Vif Proteins of Human and Simian Immunodeficiency Viruses Require Cellular CBFβ to Degrade APOBEC3 Restriction Factors

Similar documents
Supplementary Materials for

Induction of APOBEC3G Ubiquitination and Degradation by an HIV-1 Vif-Cul5-SCF Complex

Small molecules that inhibit Vif-induced degradation of APOBEC3G

Macmillan Publishers Limited. All rights reserved

Zinc chelation inhibits HIV Vif activity and liberates antiviral function of the cytidine deaminase APOBEC3G

Moderate Influence of Human APOBEC3F on HIV-1 Replication in Primary Lymphocytes

NIH Public Access Author Manuscript Expert Rev Mol Med. Author manuscript; available in PMC 2010 April 28.

Interactions between HIV-1 Vif and human ElonginB-ElonginC are important for CBF-β binding to Vif

Requirement of HIV-1 Vif C-terminus for Vif-CBF-β interaction and assembly of CUL5-containing E3 ligase

Analysis of Vif-induced APOBEC3G degradation using an α-complementation assay

Role of poly-proline motif in HIV-2 Vpx expression

Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy

Report. Evolution of HIV-1 Isolates that Use a Novel Vif-Independent Mechanism to Resist Restriction by Human APOBEC3G

Degradation of the cancer genomic DNA deaminase APOBEC3B by SIV Vif

Dispersed Sites of HIV Vif-Dependent Polyubiquitination in the DNA Deaminase APOBEC3F

JBC Papers in Press. Published on October 22, 2010 as Manuscript M

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins

Human Immunodeficiency Virus

supplementary information

Supplementary Materials

Supplementary Figure 1. SC35M polymerase activity in the presence of Bat or SC35M NP encoded from the phw2000 rescue plasmid.

SUPPLEMENTARY INFORMATION

Viral Vectors In The Research Laboratory: Just How Safe Are They? Dawn P. Wooley, Ph.D., SM(NRM), RBP, CBSP

Identification of a Single Amino Acid Required for APOBEC3 Antiretroviral Cytidine Deaminase Activity

Supplemental Information

RAW264.7 cells stably expressing control shrna (Con) or GSK3b-specific shrna (sh-

Tel: ; Fax: ;

DATA SHEET. Provided: 500 µl of 5.6 mm Tris HCl, 4.4 mm Tris base, 0.05% sodium azide 0.1 mm EDTA, 5 mg/liter calf thymus DNA.

SUPPLEMENTARY INFORMATION

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

SUPPLEMENTARY INFORMATION

Tetherin/BST-2 Antagonism by Nef Depends on a Direct Physical Interaction between Nef and Tetherin, and on Clathrin-mediated Endocytosis

Supplementary Figure 1

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia

Identification of the Function of the Vpx Protein of Primate Lentiviruses: A Dissertation

SUPPLEMENTAL FIGURE LEGENDS

Choosing Between Lentivirus and Adeno-associated Virus For DNA Delivery

VIRAL TITER COUNTS. The best methods of measuring infectious lentiviral titer

Primate lentiviral Vpx commandeers DDB1 to counteract a macrophage restriction

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Nature Medicine: doi: /nm.2109

Appendix. Table of Contents

Retroviruses. ---The name retrovirus comes from the enzyme, reverse transcriptase.

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

Supplementary Figure 1

Reviewers' comments: Reviewer #1 (Remarks to the Author):

Certificate of Analysis

The effect of HIV 1 Vif polymorphisms on A3G anti viral activity in an in vivo mouse model

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

Host hindrance to HIV replication

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Supplementary Figure 1. PD-L1 is glycosylated in cancer cells. (a) Western blot analysis of PD-L1 in breast cancer cells. (b) Western blot analysis

Role of cullin-elonginb-elonginc E3 complex in bovine immunodeficiency virus and maedi-visna virus Vif-mediated degradation of host A3Z2-Z3 proteins

This training module is required for all personnel listed on an IBC protocol that describes work utilizing viral vectors (both replication competent

A Single Amino Acid Difference in Human APOBEC3H Variants Determines HIV-1 Vif Sensitivity

Moloney leukemia virus 10 (MOV10) through interference with the Vif mediated ubiquitin proteasome pathway

