invaluable to the A3 community by providing the relevant structural scaffold for the development of effective HIV-1 inhibitors aimed at selectively disrupting the A3F-Vif interface The structures of A3A and A3F11X-CD2 align well with A3Fc-CD2 no conformational changes in the chimeric region of A3Fc-CD2 exist A3Fc-CD2 and mutants were expressed in Rosetta-2 (DE3) E Cell cultures were grown to OD600=0.8 and induced with a final concentration of 0.5 mM IPTG for 18 h at 25 °C Cells were resuspended in Ni-binding buffer (50 mM Tris-HCl pH 8.0 300 mM NaCl and 20 mM imidazole) with EDTA-free protease inhibitor cocktail and lysed at 30 kpsi using a hydraulic cell disruption system (Constant Systems TS benchtop) The lysate was centrifuged to remove cellular debris prior to loading onto Ni-NTA resin (Thermo Pierce) A3Fc-CD2 was washed with Ni-NTA binding buffer with 50 mM imidazole A3Fc-CD2 was eluted by a two-step gradient of Ni-binding buffer with 125 mM imidazole and Ni-binding buffer with 500 mM imidazole Fractions that contained A3Fc-CD2 were concentrated and purified on a Superdex-200 10/300 GL column equilibrated in Buffer A (10 mM Tris-HCl pH 7.5 A3Fc-CD2 was further purified by anion exchange chromatography Here A3Fc-CD2 was pooled and loaded onto a MonoQ HR 5/5 column equilibrated in Buffer B (10 mM Tris-HCl pH 7.5 A3Fc-CD2 was eluted using a linear gradient of 0–100% Buffer B with 1 M NaCl Purified A3Fc-CD2 was quantified by A280 and concentrated to 20 mg ml−1 for crystallization All molecular ribbon diagrams and vacuum electrostatic calculations were generated using the programme MacPyMOL This ssDNA has one A3F deamination site (5′-CTCA-3′) Biotinylated ssDNA was diluted to 50 μM in kinetics buffer (PBS immobilized on a streptavidin biosensor for 120 s 20 μM of biotinylated ssDNA was immobilized on the probe A3Fc-CD2 was diluted into kinetics buffer (0 2 and 4 μM) and association to the biotinylated ssDNA was measured over 300 s the biosensor was immersed in kinetics buffer for 300 s to measure dissociation The Kd was calculated using the BLItz Pro v.1.1.0.28 software Single-stranded 42-mer DNA (Fig. 1c) was synthesized (Integrated DNA Technologies) Biotinylated ssDNA (1.5 pmol) was immobilized onto a streptavidin-coated ELISA plate (Quidel) and washed three times with PBS with 0.01% (v/v) Tween-20 (PBS-T) Wells were blocked with 3% (w/v) BSA in PBS-T overnight at 4 °C 75 and 100 μg of A3Fc-CD2 or A3Fc-CD2 mutants were incubated at 22 °C for 1.5 h and subsequently washed three times with PBS-T A mouse anti-His primary mAb (Roche) diluted 1:3,000 in PBS-T was incubated for 1 h at 22 °C The plate was washed three times prior to incubation with a goat anti-mouse HRP-conjugated secondary mAb (Pierce) diluted 1:3,000 The plates were developed using TMB-One substrate solution (Kem-En-Tec Diagnostics) for 5 min Colour development was stopped with 2 N sulphuric acid and measured at 495 nm the biotinylated sense and antisense DNA strands (3′-AGG GAG TCT GGG AAA ATC AGT CAC ACC TTT TAG AGA TCG TCA-5′) were heated at 95 °C for 15 min and allowed to cool to 22 °C in annealing buffer (10 mM Tris-HCl (pH 7.5) The double-stranded DNA probe was then immobilized onto the streptavidin-coated ELISA plate and performed as described above All experiments were performed in triplicate A3Fc-CD2 or A3Fc-CD2 ssDNA-binding mutants cloned into pET46-Ek/LIC were transformed into BL21-(DE3) E Single colonies were used to inoculate a 10 ml culture of LB supplemented with 100 μg ml−1 ampicillin the cultures were induced with 1 mM final concentration of IPTG and grown overnight at 37 °C Cell cultures were normalized (7 ml of cells at OD600=0.5) and plated onto LB-agar plates containing 100 μg ml−1 rifampicin to select for RifR clones Single colonies were picked from plates and colony PCR was performed to amplify the bacterial RNA polymerase gene rpoB The rpoB PCR products were DNA sequenced with an rpoB sequencing primer (5′-GGC-GAA-ATG-GCG-GAA-AAC-3′) A3F or A3G DNA-binding motifs in the rpoB gene were analysed for C–to-T mutations CD wavelength scans and thermal titrations were performed on all A3Fc-CD2 proteins at a concentration of 1.0 mg ml−1 in PBS 0.05% (w/v) CHAPS and 1 M guanidine-HCl on a Jasco J-810 spectropolarimeter