Regulation of CHMP4/ESCRT-III Function in Human ...

11
Regulation of CHMP4/ESCRT-III Function in Human Immunodeficiency Virus Type 1 Budding by CC2D1A Yoshiko Usami, a Sergei Popov, a Eric R. Weiss, a Christie Vriesema-Magnuson, a Arianna Calistri, b * and Heinrich G. Göttlinger a,b Program in Gene Function and Expression, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA, a and Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA b The detachment of human immunodeficiency type 1 (HIV-1) virions depends on CHPM4 family members, which are late-acting components of the ESCRT pathway that mediate the cleavage of bud necks from the cytosolic side. We now show that in human cells, CHMP4 proteins are to a considerable extent bound to two high-molecular-weight proteins that we have identified as CC2D1A and CC2D1B. Both proteins bind to the core domain of CHMP4B, which has a strong propensity to polymerize and to inhibit HIV-1 budding. Further mapping showed that CC2D1A binds to an N-terminal hairpin within the CHMP4 core that has been implicated in polymerization. Consistent with a model in which CC2D1A and CC2D1B regulate CHMP4 polymerization, the overexpression of CC2D1A inhibited both the release of wild-type HIV-1 and the CHMP4-dependent rescue of an HIV-1 L domain mutant by exogenous ALIX. Furthermore, small interfering RNA against CC2D1A or CC2D1B increased HIV-1 budding under certain conditions. CC2D1A and CC2D1B possess four Drosophila melanogaster 14 (DM14) domains, and we demon- strate that these constitute novel CHMP4 binding modules. The DM14 domain that bound most avidly to CHMP4B was by itself sufficient to inhibit the function of ALIX in HIV-1 budding, indicating that the inhibition occurred through CHMP4 sequestra- tion. However, N-terminal fragments of CC2D1A that did not interact with CHMP4B nevertheless retained a significant level of inhibitory activity. Thus, CC2D1A may also affect HIV-1 budding in a CHMP4-independent manner. R etroviruses hijack components of the host cell’s endosomal sorting complex required for transport (ESCRT) pathway via so-called late-assembly (L) domains in Gag to promote the de- tachment of nascent virions from the cell surface and from each other (3, 9, 14, 32, 54). The ESCRT pathway was discovered based on its requirement for the budding of cellular vesicles from the limiting membrane of multivesicular bodies (MVBs) into their lumen, which occurs away from the cytosol and thus resembles retroviral budding from the plasma membrane (21, 45). The com- ponents of the ESCRT pathway are highly conserved throughout eukaryotic evolution, and most of these components participate in the formation of five heterooligomeric complexes known as the ESCRT-0 to ESCRT-III and VPS4 complexes (22, 45). During MVB biogenesis, ESCRT-I and -II induce bud formation, and ESCRT-III, in concert with VPS4, carries out the scission of bud necks from the cytosolic side (55). ESCRT-III also carries out the scission of the membrane neck that forms between dividing cells during cytokinesis (4, 5, 38). In contrast to the other ESCRT complexes, ESCRT-III is not a stable complex of a defined composition. Rather, ESCRT-III po- lymerizes on membranes in a highly regulated manner from mo- nomeric cytosolic subunits (21). Humans encode at least 12 po- tential ESCRT-III subunits, most of which belong to seven charged MVB protein (CHMP) families (22). Six of these families also have a single member each in the yeast Saccharomyces cerevi- siae, and four of these are considered core ESCRT-III subunits based on their essential role in yeast MVB biogenesis (22). The CHMP proteins share an N-terminal core domain that folds into a four-helix bundle dominated by a hairpin formed by the first two helices (1, 40). Downstream of the core domain is an autoinhibi- tory region that is required to keep CHMP proteins in a closed soluble conformation (1, 29, 40, 47, 57). Human immunodeficiency virus type 1 (HIV-1) recruits the ESCRT-III scission complex and VPS4 transiently to sites of virus assembly (2, 24). The recruitment of ESCRT-III occurs indirectly via a PTAP-type L domain in HIV-1 Gag that binds to Tsg101, a subunit of ESCRT-I (10, 17, 34, 52). However, how this ultimately triggers ESCRT-III assembly is not clear, because ESCRT-I cou- ples to ESCRT-II, which is dispensable for HIV-1 budding (28). In addition to ESCRT-I, HIV-1 Gag engages an accessory compo- nent of the ESCRT pathway called ALIX, which binds directly to the ESCRT-III subunits CHMP4A to CHMP4C (11, 13, 15, 31, 33, 36, 43, 48, 53, 58). ALIX is less important for HIV-1 budding than ESCRT-I, but exogenous ALIX nonetheless potently rescues the budding defect of HIV-1 PTAP L domain mutants in a CHMP4- dependent manner (13, 51). Most if not all CHMP proteins polymerize and become potent inhibitors of HIV-1 budding if their autoinhibition is released (47, 57). Nevertheless, only the CHMP2 and CHMP4 families are re- quired for HIV-1 budding (39). When overexpressed, CHMP4 proteins have a propensity to form membrane-attached circular filaments that can promote outward budding (20). Interestingly, the single yeast homologue of human CHMP4A to CHMP4C (called Snf7) is the most crucial core ESCRT-III component for membrane scission (56). Snf7 is also the most abundant ESCRT- III component, and the other core ESCRT-III subunits are thought to regulate Snf7/CHMP4 polymerization (49). Certain ESCRT-III subunits assemble into dome-like structures in vitro, Received 12 October 2011 Accepted 5 January 2012 Published ahead of print 18 January 2012 Address correspondence to Heinrich G. Göttlinger, heinrich [email protected]. * Present address: University of Padova, Padua, Italy. Copyright © 2012, American Society for Microbiology. All Rights Reserved. doi:10.1128/JVI.06539-11 3746 jvi.asm.org 0022-538X/12/$12.00 Journal of Virology p. 3746 –3756 on February 13, 2018 by guest http://jvi.asm.org/ Downloaded from

Transcript of Regulation of CHMP4/ESCRT-III Function in Human ...

Page 1: Regulation of CHMP4/ESCRT-III Function in Human ...