A review of HIV restriction factors and viral countering mechanisms

Lecture 6. Burr BIO 4353/6345 HIV/AIDS. Tetramer staining of T cells (CTL s) Andrew McMichael seminar: Background

Proteasome-dependent disruption of the E3 ubiquitin ligase anaphase-promoting complex by HCMV protein pul21a

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures

FIG S1 Examination of eif4b expression after virus infection. (A) A549 cells

Identification of Specific Determinants of Human APOBEC3F, APOBEC3C, and APOBEC3DE and African Green Monkey APOBEC3F That Interact with HIV-1 Vif

A. List of selected proteins with high SILAC (H/L) ratios identified in mass

Overview: Chapter 19 Viruses: A Borrowed Life

Co-evolution of host and pathogen: HIV as a model. Can Keşmir Theoretical Biology/Bioinformatics Utrecht University, NL

Running Loose or Getting Lost: How HIV-1 Counters and Capitalizes on APOBEC3-Induced Mutagenesis through Its Vif Protein

Severe Restriction of Xenotropic Murine Leukemia Virus-Related Virus Replication and Spread in Cultured Human Peripheral Blood Mononuclear Cells

m 6 A mrna methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer

Translation. Host Cell Shutoff 1) Initiation of eukaryotic translation involves many initiation factors

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

The functional investigation of the interaction between TATA-associated factor 3 (TAF3) and p53 protein

Supplemental Information. NRF2 Is a Major Target of ARF. in p53-independent Tumor Suppression

Does A Significant Change in CD4 Cell Count Affect The Number of Amino Acid Mutations In A Subject s Corresponding DNA Sequence

Supplementary Information. Supplementary Figure 1

~Lentivirus production~

SUPPLEMENTARY INFORMATION

CRISPRaTest Functional dcas9-activator Assay Kit v1 Last update: 2018/07/04 Cellecta, Inc.

Chapter 6- An Introduction to Viruses*

, virus identified as the causative agent and ELISA test produced which showed the extent of the epidemic

Lecture 2: Virology. I. Background

HIV Anti-HIV Neutralizing Antibodies

Jumpstart your research with ViraPower Lentiviral Expression Systems

Gag. Gag-Gag. TSG101 Nedd4. Two-Hybrid HIV Gag. Two-Hybrid. Two-Hybrid

APOBEC3G Contributes to HIV-1 Variation through Sublethal Mutagenesis

Inhibition of trna 3 Lys -Primed Reverse Transcription by Human APOBEC3G during Human Immunodeficiency Virus Type 1 Replication

The use of nonhuman primates in biomedical research has led to the isolation of many

DNA context and promoter activity affect gene expression in lentiviral vectors

Nature Immunology doi: /ni Supplementary Figure 1. Raf-1 inhibition does not affect TLR4-induced type I IFN responses.

Adaptive Evolution and Antiviral Activity of the Conserved Mammalian Cytidine Deaminase APOBEC3H

Inhibition of HIV-1 Integration in Ex Vivo-Infected CD4 T Cells from Elite Controllers

A second human antiretroviral factor, APOBEC3F, is suppressed by the HIV-1 and HIV-2 Vif proteins

Human immunodeficiency virus type 1 splicing at the major splice donor site is controlled by highly conserved RNA sequence and structural elements

19 Viruses BIOLOGY. Outline. Structural Features and Characteristics. The Good the Bad and the Ugly. Structural Features and Characteristics

p47 negatively regulates IKK activation by inducing the lysosomal degradation of polyubiquitinated NEMO

HIV-1 Viral Infectivity Factor Recruitment of Cellular. Protein Complexes to Counter Host Antiviral Response

Figure S1. Reduction in glomerular mir-146a levels correlate with progression to higher albuminuria in diabetic patients.

Certificate of Analysis

SUPPLEMENTARY INFORMATION

The humoral immune responses to IBV proteins.

Materials and Methods , The two-hybrid principle.