CD wavelength scans collected between 190–250 nm using a 1-mm quartz cuvette (Helma) were averaged over five scans Thermal denaturation of A3Fc-CD2 and mutants were performed by increasing the temperature from 20–95 °C and monitoring the loss in CD signal at 222 nm HIV-1 Vif was expressed in BL21-(DE3) E. coli cells, purified and refolded from inclusion bodies, as previously described32 Vif inclusion bodies were resolubilized in 6 M guanidine–HCl and 10 mM Tris–HCl (pH 7.4) overnight at 22 °C and subsequently clarified by centrifugation prior to Ni-NTA purification The Ni-NTA column was washed with 8 M urea 100 mM NaH2PO4 and 10 mM Tris-HCl (pH 6.9) and Vif was eluted in the same buffer at pH 4.5 Vif was subsequently purified on a prep grade Superdex-75 10/300 column equilibrated in 8 M urea 10 mM β-mercaptoethanol and 10 mM Tris-HCl pH 4.5 The peak corresponding to monomeric Vif was collected and diluted to 0.1 mg ml−1 and dialyzed against 100 mM NaH2PO4 20% (v/v) glycerol (pH 6.0) with decreasing concentration of urea (6 The final refolded HIV-1 Vif was dialyzed against 10 mM NaH2PO4 pH 6.0 and biotinylated using the EZ-Link Sulfo-NHS-LC-Biotinylation kit (Thermo Pierce) according to the manufacturer’s protocol Biotinylated Vif was diluted to 40 μg ml−1 in 10 mM NaH2PO4 1 mg ml−1 BSA and 0.002% (v/v) Tween-20 and immobilized onto a BLI streptavidin probe for 120 s A3Fc-CD2 mutants were diluted into 10 mM Tris-HCl (pH 7.5) 1 mg ml−1 BSA and 0.002% (v/v) Tween-20 and allowed to associate over 80 s Purified A3G-CD2 was used as a negative control Accession codes: Atomic coordinates and structure factors for A3Fc-CD2 have been deposited in the Protein Data Bank (PDB) with the accession code 4J4J An anthropoid-specific locus of orphan C to U RNA-editing enzymes on chromosome 22 Quantitative profiling of the full APOBEC3 mRNA repertoire in lymphocytes and tissues: implications for HIV-1 restriction APOBEC3G multimers are recruited to the plasma membrane for packaging into human immunodeficiency virus type 1 virus-like particles in an RNA-dependent process requiring the NC basic linker HIV-1 Vif alters processive single-stranded DNA scanning of the retroviral restriction factor APOBEC3G Multiple ways of targeting APOBEC3-virion infectivity factor interactions for anti-HIV-1 drug development interaction and real-time monitoring of the enzymatic reaction of wild-type APOBEC3G Crystal structure of the anti-viral APOBEC3G catalytic domain and functional implications Crystal structure of the APOBEC3G catalytic domain reveals potential oligomerization interfaces Rationalisation of the differences between APOBEC3G structures from crystallography and NMR studies by molecular dynamics simulations APOBEC2 is a monomer in solution: implications for APOBEC3G models Dissecting APOBEC3G substrate specificity by nucleoside analog interference Biophysical characterization of recombinant HIV-1 subtype C virus infectivity factor Single-strand specificity of APOBEC3G accounts for minus-strand deamination of the HIV genome Mutational comparison of the single-domained APOBEC3C and double-domained APOBEC3F/G anti-retroviral cytidine deaminases provides insight into their DNA target site specificities Hydrophobicities of the nucleic acid bases: distribution coefficients from water to cyclohexane An automated microseed matrix-screening method for protein crystallization The finer things in X-ray diffraction data collection Protein structure prediction on the Web: a case study using the Phyre server A new bioinformatics analysis tools framework at EMBL-EBI ENDscript: a workflow to display sequence and structure information Download references This work was supported by an Ontario HIV Treatment Network Operating Grant (ROG-G645) Canada Research Chair in Structural Virology and a Canadian Institutes of Health Research (CIHR) New Investigator Award (MSH-113554) to J.E.L A CIHR Postdoctoral Fellowship was awarded to K.K.S. was supported by a University of Toronto Graduate Scholarship and CIHR Masters Graduate Fellowship We are indebted to Douglas Instruments (UK) for the generous use of their Oryx 8 crystallization robot for microseeding and Aiping Dong Professors Cheryl Arrowsmith and Aled Edwards from the Structural Genomics