Regulation of CHMP4/ESCRT-III Function in HumanImmunodeficiency Virus Type 1 Budding by CC2D1A

Yoshiko Usami,a Sergei Popov,a Eric R. Weiss,a Christie Vriesema-Magnuson,a Arianna Calistri,b* and Heinrich G. Göttlingera,b

Program in Gene Function and Expression, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA,a and Departmentof Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USAb

The detachment of human immunodeficiency type 1 (HIV-1) virions depends on CHPM4 family members, which are late-actingcomponents of the ESCRT pathway that mediate the cleavage of bud necks from the cytosolic side. We now show that in humancells, CHMP4 proteins are to a considerable extent bound to two high-molecular-weight proteins that we have identified asCC2D1A and CC2D1B. Both proteins bind to the core domain of CHMP4B, which has a strong propensity to polymerize and toinhibit HIV-1 budding. Further mapping showed that CC2D1A binds to an N-terminal hairpin within the CHMP4 core that hasbeen implicated in polymerization. Consistent with a model in which CC2D1A and CC2D1B regulate CHMP4 polymerization,the overexpression of CC2D1A inhibited both the release of wild-type HIV-1 and the CHMP4-dependent rescue of an HIV-1 Ldomain mutant by exogenous ALIX. Furthermore, small interfering RNA against CC2D1A or CC2D1B increased HIV-1 buddingunder certain conditions. CC2D1A and CC2D1B possess four Drosophila melanogaster 14 (DM14) domains, and we demon-strate that these constitute novel CHMP4 binding modules. The DM14 domain that bound most avidly to CHMP4B was by itselfsufficient to inhibit the function of ALIX in HIV-1 budding, indicating that the inhibition occurred through CHMP4 sequestra-tion. However, N-terminal fragments of CC2D1A that did not interact with CHMP4B nevertheless retained a significant level ofinhibitory activity. Thus, CC2D1A may also affect HIV-1 budding in a CHMP4-independent manner.

Retroviruses hijack components of the host cell’s endosomalsorting complex required for transport (ESCRT) pathway via

so-called late-assembly (L) domains in Gag to promote the de-tachment of nascent virions from the cell surface and from eachother (3, 9, 14, 32, 54). The ESCRT pathway was discovered basedon its requirement for the budding of cellular vesicles from thelimiting membrane of multivesicular bodies (MVBs) into theirlumen, which occurs away from the cytosol and thus resemblesretroviral budding from the plasma membrane (21, 45). The com-ponents of the ESCRT pathway are highly conserved throughouteukaryotic evolution, and most of these components participatein the formation of five heterooligomeric complexes known as theESCRT-0 to ESCRT-III and VPS4 complexes (22, 45). DuringMVB biogenesis, ESCRT-I and -II induce bud formation, andESCRT-III, in concert with VPS4, carries out the scission of budnecks from the cytosolic side (55). ESCRT-III also carries out thescission of the membrane neck that forms between dividing cellsduring cytokinesis (4, 5, 38).

In contrast to the other ESCRT complexes, ESCRT-III is not astable complex of a defined composition. Rather, ESCRT-III po-lymerizes on membranes in a highly regulated manner from mo-nomeric cytosolic subunits (21). Humans encode at least 12 po-tential ESCRT-III subunits, most of which belong to sevencharged MVB protein (CHMP) families (22). Six of these familiesalso have a single member each in the yeast Saccharomyces cerevi-siae, and four of these are considered core ESCRT-III subunitsbased on their essential role in yeast MVB biogenesis (22). TheCHMP proteins share an N-terminal core domain that folds into afour-helix bundle dominated by a hairpin formed by the first twohelices (1, 40). Downstream of the core domain is an autoinhibi-tory region that is required to keep CHMP proteins in a closedsoluble conformation (1, 29, 40, 47, 57).

Human immunodeficiency virus type 1 (HIV-1) recruits theESCRT-III scission complex and VPS4 transiently to sites of virus

assembly (2, 24). The recruitment of ESCRT-III occurs indirectlyvia a PTAP-type L domain in HIV-1 Gag that binds to Tsg101, asubunit of ESCRT-I (10, 17, 34, 52). However, how this ultimatelytriggers ESCRT-III assembly is not clear, because ESCRT-I cou-ples to ESCRT-II, which is dispensable for HIV-1 budding (28). Inaddition to ESCRT-I, HIV-1 Gag engages an accessory compo-nent of the ESCRT pathway called ALIX, which binds directly tothe ESCRT-III subunits CHMP4A to CHMP4C (11, 13, 15, 31, 33,36, 43, 48, 53, 58). ALIX is less important for HIV-1 budding thanESCRT-I, but exogenous ALIX nonetheless potently rescues thebudding defect of HIV-1 PTAP L domain mutants in a CHMP4-dependent manner (13, 51).

Most if not all CHMP proteins polymerize and become potentinhibitors of HIV-1 budding if their autoinhibition is released (47,57). Nevertheless, only the CHMP2 and CHMP4 families are re-quired for HIV-1 budding (39). When overexpressed, CHMP4proteins have a propensity to form membrane-attached circularfilaments that can promote outward budding (20). Interestingly,the single yeast homologue of human CHMP4A to CHMP4C(called Snf7) is the most crucial core ESCRT-III component formembrane scission (56). Snf7 is also the most abundant ESCRT-III component, and the other core ESCRT-III subunits arethought to regulate Snf7/CHMP4 polymerization (49). CertainESCRT-III subunits assemble into dome-like structures in vitro,

Received 12 October 2011 Accepted 5 January 2012

Published ahead of print 18 January 2012

Address correspondence to Heinrich G. Göttlinger, [email protected].

* Present address: University of Padova, Padua, Italy.

Copyright © 2012, American Society for Microbiology. All Rights Reserved.

doi:10.1128/JVI.06539-11

3746 jvi.asm.org 0022-538X/12/$12.00 Journal of Virology p. 3746–3756

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 2: Regulation of CHMP4/ESCRT-III Function in Human ...

and dome-shaped CHMP4 polymers may thus drive HIV-1 bud-ding (12, 30). Consistent with this model, ringlike structures po-tentially formed by CHMP4 were frequently detected within thenecks of arrested HIV-1 buds on the surface of cells depleted ofCHMP2 proteins (39).

In the present study, we observed that in human cells, CHMP4family members are complexed with two high-molecular-weightproteins, and microsequencing revealed that these proteins areCC2D1A and CC2D1B. In contrast to other CHMP4 bindingpartners, CC2D1A and CC2D1B interact with a CHMP4B coredomain fragment that blocks HIV-1 budding. Specifically,CC2D1A binds to an N-terminal helical hairpin that is thought tobe involved in CHMP4 polymerization. When overexpressed,CC2D1A inhibited HIV-1 budding and abolished the ability ofALIX to rescue a Tsg101 binding-site mutant. Taken together, ourresults suggest a model in which CC2D1A regulates CHMP4polymerization and determines the size of the pool that is availableto function in HIV-1 budding.