Transcription:

JVI Accepts, published online ahead of print on 28 December 2011 J. Virol. doi:10.1128/jvi.06950-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 Vif Proteins of Human and Simian Immunodeficiency Viruses Require Cellular CBFβ to Degrade APOBEC3 Restriction Factors Running Head: Functional Conservation of the Vif-CBFβ Interaction Judd F. Hultquist 1,2, Mawuena Binka 3, Rebecca S. LaRue 1, Viviana Simon 3,4,5, and Reuben S. Harris1, 2,* Figures: 4 Abstract: 75 words Manuscript: 1028 words 1Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA. 2Department of Molecular, Cellular, Developmental Biology and Genetics, University of Minnesota, Minneapolis, Minnesota 55455, USA. 3Department of Microbiology, Mount Sinai School of Medicine, New York, New York 10029, USA. 4The Global Health and Emerging Pathogens Institute, Mount Sinai School of Medicine, New York, New York 10029, USA. 5Division of Infectious Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA. * Corresponding author. Mailing address: University of Minnesota, Department of Biochemistry, Molecular Biology and Biophysics, 321 Church Street S.E., 6-155 Jackson Hall, Minneapolis, MN 55455. Phone: (612) 624-0457. Fax: (612) 625-2163. E-mail: rsh@umn.edu 1

45 46 47 48 49 50 51 52 ABSTRACT HIV-1 requires the cellular transcription factor CBFβ to stabilize its accessory protein Vif and promote APOBEC3G degradation. Here, we demonstrate that both isoforms of CBFβ allow for increased steady-state levels of Vif, enhanced APOBEC3G degradation, and increased viral infectivity. This conserved functional interaction enhances the steady-state levels of Vif proteins from multiple HIV-1 subtypes and is required for the degradation of all human and rhesus Vif-sensitive APOBEC3 proteins by their respective lentiviral Vif proteins. 53 54 2

55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 HIV-1 and related lentiviruses require the viral accessory protein Vif to neutralize members of the APOBEC3 family of retroviral restriction factors and render host cells permissive for productive viral replication. HIV-1 Vif neutralizes the APOBEC3 proteins by recruitment of an E3 ubiquitin ligase complex that polyubiquitinates APOBEC3 proteins and targets them for proteasomal degradation [(13), reviewed by (1, 9, 12)]. Recently, the cellular transcription factor CBFβ was found to be associated with this complex and allows for its reconstitution in vitro (6). Furthermore, CBFβ was found to be required for the stability of HIV-1 IIIB Vif in vivo, allowing for efficient degradation of APOBEC3G (A3G) and increased viral infectivity (6). The current model is that HIV-1 Vif hijacks cellular CBFβ to facilitate Vif folding and/or stability as well as nucleation of the E3 ubiquitin ligase complex. While it has been shown that SIV mac239 Vif also requires CBFβ to degrade rhesus A3G (6), the generality of the CBFβ/Vif/APOBEC3 functional interplay remains to be determined. The goal of the current study was to determine which isoforms of CBFβ contribute to Vif stabilization, whether CBFβ is required to stabilize Vif proteins of multiple different HIV subtypes, and, finally, if CBFβ is required by Vif to neutralize the entire repertoire of Vif-sensitive APOBEC3 proteins. Alternative splicing generates at least two isoforms of CBFβ in human cells (Genbank accessions NM_022845.2 and NM_001755.2). Though they differ in size and amino acid sequence at their C-terminal end, these splice variants share 165 N- terminal residues, including the RUNX heterodimerization domain, and a clear functional difference has yet to be delineated. To determine if HIV-1 Vif distinguishes between these CBFβ isoforms, a stable CBFβ knockdown clone of 3