Consortium (University of Toronto) for access to their X-ray diffraction facility This work is based upon research conducted at the NE-CAT 24-ID-E beamline (Advanced Photon Source supported by award RR-15301 from the National Center for Research Resources (NIH) Use of the APS is supported by the US Department of Energy Department of Laboratory Medicine and Pathobiology conceived and supervised the project; K.K.S. crystallization and structure determination; A.S bacterial RifR DNA cytosine deaminase assays and CD spectroscopy; F.C.A purification and refolding of HIV-1 Vif for binding studies; K.K.S Supplementary Table S1 and Supplementary Discussion (PDF 1082 kb) Download citation Metrics details The essential HIV-1 viral infectivity factor (Vif) allows productive infection of non-permissive cells expressing cytidine deaminases APOBEC3G (A3G) and A3F by decreasing their cellular level and preventing their incorporation into virions the functional role of the inhibition of A3G translation by Vif remained unclear we show that two stem-loop structures within the 5′-untranslated region of A3G mRNA are crucial for translation inhibition by Vif in cells and most Vif alleles neutralize A3G translation efficiently K26R mutation in Vif abolishes degradation of A3G by the proteasome but has no effect at the translational level indicating these two pathways are independent proteasomal degradation and translational inhibition similarly contribute to decrease the cellular level of A3G by Vif and to prevent its incorporation into virions inhibition of A3G translation is sufficient to partially restore viral infectivity in the absence of proteosomal degradation These findings demonstrate that HIV-1 has evolved redundant mechanisms to specifically inhibit the potent antiviral activity of A3G the relative importance of the translational inhibition of A3G by Vif compared to the well-documented A3G degradation and its impact on viral infectivity remained to be established we used several A3G mRNA expression plasmids mutated in their UTRs with and without inhibitors of A3G degradation by the proteasome Our data show that two stem-loop structures in the 5′-UTR of A3G mRNA are required for translational inhibition by Vif The property of Vif to inhibit the translation of A3G is common to a large variety of Vif alleles and was also demonstrated in HIV-1 chronically infected H9 cells which abolishes degradation of A3G by the proteasome but has no effect on the translational repression of A3G demonstrating that these two pathways are independent These two mechanisms contribute to the decrease of the intracellular level of A3G by Vif and to the subsequent A3G incorporation into virions the inhibition of A3G translation by Vif is sufficient to partially restore viral infectivity in A3G expressing cells in the absence of proteasomal degradation These findings demonstrate that HIV-1 has evolved several redundant mechanisms to specifically inhibit the potent antiviral activity of A3G proteins Schematic representation of A3G constructs used in this study. Wild-type authentic A3G mRNA and mutants deleted from their 5′, 3′ or 5′ and 3′-UTRs are represented. Secondary structures of the 5′- and 3′UTRs of wild-type A3G mRNA are also indicated with high affinity binding sites for Vif depicted in grey. Dotted lines represent the deletions. Vif inhibits A3G translation in a 5′UTR dependent manner All samples derive from the same experiment and blots were processed in parallel Vif inhibits A3G translation in HIV-1 chronically infected H9 cells Heterologous 5′UTRs do not allow inhibition of A3G translation by Vif Vif requires SL2 and SL3 to impair A3G translation Vif K26 residue is required for the translational inhibition of A3G Effect of the inhibition of A3G translation by Vif on A3G packaging and viral infectivity these two mechanisms significantly contribute to exclude A3G from viral particles the amount of Vif protein encapsidated into viral particles was constant under all conditions studied indicating that a direct competition between Vif and A3G for packaging is unlikely These results indicate that the inhibition of A3G translation by Vif is sufficient to partially restore HIV-1 infectivity Translational repression of A3G by