MATERIALS AND METHODSRetroviral vectors and stable cell lines. The coding sequences for full-length CHMP2, CHMP3, CHMP4A to CHMP4C, and CHMP6 with aC-terminal FLAG tag were inserted between the XhoI and EcoRI sites ofthe retroviral vector pMSCVpuro (Clontech). Additionally, the codingsequences for all CHMP proteins except CHMP7 were inserted betweenthe XhoI and NotI sites of the pOZ-FH-N or pOZ-FH-C retroviral vector(42), in each case in frame with either N-terminal or C-terminal FLAGand hemagglutinin (HA) tags encoded by the vector. Stable cell lines wereobtained by retroviral transduction followed by selection with puromycinor, in the case of the pOZ-based vectors, by multiple rounds of magnetic

affinity sorting with an anti-interleukin-2 receptor alpha antibody as de-scribed previously (42).

Mammalian expression vectors. The pBJ5-based expression vectorsfor CHMP4B-FLAG, FLAG-tagged CHMP4B1-153 (CHMP4B1-153FLAG),and HA-ALIX were described previously (48, 57). DNAs encodingCHMP4B residues 1 to 81, 1 to 92, 1 to 100, 1 to 108, 1 to 124, and 1 to 153with an N-terminal FLAG tag were amplified from cDNA clone BC011429(Open Biosystems, Huntsville, AL) and cloned into pBJ5. Additionally,DNAs encoding CHMP4B residues 19 to 224, 31 to 224, 33 to 224, 35 to224, 37 to 224, 39 to 224, and 41 to 224 with a C-terminal FLAG tag wereamplified from CHMP4B-FLAG and cloned into pBJ5. The coding se-quence for full-length CC2D1A with an N-terminal HA tag was amplifiedfrom BC064981 (Open Biosystems) and cloned into pBJ5. The resultingconstruct was used as a template to amplify DNAs encoding CC2D1Aresidues 1 to 141, 1 to 181, 1 to 203, 1 to 350, 1 to 494, 1 to 554, 1 to 655,and 1 to 759 with an N-terminal HA tag, which were all cloned into pBJ5.A vector expressing full-length CC2D1A with a C-terminal HA tag wasassembled from clone 023N (35) and BC006556 (Open Biosystems), andthe coding sequence for full-length CC2D1B with an N-terminal HA tagwas assembled from BQ073406 (Open Biosystems) and KIAA1836 (Ka-zusa DNA Research Institute) and cloned into pBJ5. For the mammalianexpression of glutathione S-transferase (GST)-tagged proteins, PCR frag-ments encompassing CC2D1A residues 1 to 141, 1 to 181, 1 to 203, 135 to198 (DM14 domain 1 [DM14/1]), 255 to 318 (DM14/2), 347 to 412(DM14/3), and 492 to 556 (DM14/4) were inserted in frame between theEcoRI and XhoI sites of pCAGGS/GST (33).

Immunoaffinity purification and identification of CHMP proteininteraction partners. Cells stably expressing FLAG- or FLAG/HA-taggedCHMP proteins were lysed in 0.5% Nonidet P-40 (NP-40) in phosphate-buffered saline. Clarified lysates were incubated with anti-FLAG M2antibody-conjugated agarose (Sigma-Aldrich), followed by extensivewashing and elution of bound proteins with 300 �g/ml FLAG peptide, as

FIG 1 CHMP4B interacts with CC2D1A and CC2D1B in vivo. (A) CHMP4B specifically interacts with two high-molecular-weight proteins in human cells. HeLa S3cells stably expressing FLAG-tagged CHMP6, CHMP4B, or CHMP1B were lysed, and proteins precipitated with anti-FLAG antibody were eluted with FLAG peptide,separated by SDS-PAGE, and stained with colloidal Coomassie. Microsequencing revealed that p115 and p130 are CC2D1A and CC2D1B, respectively. Two proteins thatcoprecipitated with CHMP1B-FLAG were identified as LIP5 and CHMP5. (B) CHMP4B does not induce the expression of p115/CC2D1A. The lysates of HeLa S3 cellsstably transduced with a retroviral vector expressing CHMP4B-FLAG or the empty vector were analyzed by Western blotting (WB) with antibodies against CC2D1A andactin. (C) Immunoprecipitations (IP) from 293T cells transiently expressing HA-tagged CC2D1A or CC2D1B together with full-length CHMP4B-FLAG.

CC2D1A Regulates CHMP4 Function in HIV Budding

April 2012 Volume 86 Number 7 jvi.asm.org 3747

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 3: Regulation of CHMP4/ESCRT-III Function in Human ...

described previously (42). Eluted proteins were separated by sodium do-decyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and stainedwith colloidal Coomassie. Specific bands were excised and subjected toin-gel tryptic digestion, and extracted peptides were analyzed on a matrix-assisted laser desorption ionization–time of flight mass spectrometer.

Coimmunoprecipitations. Expression vectors for FLAG- and HA-tagged proteins were cotransfected into 293T cells by using a calcium-phosphate precipitation technique. At 24 h posttransfection, the cells werelysed in NP-40 buffer (0.5% NP-40, 20 mM Tris HCl [pH 7.4], 150 mMNaCl, and protease inhibitor cocktail [Complete; Roche Molecular Bio-chemicals]). The lysates were centrifuged at 16,000 � g and immunopre-cipitated for 2.5 h at 4°C with anti-FLAG M2 antibody (Sigma-Aldrich).Immunoprecipitates and the cell lysates were analyzed by immunoblot-ting with anti-HA (HA.11; Covance) or anti-FLAG M2 antibody, as indi-cated.

GST pulldown assay. 293T cells were cotransfected with mammalianexpression vectors for GST- and either HA- or FLAG-tagged proteins.Twenty-four hours later, the cells were lysed in 0.5% NP-40 buffer, andclarified lysates were incubated with glutathione-Sepharose beads (GEHealthcare) for 2.5 h at 4°C. After extensive washing in NP-40 buffer,bound proteins were eluted by boiling in SDS-PAGE sample buffer andresolved by SDS-PAGE. Epitope-tagged proteins were detected by West-ern blotting with anti-HA or anti-FLAG M2 antibody, and GST fusionproteins were visualized with colloidal Coomassie brilliant blue G-250.