78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 HEK293T was created using a stably integrated shrna that targets both isoforms (6). This line was transiently transfected with a Vif-proficient or Vif-deficient A200C HIV-1 IIIB molecular clone (3) in the presence or absence of human A3G and complemented with either the 187 amino acid CBFβ isoform 1 (cloned from CEM cdna by PCR and standard molecular biology techniques) or the shorter 182 amino acid CBFβ isoform 2 [as used previously; (6)]. 48 hours after transient transfection, cell lysates and viral particles were collected for immunoblotting and viral infectivity was monitored by infection of the reporter cell line CEM-GFP (5). Both isoforms resulted in a comparable increase in HIV-1 IIIB Vif steady-state levels, enhanced degradation of A3G, and a rescue of viral infectivity (Figure 1). In the absence of A3G, neither CBFβ isoform impacted viral infectivity. Most laboratory strains of HIV-1, including HIV-1 IIIB, HIV-1 NL4-3, and HIV-1 LAI, are subtype B, but over ten different HIV-1 subtypes are found worldwide with subtype C being the most prevalent (4). To determine if CBFβ can stabilize Vif proteins from multiple subtypes, representative Vif alleles from HIV-1 subtypes A, B, C, D, AE, F, and G [as described (2)] were co-transfected into the HEK293T CBFβ knockdown cell line with A3G in the presence or absence of CBFβ isoform 2 and GST as a transfection control. In every case, CBFβ increased the steady-state level of the Vif variant and resulted in increased degradation of A3G (Figure 2). While basal Vif expression levels varied, CBFβ increased the steady-state level of each Vif variant by an average of approximately 4-fold. Furthermore, while each variant also differs in its ability to neutralize A3G (2), steady-state levels of A3G were decreased upon CBFβ complementation in every case by an average of 2-fold. A3G levels were not 4

101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 affected by CBFβ in the absence of Vif or in the presence of HIV-1 IIIB Vif C133S, which fails to recruit the E3 ubiquitin ligase complex (7, 8). CBFβ did not effect the expression of the GST control. Thus, the dependency of Vif for CBFβ is broadly conserved across multiple HIV-1 subtypes. Human CD4 + T cells express six APOBEC3 proteins, of which HIV-1 Vif degrades five: A3C, A3D, A3F, A3G, and A3H (5, 10). To determine if HIV-1 Vif requires CBFβ to neutralize not only A3G, but the other Vif-sensitive APOBEC3 proteins as well, HIV-1 LAI molecular clone was transfected into HEK293T CBFβ knockdown cells with increasing amounts of each human APOBEC3 protein in the presence or absence of CBFβ isoform 2. CBFβ increased Vif steady-state levels and resulted in decreased cellular levels of all Vif-sensitive APOBEC3 proteins (A3C, A3D, A3F, A3G, and A3H haplotype II, Figure 3). In the presence of CBFβ, packaging of A3D, A3F, A3G, and A3H was also decreased and viral infectivity increased accordingly. Neither A3A nor A3B are sensitive to HIV-1 LAI Vif and consequently their expression, packaging, and impact on viral infectivity were not affected by CBFβ. Thus, HIV-1 Vif requires CBFβ to neutralize not only A3G, but the entire repertoire of Vif-sensitive human APOBEC3 proteins. Rhesus macaques also encode seven distinct APOBEC3 proteins, of which rhesus A3D, A3F, A3G, and A3H can restrict Vif-deficient HIV-1 and SIV (5, 11). SIV mac239 Vif neutralizes all four restrictive rhesus APOBEC3 proteins and also degrades rhesus A3B and rhesus A3C (5). To determine if SIV mac239 Vif requires CBFβ to neutralize the rhesus APOBEC3 proteins, Vif-deficient HIV-1 IIIB molecular clone was transfected into HEK293T CBFβ knockdown cells alongside SIV mac239 Vif 5

124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 and increasing amounts of each rhesus APOBEC3 protein in the presence or absence of CBFβ isoform 2. Human CBFβ isoforms 1 and 2 are identical at the amino acid level to rhesus CBFβ isoforms 1 and 2, respectively. While there is no antibody for SIVmac239 Vif, the addition of CBFβ resulted in decreased cellular levels of all Vifsensitive rhesus APOBEC3 proteins (rhesus A3B, A3C, A3D, A3F, A3G, and A3H, Figure 4). In the presence of CBFβ, packaging of rhesus A3D, A3F, A3G, and A3H was also decreased and viral infectivity consequently increased. Rhesus A3A is not sensitive to SIV mac239 Vif and so its expression, packaging, and effect on viral infectivity were unaltered by CBFβ. Thus, SIV Vif demonstrates a conserved requirement for CBFβ to neutralize the rhesus repertoire of APOBEC3 proteins. HIV-1 IIIB Vif was previously shown to require CBFβ isoform 2 for stable expression and neutralization of A3G (6). Here, we demonstrate that both CBFβ isoform 1 and isoform 2 may be hijacked to stabilize HIV-1 IIIB Vif and degrade A3G. This functional interaction was conserved across all tested HIV-1 subtypes and was required for the neutralization of not only A3G, but all Vif-sensitive human APOBEC3 proteins. SIV mac239 Vif also required CBFβ to neutralize all restrictive rhesus APOBEC3 proteins. Taken together, the CBFβ-Vif interaction appears to be broadly conserved and essential for Vif function, implicating this interface as a candidate for disruption by small molecule therapeutics that would alleviate repression of multiple restrictive APOBEC3 proteins. 144 145 6