different Vif alleles HEK 293T cells were transfected with plasmids expressing wild-type A3G mRNA in the presence of Vif alleles and proteasome inhibition (ALLN) and the relative A3G expression was analyzed by western blot and quantify using Image J (1.46r) Standard deviations are representative of at least three independent experiments raising the possibility that Vif/A3G interaction might be required for translational regulation suggesting that specific sequences/domains of Vif are required to down-regulate A3G translation This translational control is corroborated by the fact that this property is shared by almost all Vif proteins and opens attractive perspectives for the development of new drugs disrupting the translational control of A3G by Vif considering that the 5′UTR of A3G and A3F mRNAs is highly conserved it is likely that Vif is also able to inhibit A3F translation into the double digested pCMV A3G expression vector All constructs were confirmed by DNA sequencing (GATC Biotech HEK 293T cells were co-transfected with pA3G-HA and plasmids expressing wild type or mutant Vif 0.05% SDS) supplemented with protease inhibitors (cOmplete EDTA Free cocktail an aliquot fraction (50 μl) was used for determination of the protein expression level and the remaining was incubated 2 h at 4 °C with 1 μg of HA antibody (Santa Cruz protein A Dynabeads (Life Technologies) were added and incubated for 1.5 h at 4 °C NuPAGE LDS sample buffer (Life Technologies) supernatant was loaded on NuPAGE gel (Life Technologies) and analyzed by western blot Polyclonal anti-hA3G (#9968) and monoclonal anti-HIV-1 Vif (#319) antibodies were obtained through the NIH AIDS Research and Reference Reagent Program Monoclonal anti-β-actin antibody was purchased from SIGMA (#A5316) An HIV-positive patient serum was used for the identification of HIV-1 p24 protein The PVDF membranes were then incubated with horseradish peroxidase-conjugated secondary antibodies (BIO-RAD) and the proteins were visualized by enhanced chemiluminescence (ECL) using the ECL Prime Western blotting detection reagent (GE Healthcares) Bands were quantified using Image J (1.46r) by analyzing pixel density Student’s T-test was used to determine statistical significance 5.106 cells (treated or not with ALLN/DMSO) were harvested by centrifugation and lysed for 10 min at 4 °C in RIPA 1X supplemented with protease inhibitors Lysates were cleared by centrifugation for 30 min at 14,000 g and protein concentration was determined using a Bradford assay in order to load the equivalent of 150 μg of total proteins on a NuPAGE® Novex® 4–12% Bis-Tris gels (Life Technologies) Western blot was then performed as above using antibodies directed against A3G (NIH#9968) GAPDH (ABD Serotec-Bio-Rad); p24 (HIV-positive patient serum) and Ubiquitin (Santa Cruz Appropriate HRP-conjugated secondary antibodies were used and revealed by chemiluminescence total RNA was isolated from 293T cells using TRI Reagent (SIGMA) total RNA was isolated by phenol/chloroform extraction followed by ethanol precipitation Total RNA (1 μg) was then reverse-transcribed using the iScriptTM Reverse Transcription Supermix (BIO-RAD) as recommended by the manufacturer Subsequent qPCR analysis was performed using the KAPA SYBR® FAST qPCR Master Mix (KAPA BIOSYSTEMS) and was monitored on a CFX Real Time System (BIO-RAD) Gene-specific primers were: A3G forward primer and reverse primer 5′-TTCCAAAAGGGAATCACGTC-3′; β-actin forward primer and reverse primer 5′-AGCACTGTGTTGGCGTACAG-3′ The A3G mRNA levels were normalized to those of β-actin mRNA and relative quantification was determined using the standard curve based method Translational regulation of APOBEC3G mRNA by Vif requires its 5′UTR and contributes to restoring HIV-1 infectivity The restriction factors of human immunodeficiency virus Tumultuous relationship between the human immunodeficiency virus type 1 viral infectivity factor (Vif) and the human APOBEC-3G and APOBEC-3F restriction factors APOBEC proteins and intrinsic resistance to HIV-1 infection Remarkable lethal G-to-A mutations in vif-proficient HIV-1 provirus by individual APOBEC3 proteins in humanized mice APOBEC3G is incorporated into virus-like particles by a direct interaction with HIV-1 Gag