Analysis of viral particle production. 293T cells were cotransfectedwith HIV-1 proviral DNA together with vectors expressing FLAG- orHA-tagged proteins and, in some cases, with small interfering RNA(siRNA), as indicated. The cells were transfected with calcium-phosphate-precipitated DNA or, in cases where siRNA was cotransfected, with Lipo-fectamine 2000 (Invitrogen). The total amount of transfected DNA waskept constant with carrier DNA when calcium-phosphate precipitationwas used. The HIV-1 proviral plasmids used were the infectious molecularclone HXBH10 and a variant (�PTAPP) with an in-frame deletion thatremoves the binding site for Tsg101 (27). Previously described stealthsiRNA duplexes targeting CC2D1A (sense, CCCUGGCGAUCUGGAUGUCUUUGUU) (41) and CC2D1B (sense, CCCUGCAGCAGAGGCUGAACAAGUA) (19) and a matched stealth negative-control duplex (sense,

CCCAGCGGUCUGUAGUUCUUGUGUU) were purchased from Invit-rogen and used at 80 nM. At 24 h posttransfection, or 54 h posttransfec-tion if siRNA was cotransfected, the cells were lysed in radioimmunopre-cipitation assay buffer (140 mM NaCl, 8 mM Na2HPO4, 2 mM NaH2PO4,1% NP-40, 0.5% sodium deoxycholate, 0.05% SDS). Culture superna-tants were harvested from 6 to 24 h posttransfection, or from 48 to 54 hposttransfection if siRNA was used; clarified by low-speed centrifugation;and passaged through 0.45-�m filters. Virions released into the superna-tants were then pelleted through 20% sucrose cushions by ultracentrifu-gation for 2 h at 27,000 rpm at 4°C in a Beckman SW41 rotor. Pelletablematerial and the cell lysates were analyzed by SDS-PAGE and Westernblotting with anti-HIV CA antibody 183-H12-5C (7). The cell lysates werealso analyzed by Western blotting with anti-FLAG or anti-HA antibodyand, where indicated, with a rabbit anti-CC2D1A polyclonal antibody(Bethyl Laboratories) and anti-actin antibody AC-40 (Sigma-Aldrich).

RESULTSHuman CHMP4 proteins interact with CC2D1A and CC2D1Bin vivo. Among the ESCRT-III protein family members,CHMP4B is particularly important for HIV-1 virion release (39).To search for specific cellular binding partners, FLAG-tagged ver-sions of CHMP4B and of other human CHMP proteins were sta-bly expressed in 293T or HeLa S3 cells via retroviral transduction.The stable cell lines were lysed in 0.5% NP-40 buffer, and proteinsprecipitated from the lysates with anti-FLAG antibody were elutedwith FLAG peptide, separated by SDS-PAGE, and visualized byCoomassie staining. As shown in Fig. 1A, two proteins of approx-imately 115 and 130 kDa (provisionally designated p115 andp130) specifically coprecipitated with CHMP4B but not withCHMP1B or CHMP6. The intensities of the p115 and p130 bandswere comparable to that of the CHMP4B-FLAG band itself, sug-gesting that a considerable fraction of CHMP4B in human cells isbound to p115 and p130. The p115 and p130 bands also copre-cipitated with CHMP4A and to a lesser extent with CHMP4C but

FIG 2 CC2D1A and CC2D1B interact with the core domain of CHMP4B. Shown are immunoprecipitations from 293T cells transiently expressing HA-CC2D1A(A) or HA-CC2D1B (B) and FLAG-tagged full-length CHMP4B or its core domain (CHMP4153). Proteins precipitated with anti-FLAG antibody and the celllysates were analyzed by Western blotting (WB), as indicated. The pulldown of HA-CC2D1B was examined in duplicate.

Usami et al.

3748 jvi.asm.org Journal of Virology

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 4: Regulation of CHMP4/ESCRT-III Function in Human ...

not with CHMP1A, CHMP2A, CHMP2B, CHMP3, or CHMP5(data not shown).

Microsequencing of tryptic peptides derived from proteins co-precipitating with CHMP4B-FLAG unequivocally identified p115as coiled-coil- and C2 domain-containing protein 1A (CC2D1A)(64% coverage) and p130 as the related protein CC2D1B (67%coverage). Consistent with these results, CC2D1A previouslyscored positive as a potential binding partner for human CHMP4proteins in a yeast two-hybrid screen (50). Of note, Western blot-ting showed that the steady-state levels of endogenous CC2D1A incells stably expressing CHMP4B-FLAG were not higher than thosein control cells (Fig. 1B). We thus conclude that the remarkablyhigh pulldown levels particularly of CC2D1A relative to the levelsof CHMP4B-FLAG shown in Fig. 1A were not caused by an in-duction of CC2D1A expression by CHMP4B-FLAG.

CC2D1A and CC2D1B have calculated molecular masses of104 and 94 kDa, respectively. It was therefore unexpected thatCC2D1A (p115) exhibited a slower electrophoretic mobility thanCC2D1B (p130). To confirm that p115 and p130 are CC2D1A andCC2D1B, we coexpressed CHMP4B-FLAG in transiently trans-fected 293T cells together with HA-tagged versions of CC2D1A orCC2D1B. The transfected cells were then lysed in 0.5% NP-40buffer, and proteins precipitated from the lysates with anti-FLAGantibody were analyzed by Western blotting. As shown in Fig. 1C,

HA-CC2D1A and HA-CC2D1B both coprecipitated with CHMP4B-FLAG. Importantly, when HA-CC2D1A and HA-CC2D1B were ex-pressed alone, both were absent from anti-FLAG immunoprecipi-tates (Fig. 1C). As anticipated, HA-CC2D1B exhibited a slowerelectrophoretic mobility than HA-CC2D1A, confirming thatCC2D1B migrates aberrantly in SDS-PAGE gels.

CC2D1A and CC2D1B interact with a CHMP4B core frag-ment that blocks HIV-1 budding. The previously identifiedCHMP4 interaction partners VPS4 and ALIX bind to elementsnear or at the C terminus of CHMP4 family members (26, 36).Apart from providing docking sites, the C terminus of CHMP4has an autoinhibitory role (47, 57). Notably, the removal ofC-terminal autoinhibitory sequences triggers CHMP protein po-lymerization along with a potent anti-HIV-1 budding activity (47,57). For instance, we previously showed that HIV-1 release is pro-foundly inhibited by CHMP4B1–153FLAG, which lacks autoin-hibitory helix 5 and the VPS4 and ALIX binding sites (57).CHMP4B1–153FLAG corresponds to the asymmetric four-helixbundle that has been termed the CHMP protein core (57). There-fore, we used CHMP4B1–153FLAG to determine whether the in-teraction with CC2D1A is mediated by the core or the C-terminalregulatory region of CHMP4B. As shown in Fig. 2A, HA-CC2D1Aspecifically coprecipitated with both full-length CHMP4B-FLAGand C-terminally truncated CHMP4B1–153FLAG. Indeed, in sev-

FIG 3 Ability of N-terminal CHMP4B fragments to bind CC2D1A and to inhibit HIV-1 budding. (A) Schematic illustration of the N-terminal CHMP4Bfragments examined. Boxes indicate the predicted positions of �-helical regions in wild-type (WT) CHMP4B. (B) CC2D1A interacts with the N-terminal helicalhairpin of the CHMP4B core. 293T cells were cotransfected with vectors expressing FLAG-tagged CHMP4B truncation mutants and HA-CC2D1A. The celllysates and proteins immunoprecipitated with an anti-FLAG antibody were analyzed by Western blotting as indicated. (C) Effects on HIV-1 budding. 293T cellswere cotransfected with wild-type HIV-1 proviral DNA and vectors expressing the indicated CHMP4B truncation mutants. Virion production and Gag proteinexpression levels were compared by Western blotting with an anti-CA antibody, and the expression of the CHMP4B truncation mutants was examined byWestern blotting with an anti-FLAG antibody.