146 147 148 149 150 151 We thank N. Krogan and J. Gross for discussion and data sharing prior to publication, and the NIH AIDS Research and Reference Reagent Program for materials. This research was funded by NIH R01 AI064046 and P01 GM091743 to RSH and NIH R01 AI064001 and NIH R01 AI089246 to VS. JFH was supported by an NSF Predoctoral Fellowship. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 REFERENCES 1. Albin, J. S., and R. S. Harris. 2010. Interactions of host APOBEC3 restriction factors with HIV-1 in vivo: implications for therapeutics. Expert Rev Mol Med 12:e4. 2. Binka, M., M. Ooms, M. Steward, and V. Simon. 2011. The activity spectrum of Vif from multiple HIV-1 Subtypes against APOBEC3G, APOBEC3F and APOBEC3H. J Virol. 3. Haché, G., T. E. Abbink, B. Berkhout, and R. S. Harris. 2009. Optimal translation initiation enables Vif-deficient human immunodeficiency virus type 1 to escape restriction by APOBEC3G. J Virol 83:5956-60. 4. Hemelaar, J., E. Gouws, P. D. Ghys, and S. Osmanov. 2011. Global trends in molecular epidemiology of HIV-1 during 2000-2007. AIDS 25:679-89. 5. Hultquist, J. F., J. A. Lengyel, E. W. Refsland, R. S. LaRue, L. Lackey, W. L. Brown, and R. S. Harris. 2011. Human and rhesus APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H demonstrate a conserved capacity to restrict Vifdeficient HIV-1. J Virol 85:11220-34. 7

169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 6. Jäger, S., D. Kim, J. Hultquist, K. Shindo, R. LaRue, E. Kwon, M. Li, B. Anderson, L. Yen, D. Stanley, C. Mahon, J. Kane, K. Franks-Skiba, P. Cimermancic, A. Burlingame, A. Sali, C. Craik, R. Harris, J. Gross, and N. Krogan. 2011. Vif Hijacks CBFβ to Degrade APOBEC3G and Promote HIV-1 Infection. Nature In Press. 7. Kobayashi, M., A. Takaori-Kondo, Y. Miyauchi, K. Iwai, and T. Uchiyama. 2005. Ubiquitination of APOBEC3G by an HIV-1 Vif-Cullin5-Elongin B-Elongin C complex is essential for Vif function. J Biol Chem 280:18573-8. 8. Luo, K., Z. Xiao, E. Ehrlich, Y. Yu, B. Liu, S. Zheng, and X. F. Yu. 2005. Primate lentiviral virion infectivity factors are substrate receptors that assemble with cullin 5-E3 ligase through a HCCH motif to suppress APOBEC3G. Proc Natl Acad Sci U S A 102:11444-9. 9. Malim, M. H., and M. Emerman. 2008. HIV-1 accessory proteins--ensuring viral survival in a hostile environment. Cell Host Microbe 3:388-98. 10. Refsland, E. W., M. D. Stenglein, K. Shindo, J. S. Albin, W. L. Brown, and R. S. Harris. 2010. Quantitative profiling of the full APOBEC3 mrna repertoire in lymphocytes and tissues: implications for HIV-1 restriction. Nucleic Acids Res 38:4274-84. 11. Virgen, C. A., and T. Hatziioannou. 2007. Antiretroviral activity and Vif sensitivity of rhesus macaque APOBEC3 proteins. J Virol 81:13932-7. 12. Wolf, D., and S. P. Goff. 2008. Host restriction factors blocking retroviral replication. Annu Rev Genet 42:143-63. 8