nucleocapsid protein HIV-1 and MLV Gag proteins are sufficient to recruit APOBEC3G into virus-like particles The role of innate APOBEC3G and adaptive AID immune responses in HLA-HIV/SIV immunized SHIV infected macaques Suppression of HIV-1 infection by APOBEC3 proteins in primary human CD4(+) T cells is associated with inhibition of processive reverse transcription as well as excessive cytidine deamination APOBEC3F/G and Vif: action and counteraction Host restriction factors in retroviral infection: promises in virus-host interaction Identification of the HIV-1 Vif and Human APOBEC3G Protein Interface The Binding Interface between Human APOBEC3F and HIV-1 Vif Elucidated by Genetic and Computational Approaches Vif hijacks CBF-β to degrade APOBEC3G and promote HIV-1 infection CBF-β stabilizes HIV Vif to counteract APOBEC3 at the expense of RUNX1 target gene expression Differential requirements for HIV-1 Vif-mediated APOBEC3G degradation and RUNX1-mediated transcription by core binding factor beta Identification of HIV-1 Vif regions required for CBF-β interaction and APOBEC3 suppression The human immunodeficiency virus type 1 Vif protein reduces intracellular expression and inhibits packaging of APOBEC3G (CEM15) The mechanism of eukaryotic translation initiation and principles of its regulation Regulation of mRNA translation by 5′- and 3′-UTR-binding factors HIV-1 replication and the cellular eukaryotic translation apparatus Stoichiometry of the antiviral protein APOBEC3G in HIV-1 virions Highly purified human immunodeficiency virus type 1 reveals a virtual absence of Vif in virions Multiple lysines combined in HIV-1 Vif determines the responsiveness to CBF-beta Identification of an APOBEC3G binding site in human immunodeficiency virus type 1 Vif and inhibitors of Vif-APOBEC3G binding Evidence that ecotropic murine leukemia virus contamination in TZM-bl cells does not affect the outcome of neutralizing antibody assays with human immunodeficiency virus type 1 Viral RNA is required for the association of APOBEC3G with human immunodeficiency virus type 1 nucleoprotein complexes Effects of lysine to arginine mutations in HIV-1 Vif on its expression and viral infectivity Production of acquired immunodeficiency syndrome-associated retrovirus in human and nonhuman cells transfected with an infectious molecular clone Cytoskeleton association and virion incorporation of the human immunodeficiency virus type 1 Vif protein Different effects of the TAR structure on HIV-1 and HIV-2 genomic RNA translation HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis Download references Redmond Smyth for critical reading of the manuscript France) who kindly provided us vectors expressing the different Vif alleles and vectors expressing heterologous 5′UTR The following reagents were obtained through the AIDS Research and Reference Reagent Program NIH: A3G polyclonal antibody (#9968) from Dr Warner Greene and Vif monoclonal antibody (#319) from Dr This work was supported by a grant from the French National Agency for Research on AIDS and Viral Hepatitis (ANRS) and SIDACTION to J.C.P. and by post-doctoral (J.B.) and doctoral (S.G also received funding from the Ecuadorian government through the National Secretary of Higher Education Technology and Innovation (Secretaría Nacional de Educación Superior Funding for open access charge: French National Center for Research (CNRS) The Barcelona Institute for Science and Technology Roland Marquet & Jean-Christophe Paillart Laboratoire d’ImmunoRhumatologie Moléculaire Fédération de Médecine Translationnelle de Strasbourg (FMTS) wrote the paper with contributions from R.M Download citation Metrics details using an affinity tag/purification mass spectrometry approach that Vif additionally recruits the transcription cofactor CBF-β to this ubiquitin ligase complex which normally functions in concert with RUNX DNA binding proteins allows the reconstitution of a recombinant six-protein assembly that elicits specific polyubiquitination activity with APOBEC3G Using RNA knockdown and genetic complementation studies we also demonstrate that CBF-β is required for Vif-mediated degradation of APOBEC3G and therefore for preserving HIV-1 infectivity simian immunodeficiency virus (SIV) Vif also binds