CC2D1A Regulates CHMP4 Function in HIV Budding

April 2012 Volume 86 Number 7 jvi.asm.org 3749

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 5: Regulation of CHMP4/ESCRT-III Function in Human ...

eral independent experiments, the amount of HA-CC2D1A recov-ered with the truncation mutant exceeded that obtained with full-lengthCHMP4B (Fig. 2A and data not shown). Similarly, HA-CC2D1B copre-cipitated at least as efficiently with CHMP4B1–153FLAG as with full-length CHMP4B (Fig. 2B). Thus, unlike VPS4 or ALIX, CC2D1A andCC2D1B bind to a CHMP4B core fragment that is capable of blockingHIV-1 budding.

CC2D1A binds to an N-terminal helical hairpin of CHMP4B.Members of the CHMP4 family are predicted to contain 6 helicalsegments, the first 5 of which are equivalent to helices in crystalstructures of CHMP3 (1, 40). As illustrated in Fig. 3A, CHMP4B1–153

retains predicted helices 1 through 4, which, by analogy withCHMP3, form the core of CHMP4B (39). To examine the roles ofindividual helical segments of the core in the interaction withCC2D1A, we further truncated CHMP4B1–153 from the C termi-nus (Fig. 3A). The analysis of a panel of truncation mutants re-vealed that the first 100 CHMP4B residues were fully sufficient forthe interaction with CC2D1A (Fig. 3B, lane 5). Within the CHMPprotein core, these residues form a prominent N-terminal helicalhairpin (40). Furthermore, CC2D1A coprecipitated equally wellwith CHMP4B1–92, indicating that the C terminus of the helicalhairpin is not required for the interaction (Fig. 3B, lane 6). How-ever, CC2D1A did not coprecipitate with CHMP4B1– 81, whichlacks almost half of the residues predicted to form helix 2 (Fig. 3B,lane 9).

It was suggested previously that the N-terminal helical hairpingoverns the ability of CHMP proteins to polymerize (1). Consis-tent with this notion, CHMP31–113, which essentially representsthe CHMP3 helical hairpin, inhibits HIV-1 budding even when

expressed at relatively low levels (57). However, it is not knownwhether the helical hairpin of CHMP4B shares this activity, andwe therefore examined whether CHMP4B1–100 affects HIV-1budding. Although CHMP4B1–100 did not share the effect ofCHMP4B1–153 on HIV-1 budding, CHMP4B1–100 caused someaccumulation of the Gag cleavage intermediates CA-p2 and p41(Fig. 3C, lane 3), a hallmark of late-assembly defects (17, 18).However, CHMP4B1-92, the shortest C-terminal truncation mu-tant that still interacted with CC2D1A, inhibited neither HIV-1release nor Gag processing (Fig. 3C, lane 4). Thus, the ability ofCHMP4B hairpin fragments to interact with CC2D1A is notsufficient for the dominant negative inhibition of the ESCRTpathway.

To determine the role of the N-terminal end of the helicalhairpin in the interaction with CC2D1A, we generated N-terminaltruncation mutants of CHMP4B-FLAG (Fig. 4A) and used thesemutants in coprecipitation experiments. CHMP4B31– end re-mained fully capable of interacting with CC2D1A (Fig. 4B, com-pare lanes 2 and 4), whereas CHMP4B41– end had lost this ability(lane 5). An analysis of additional truncation mutants revealedthat the N-terminal 34 residues of CHMP4B are entirely dispens-able for the interaction with CC2D1A (Fig. 4B, lane 8), whereasthe deletion of an additional 2 residues in CHMP4B37– end nearlyabolished the interaction (Fig. 4B, lane 9). We also examined theeffects of the N-terminal truncations on the ability of theCHMP4B core fragment CHMP4B1-153FLAG to inhibit HIV-1budding. CHMP4B19-153FLAG, which retained the entire helicalhairpin, remained capable of inhibiting HIV-1 Gag processingand budding (Fig. 4C, lane 3) albeit to a lesser extent than

FIG 4 Role of the CHMP4B N terminus. (A) Schematic illustration of N-terminal CHMP4B truncation mutants. (B) Effects of the truncations on the interactionbetween CHMP4B and CC2D1A, analyzed as described in the legend of Fig. 3B. (C) Effects of selected N-terminal truncations on the ability of the CHMP4B coredomain to inhibit HIV-1 budding and Gag processing, analyzed as described in the legend of Fig. 3C.

Usami et al.

3750 jvi.asm.org Journal of Virology

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 6: Regulation of CHMP4/ESCRT-III Function in Human ...

CHMP4B1-153FLAG (lane 2). However, all truncations which ex-tended into the region forming helix 1 abolished the inhibition ofHIV-1 Gag processing and budding by the CHMP4B core (Fig.4C, lane 4, and data not shown). Together, these data imply thatCC2D1A binds to a central portion of a helical hairpin, whoseintegrity is critical for the dominant negative inhibition of HIV-1budding by the CHMP4B core.

CC2D1A inhibits HIV-1 budding. A recent study demon-strated that CHMP4 proteins are transiently recruited late duringHIV-1 assembly, presumably to carry out the membrane fissionrequired for virion release (24). Consistent with this notion, thecodepletion of CHMP4 family members profoundly inhibits therelease of HIV-1 virions (39). Since our data indicated thatCHMP4A and CHMP4B bind avidly to CC2D1 proteins, we con-sidered the possibility that the expression levels of CC2D1A orCC2D1B affect the availability of CHMP4 family members duringHIV-1 budding. To test this possibility, we cotransfected 293Tcells with HXBH10, a replication-competent HIV-1 provirus, anddifferent amounts of a vector expressing CC2D1A-HA. As shownin Fig. 5A, CC2D1A inhibited the release of wild-type HIV-1 viri-ons in a dose-dependent manner, as monitored by the release ofparticle-associated CA into the medium. Furthermore, the over-expression of CC2D1A led to the accumulation of the Gag cleav-age intermediates CA-p2 and p41 in the transfected cells, as ex-pected if a late assembly step is defective (Fig. 5A).