191 192 193 13. Yu, X., Y. Yu, B. Liu, K. Luo, W. Kong, P. Mao, and X. F. Yu. 2003. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science 302:1056-60. 194 195 196 197 198 199 200 201 202 203 204 205 206 FIGURE LEGENDS Figure 1 CBFβ Isoform 1 and Isoform 2 Stabilize HIV-1 Vif to Degrade A3G and Increase Viral Infectivity. (A) Percent infectivity of HIV-1 IIIB measured by duplicate infection of CEM-GFP and flow cytometry, reported as the mean +/- the standard deviation of the technical replicate. A constant amount of Vif-deficient or Vif-proficient A200C HIV-1 IIIB molecular clone (1 μg) was co-transfected with A3G or empty plasmid (50 ng) in the presence or absence of CBFβ complementation vector (25 ng) as indicated. (B) Immunoblots of CBFβ, Vif, and HA-tagged human A3G in cell lysates (lower panels) and of A3G in HIV-1 particles produced by those cells (upper panels). Tubulin (TUB) and p24 (CA) served as cell and viral lysate loading controls. 207 208 209 210 211 212 213 Figure 2 CBFβ Stabilizes Vif Proteins from Multiple HIV-1 Subtypes. Immunoblots of HA-tagged CBFβ, HIV-1 Vif, and FLAG-tagged human A3G in cell lysates. Tubulin (TUB) and V5-tagged GST served as cell lysate loading and transfection controls respectively. A constant amount of the indicated Vif variants (pcrv1 expression vector, 50 ng) were co-transfected with A3G (300 ng), GST (200 ng), and either CBFβ isoform 2 (100 ng) or empty vector. The untagged Vif variants 9

214 215 216 217 218 219 were detected with a polyclonal rabbit anti-vif antibody (NIH Cat#2221). One representative experiment of three independent transfections is shown. The Vif ratio represents the average ratio of Vif in the presence versus the absence of CBFβ (relative to GST) over three experiments unless otherwise noted (*n=2, #n=6). The A3G ratio was calculated analogously. Quantification was performed using Image Gauge v4.0. 220 221 222 223 224 225 226 227 228 229 230 231 232 Figure 3 CBFβ Is Required for HIV-1 Vif to Degrade all Vif-sensitive Human APOBEC3 Proteins. (A) Percent infectivity of HIV-1 LAI measured by duplicate infection of CEM-GFP and flow cytometry, reported as the mean +/- the standard deviation of the technical replicate. A constant amount of Vif-proficient HIV-1 LAI proviral construct (1 μg) was co-transfected with an increasing concentration of each human HA-tagged APOBEC3 protein (0, 50, or 100 ng) in the presence or absence of CBFβ isoform 2 complementation vector (25 ng) as indicated. (B) Immunoblots of CBFβ, Vif, and the HA-tagged human APOBEC3 proteins in cell lysates (lower panels) and of the APOBEC3 proteins in HIV virus particles produced by those cells (upper panels). Tubulin (TUB) and p24 (CA) served as cell and viral lysate loading controls. 233 234 235 Figure 4 CBFβ Is Required for SIV Vif to Degrade all Vif-sensitive Rhesus APOBEC3 Proteins. 10

236 237 238 239 240 241 242 243 244 245 246 (A) Percent infectivity of Vif-deficient HIV-1 IIIB supplemented with SIV mac239 Vif measured by duplicate infection of CEM-GFP and flow cytometry, reported as the mean +/- the standard deviation of the technical replicate. A constant amount of Vifdeficient A200C HIV-1 IIIB proviral construct (1 μg) was co-transfected with untagged SIV mac239 Vif (pvr1012 expression vector, 50 ng) and an increasing concentration of each rhesus, HA-tagged APOBEC3 protein (0, 50, or 100 ng) in the presence or absence of CBFβ isoform 2 complementation vector (25 ng) as indicated. (B) Immunoblots of CBFβ, Vif, and the HA-tagged rhesus APOBEC3 proteins in cell lysates (lower panels) and of the rhesus APOBEC3 proteins in HIV virus particles produced by those cells (upper panels). Tubulin (TUB) and p24 (CA) served as cell and viral lysate loading controls. 11