to and requires CBF-β to degrade rhesus macaque APOBEC3G Methods of disrupting the CBF-β–Vif interaction might enable HIV-1 restriction and provide a supplement to current antiviral therapies that primarily target viral proteins HIV-1 accessory proteins—ensuring viral survival in a hostile environment Interactions of host APOBEC3 restriction factors with HIV-1 in vivo: implications for therapeutics Getting into position: the catalytic mechanisms of protein ubiquitylation Recognition of the polyubiquitin proteolytic signal Purification and characterization of HIV-human protein complexes Global landscape of HIV–human protein complexes Nature doi:10.1038/nature10719 (this issue) VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases E2-RING expansion of the NEDD8 cascade confers specificity to cullin modification Interplay of transcription factors in T-cell differentiation and function: the role of Runx Mechanism of lysine 48-linked ubiquitin-chain synthesis by the cullin-RING ubiquitin-ligase complex SCF-Cdc34 Certain pairs of ubiquitin-conjugating enzymes (E2s) and ubiquitin-protein ligases (E3s) synthesize nondegradable forked ubiquitin chains containing all possible isopeptide linkages APOBEC3 proteins mediate the clearance of foreign DNA from human cells Sequential E2s drive polyubiquitin chain assembly on APC targets Priming and extending: a UbcH5/Cdc34 E2 handoff mechanism for polyubiquitination on a SCF substrate Multimodal activation of the ubiquitin ligase SCF by Nedd8 conjugation Download references Gross and Harris laboratories for comments Schulman and the AIDS Research and Reference Reagent Program for reagents This research was funded by grants from QB3 at University of California and the National Institutes of Health (P50 GM082250 P01 AI090935 and P50 GM081879 to N.J.K.; U54 RR022220 to A.S.; R01 AI064046 and P01 GM091743 to R.S.H.; P50 GM082250 to J.D.G and C.S.C.; P41RR001614 and P50GM081879 to A.B.) is a Searle Scholar and a Keck Young Investigator Hultquist: These authors contributed equally to this work California Institute for Quantitative Biosciences Supplementary References and Supplementary Figures 1-8 with legends Download citation The transcription cofactor CBF-β (core binding factor β) regulates the DNA binding activity of RUNX family proteins Two independent studies now show that CBF-β also regulates the ability of HIV-1 to evade host restriction mediated by the cDNA deaminase APOBEC3G a host factor that blocks viral replication They show that it associates with the HIV protein Vif and is essential for the assembly of the Vif-Cul5 E3 ubiquitin ligase complex which mediates the ubiquitination and destruction of APOBEC3 Both groups suggest that disrupting the Vif–CBF-β interaction could provide a new therapeutic target against HIV-1 infection VIF International Education, a K-12 professional development company, has acquired Participate Learning a teacher resource aggregator and search engine Financial terms of the deal were not disclosed Short for “Visiting International Faculty,” VIF was founded in 1987 to help universities recruit international faculty and later turned its attention to K-12 educators and bringing international “visiting” teachers to the U.S The organization later rebranded as VIF International Education in the 1990s focused on providing more general professional development around developing educators’ global and cultural awareness VIF International CEO David Young tells EdSurge that he and his team have been interested in turning the organization “into a full-blown edtech company,” where VIF provides educators with the tools they need to do their job “We needed to train our teachers and provide them with resources to be successful,” VIF International CEO David Young tells EdSurge adding that VIF currently has 20,000 teachers (1,000 of whom are “visiting international faculty from about 30 countries around the world”) on its platform Young explains that Participate Learning’s online platform will power VIF’s programming starting in the 2016-20­17 school year with Participate’s team developing a “better user experience” to sustain “more stability Journalism that ignites your curiosity about education EdSurge is an editorially independent project ofand