We also examined whether exogenous CC2D1A affects thefunction of ALIX in virus budding. We and others previouslyshowed that exogenous ALIX efficiently rescues the profound re-lease defect of HIV-1 mutants that lack the PTAP-type primary L

domain (13, 51). ALIX selectively interacts with CHMP4 familymembers, particularly with CHMP4B (13, 25), and point muta-tions in the Bro1 domain of ALIX that eliminate the interactionwith CHMP4B abolish the rescue of �PTAPP HIV-1 by ALIX (13,51). Because these results indicated that ALIX must interact withCHMP4 proteins to function in HIV-1 budding, we determinedthe effect of exogenous CC2D1A in the �PTAPP rescue assay.Even at relatively low expression levels, CC2D1A-HA blocked theability of HA-ALIX to rescue �PTAPP HIV-1 in terms of bothparticle production and Gag processing (Fig. 5B, lane 3). Impor-tantly, CC2D1A-HA had no effect on the expression levels of HA-ALIX. Also, CC2D1A-HA did not reduce the low level of particleproduction by �PTAPP HIV-1 obtained in the absence of ALIX-HA, implying that CC2D1A specifically affected the function ofALIX (Fig. 5B, lane 4).

We next asked whether the depletion of CC2D1A or CC2D1Bincreases particle production under conditions where HIV-1 bud-ding is attenuated. To this end, we cotransfected 293T cells withthe �PTAPP mutant along with a small amount of the HA-ALIXexpression vector. When cotransfected with a control siRNA, thisamount was insufficient to trigger the release of mature CA (Fig.5C, lane 1). However, the cotransfection of siRNAs againstCC2D1A or CC2D1B stimulated the release of CA and reducedthe levels of cell-associated CA-p2 without affecting the expres-sion levels of HA-ALIX (Fig. 5C, lanes 2 and 3). As expected, onlythe siRNA against CC2D1A depleted the transfected cells ofCC2D1A protein (Fig. 5C). The effectiveness of the siRNA againstCC2D1B was previously demonstrated (19). Taken together, theseresults support the notion that CC2D1 proteins inhibit the ability

FIG 5 CC2D1A inhibits HIV-1 budding. (A) Effect of CC2D1A overexpression on wild-type HIV-1. 293T cells were cotransfected with wild-type HIV-1 proviralDNA (1 �g) and the indicated amounts of a vector expressing CC2D1A-HA. (B) Effect of CC2D1A overexpression on the rescue of HIV-1�PTAPP by ALIX. 293Tcells were cotransfected with �PTAPP HIV-1 proviral DNA (1 �g), together with pBJ5-based vectors expressing HA-ALIX (1 �g) or CC2D1A-HA (2 �g) or withempty pBJ5, as indicated. Virion pellets and the cell lysates were analyzed by Western blotting with anti-CA and anti-HA antibodies to detect Gag, Gag cleavageproducts, and HA-tagged proteins. (C) Effects of siRNAs against CC2D1A or CC2D1B on the release of �PTAPP HIV-1 in the presence of a suboptimal amountof HA-ALIX. 293T cells were cotransfected with �PTAPP HIV-1 proviral DNA (0.5 �g), together with the vector expressing HA-ALIX (0.2 �g) and the indicatedsiRNAs (80 nM). Pelleted virions harvested at 48 to 54 h posttransfection and the cell lysates were analyzed by Western blotting with anti-CA, anti-HA,anti-CC2D1A, or anti-actin antibodies, as indicated.

CC2D1A Regulates CHMP4 Function in HIV Budding

April 2012 Volume 86 Number 7 jvi.asm.org 3751

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 7: Regulation of CHMP4/ESCRT-III Function in Human ...

of CHMP4 family members to participate in HIV-1 buddingevents.

Role of DM14 domains in CHMP4 binding and in the inhibi-tion of HIV-1 budding. CC2D1A and CC2D1B harbor four Dro-sophila melanogaster 14 (DM14) domains that are unique to theseproteins and have no known function and a protein kinase Cconserved region 2 (C2) domain that follows upon the DM14domains (Fig. 6A). To determine the contribution of these do-mains and of intervening sequences to the interaction withCHMP4B, we generated a series of C-terminal truncation mutantsof CC2D1A. Taking into account their cellular expression levels,the CC2D1A truncation mutants encompassing residues 1 to 759,1 to 655, 1 to 554, and 1 to 494 appeared to coprecipitate equallywell with CHMP4B-FLAG, indicating that the region N terminalto the fourth DM14 domain was fully sufficient for the interactionin vivo (Fig. 6A, lanes 1 to 4). However, the mutant encompassingresidues 1 to 350, which retains only 2 DM14 domains, coprecipi-tated less efficiently (about 3- to 4-fold) relative to its expressionlevel (Fig. 6A, lane 5). Nevertheless, HA-CC2D1A1-350 remainedfully capable of blocking the effect of ALIX on HIV-1 budding inthe �PTAPP rescue assay (Fig. 6B). As expected from this result,the function of ALIX in HIV-1 budding was also blocked by themutants encompassing residues 1 to 759, 1 to 655, 1 to 554, and 1

to 494 (data not shown). Together, these findings suggested a rolefor DM14 domains in the interaction with CHMP4B and in theinhibition of ALIX function.

To determine directly whether DM14 domains bind CHMP4B,each of the four DM14 domains of CC2D1A was individually fused tothe C terminus of GST in the context of a mammalian expressionvector. We then expressed the GST-DM14 fusion proteins in 293Tcells together with HA-tagged CHMP4B and performed GST pull-down assays. Although the first DM14 domain of CC2D1A did notpull down any CHMP4B-HA when fused to GST, the isolated second,third, and fourth DM14 domains clearly interacted with CHMP4B inthe same context (Fig. 7A). We also tested the effects of the CHMP4B-binding fusion proteins in the �PTAPP rescue assay and found thatonly the construct harboring the third DM14 domain of CC2D1Ainhibited the rescue of �PTAPP HIV-1 by ALIX (Fig. 7B). As shownin Fig. 7A, the third DM14 domain also pulled down more CHMP4Bthan did DM14 domains 2 and 4. Thus, 3 of the 4 DM14 domains ofCC2D1A constitute autonomous CHMP4B binding modules, andthe DM14 domain that interacts most avidly with CHMP4B is byitself capable of inhibiting the function of ALIX in HIV-1 budding.

Since the first DM14 domain (CC2D1A residues 135 to 198)was unique in its inability to interact with CHMP4B, we examinedwhether DM14 domain 1 mediates CHMP4B binding in the pres-

FIG 6 Ability of CC2D1A truncation mutants to bind CHMP4B and to inhibit the function of ALIX in HIV-1 budding. (A) Ability of CC2D1A truncationmutants to interact with CHMP4B. The schematically illustrated HA-tagged CC2D1A mutants were expressed in 293T cells together with CHMP4B-FLAG. Thecell lysates and proteins immunoprecipitated with an anti-FLAG antibody were analyzed by Western blotting, as indicated. (B) Effect of CC2D1A1–350 on therescue of HIV-1�PTAPP by ALIX. 293T cells were cotransfected with �PTAPP HIV-1 proviral DNA (1 �g), together with pBJ5-based vectors expressing HA-ALIX(2 �g) or HA-CC2D1A1–350 (2 �g) or with empty pBJ5, as indicated. Virion pellets and the cell lysates were analyzed by Western blotting with anti-CA andanti-HA antibodies to detect Gag, Gag cleavage products, ALIX, and CC2D1A1–350.

Usami et al.

3752 jvi.asm.org Journal of Virology

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 8: Regulation of CHMP4/ESCRT-III Function in Human ...

ence of additional residues that flank the domain in the context ofintact CC2D1A. Indeed, a GST fusion protein that harbored anN-terminal CC2D1A fragment that extended slightly beyond thefirst DM14 domain (GST-CC2D1A1-203) interacted robustly withCHMP4B (Fig. 8A, lane 1). To determine whether an intact DM14domain was required for CHMP4B binding in this context, theDM14 domain was either truncated or deleted, yielding GST-CC2D1A1-181 and GST-CC2D1A1-141, respectively. Both modifi-cations abolished binding to CHMP4B (Fig. 8A, lanes 2 and 3),demonstrating that the interaction depended on the integrity ofthe DM14 domain. In the �PTAPP rescue assay, CHMP4binding-competent CC2D1A1-203 completely abolished the abilityof ALIX to promote HIV-1 budding (Fig. 8B, lane 3). However,despite their inability to interact with CHMP4B, CC2D1A1-181

and CC2D1A1-141 also inhibited the effect of ALIX on HIV-1 bud-ding albeit to a lesser extent than did CC2D1A1-203 (Fig. 8B, lanes4 and 5). Taken together, these results indicate that the inhibitionof HIV-1 budding by CC2D1A occurs both via the sequestrationof CHMP4 proteins by DM14 domains and via a CHMP4-independent mechanism.

DISCUSSION

Although most CHMP proteins can be converted into strongdominant negative inhibitors of HIV-1 particle production,siRNA depletion experiments have revealed that only the CHMP2and CHMP4 families are in fact required for HIV-1 budding (39).While the members of these two families exhibit redundancy,

CHMP4B appears particularly important, because its individualdepletion had the greatest effect on HIV-1 release (39).

In the present study, we provide evidence that a large fractionof cellular CHMP4B exists in a complex with high-molecular-weight proteins that we have identified as CC2D1A and CC2D1B.Both proteins additionally interacted with CHMP4A andCHMP4C in human cells but not with any other ESCRT-III com-ponent. CC2D1A is identical to Freud-1, which has been impli-cated in transcriptional regulation and in nonsyndromic mentalretardation (44, 59). Consistent with our findings, CC2D1A wasamong a large collection of potential CHMP protein auxiliary fac-tors that emerged from a yeast two-hybrid screen, in whichCC2D1A also interacted only with CHMP4 proteins (50). A con-nection to the ESCRT pathway is further implied by the pheno-type caused by mutations in the Drosophila protein Lethal giantdiscs (Lgd), the homologue of human CC2D1A and CC2D1B.Specifically, the absence of Lgd leads to the accumulation andectopic activation of Notch in an endosomal compartment, whichis also observed if the function of ESCRT components is lost, forinstance, that of the Drosophila CHMP4 homologue (8, 16, 23).Human CC2D1A is also known as Aki1, which has been shownto localize to centrosomes (41). Interestingly, the depletion ofCC2D1A/Aki1 results in the formation of multipolar spindlesduring mitosis (41), and cells depleted of CHMP4B or CHMP4Calso exhibit multipolar spindles (37).

Our results suggest that CC2D1A and CC2D1B control thepolymerization of CHMP4 filaments, which are thought to drive

FIG 7 CHMP4B binding by isolated DM14 domains and their effects on the function of ALIX in HIV-1 budding. (A) Ability of GST-DM14 domain fusionproteins to pull down CHMP4B. The schematically illustrated GST fusion proteins were expressed in 293T cells together with CHMP4B-HA, and proteinsprecipitated from the cell lysates by glutathione-Sepharose beads were analyzed by Western blotting with an anti-HA antibody or by Coomassie staining to detectthe GST-DM14 fusion proteins. (B) Effects of the GST-DM14 domain fusion proteins on the rescue of HIV-1�PTAPP by ALIX. 293T cells were cotransfected with�PTAPP HIV-1 proviral DNA, a vector expressing HA-ALIX, and vectors expressing GST or the indicated GST-DM14 domain fusion proteins. Virion pellets andthe cell lysates were analyzed by Western blotting with anti-CA and anti-HA antibodies to detect Gag, Gag cleavage products, and ALIX.

CC2D1A Regulates CHMP4 Function in HIV Budding

April 2012 Volume 86 Number 7 jvi.asm.org 3753

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 9: Regulation of CHMP4/ESCRT-III Function in Human ...

the scission of bud necks from the cytosolic side, for instance,during HIV-1 budding. When overexpressed, human CHMP4Aand CHMP4B assemble into membrane-attached circular fila-ments that can induce the outward budding of tubules at theplasma membrane (20). Functional reconstitution studies withpurified yeast ESCRT-III proteins support the notions thatCHMP4 family members self-assemble into oligomers that inducemembrane deformation and that the other ESCRT-III core com-ponents function in the nucleation or termination of CHMP4oligomerization (46). To control ESCRT-III formation, ESCRT-III components are by default kept in a closed conformationthrough an autoinhibitory mechanism (29, 40, 47, 57). However,the propensity of CHMP4 family members to polymerize mayrequire an additional level of regulation through proteins such asCC2D1A/B that bind to the region through which activatedCHMP4 self-associates. Consistent with this model, we find thatCC2D1A and CC2D1B strongly interact with a CHMP4B corefragment that is constitutively activated, based on its ability toprevent HIV-1 budding (57). Moreover, additional mapping re-vealed that CC2D1A binds to the helical hairpin that dominatesthe CHMP core domain and has been implicated in filament for-mation (1, 40). Further support for a model in which CC2D1Aregulates CHMP4 polymerization is provided by our finding thatexogenous CC2D1A inhibited the release of wild-type HIV-1 andthe rescue of �PTAPP HIV-1 by ALIX, both of which depend onCHMP4 (13, 39, 51).

CC2D1A and CC2D1B are the only human members of a pro-tein family harboring four DM14 domains and a C-terminal C2domain. It was shown previously that the C2 domain of the

CC2D1A homologue Lgd binds phospholipids and is required forthe membrane association of Lgd in vivo (16). However, the func-tion of the DM14 domains has remained unknown. We now showthat DM14 domains constitute CHMP4 binding modules that canfunction autonomously, as demonstrated by our observation thatthe second, third, and fourth DM14 domains of CC2D1A boundCHMP4B even in isolation. However, only the isolated thirdDM14 domain, which bound most avidly, was sufficient to blockthe function of ALIX in HIV-1 budding. Together, these resultssuggest that only the third DM14 domain retained a sufficientlyhigh affinity to block CHMP4 polymerization by itself. It appearsless likely that competition with the Bro1 domain of ALIX forCHMP4 binding played a role, because our results imply thatDM14 domains bind to the CHMP4 core. In contrast, ALIX bindsto the C terminus of CHMP4 family members (36). The firstDM14 domain did not bind CHMP4B but mediated CHMP4binding in the context of a larger N-terminal fragment. Surpris-ingly, the disruption or removal of the DM14 domain, which pre-vented CHMP4 binding, only mitigated but did not abolish theability of the N-terminal fragment to inhibit the function of ALIXin HIV-1 budding. It thus appears that CC2D1A inhibits the ac-tivity of ALIX not only via the sequestration of CHMP4 proteins.Consistent with this notion, we were unable to alleviate the inhib-itory effect of CC2D1A overexpression on the function of ALIXthrough the simultaneous overexpression of CHMP4B (data notshown).

In a recent study, CC2D1A was found to localize to endolyso-somal compartments in human cells (6), which would be consis-tent with a role in regulating the polymerization of CHMP4 pro-

FIG 8 DM14 domain 1-dependent CHMP4B binding and inhibition of HIV-1 budding by an N-terminal CC2D1A fragment. (A) DM14 domain 1 is requiredfor CHMP4B binding by an N-terminal CC2D1A fragment. The schematically illustrated GST fusion proteins were expressed in 293T cells together withCHMP4B-FLAG, and proteins precipitated from the cell lysates by glutathione-Sepharose beads were analyzed by Western blotting with anti-FLAG antibody orby Coomassie staining to detect the GST-DM14 fusion proteins. (B) Effects of the N-terminal CC2D1A fragments on the rescue of HIV-1�PTAPP by ALIX.

Usami et al.

3754 jvi.asm.org Journal of Virology

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 10: Regulation of CHMP4/ESCRT-III Function in Human ...

teins on endosomal membranes. That same study and earlier workalso indicated that CC2D1A activates signaling pathways involvedin innate immunity, including the NF-�B pathway (6, 35, 59).Notably, the overexpression of CC2D1A induced endogenousbeta interferon and led to an antiviral state (6). Our observationthat CC2D1A binds to CHMP4 proteins via its DM14 domainsthus raises the possibility that components of the ESCRT pathwayplay specific roles in innate antiviral defense mechanisms.

ACKNOWLEDGMENTS

We thank Paul Bieniasz for pCAGGS/GST, Yoshihiro Nakatani for pOZ-FH-N and pOZ-FH-C, Akio Matsuda for clone 023N, Tahakiro Nagaseand the Kazusa DNA Research Institute for KIAA1836, and Steven Gygiand John Leszyk for protein microsequencing. The following reagent wasobtained through the AIDS Research and Reference Reagent Program,Division of AIDS, NIAID, NIH: HIV-1 p24 monoclonal antibody (183-H12-5C), from Bruce Chesebro and Kathy Wehrly.

This work was supported by grant R37AI029873 from the NationalInstitute of Allergy and Infectious Diseases.

REFERENCES1. Bajorek M, et al. 2009. Structural basis for ESCRT-III protein autoinhi-

bition. Nat. Struct. Mol. Biol. 16:754 –762.2. Baumgartel V, et al. 2011. Live-cell visualization of dynamics of HIV

budding site interactions with an ESCRT component. Nat. Cell Biol. 13:469 – 474.

3. Bieniasz PD. 2009. The cell biology of HIV-1 virion genesis. Cell HostMicrobe 5:550 –558.

4. Caballe A, Martin-Serrano J. 2011. ESCRT machinery and cytokinesis:the road to daughter cell separation. Traffic 12:1318 –1326.

5. Carlton JG, Martin-Serrano J. 2007. Parallels between cytokinesis andretroviral budding: a role for the ESCRT machinery. Science 316:1908 –1912.

6. Chang CH, et al. 2011. TBK1-associated protein in endolysosomes(TAPE) is an innate immune regulator modulating the TLR3 and TLR4signaling pathways. J. Biol. Chem. 286:7043–7051.

7. Chesebro B, Wehrly K, Nishio J, Perryman S. 1992. Macrophage-tropichuman immunodeficiency virus isolates from different patients exhibitunusual V3 envelope sequence homogeneity in comparison with T-cell-tropic isolates: definition of critical amino acids involved in cell tropism. J.Virol. 66:6547– 6554.

8. Childress JL, Acar M, Tao C, Halder G. 2006. Lethal giant discs, a novelC2-domain protein, restricts notch activation during endocytosis. Curr.Biol. 16:2228 –2233.

9. Demirov DG, Freed EO. 2004. Retrovirus budding. Virus Res. 106:87–102.

10. Demirov DG, Ono A, Orenstein JM, Freed EO. 2002. Overexpressionof the N-terminal domain of TSG101 inhibits HIV-1 budding byblocking late domain function. Proc. Natl. Acad. Sci. U. S. A. 99:955–960.

11. Dussupt V, et al. 2009. The nucleocapsid region of HIV-1 Gag cooperateswith the PTAP and LYPXnL late domains to recruit the cellular machinerynecessary for viral budding. PLoS Pathog. 5:e1000339.

12. Fabrikant G, et al. 2009. Computational model of membrane fissioncatalyzed by ESCRT-III. PLoS Comput. Biol. 5:e1000575.

13. Fisher RD, et al. 2007. Structural and biochemical studies of ALIX/AIP1and its role in retrovirus budding. Cell 128:841– 852.

14. Fujii K, Hurley JH, Freed EO. 2007. Beyond Tsg101: the role of Alix in‘ESCRTing’ HIV-1. Nat. Rev. Microbiol. 5:912–916.

15. Fujii K, et al. 2009. Functional role of Alix in HIV-1 replication. Virology391:284 –292.

16. Gallagher CM, Knoblich JA. 2006. The conserved c2 domain proteinlethal (2) giant discs regulates protein trafficking in Drosophila. Dev. Cell11:641– 653.

17. Garrus JE, et al. 2001. Tsg101 and the vacuolar protein sorting pathwayare essential for HIV-1 budding. Cell 107:55– 65.

18. Gottlinger HG, Dorfman T, Sodroski JG, Haseltine WA. 1991. Effect ofmutations affecting the p6 gag protein on human immunodeficiency virusparticle release. Proc. Natl. Acad. Sci. U. S. A. 88:3195–3199.

19. Hadjighassem MR, Galaraga K, Albert PR. 2011. Freud-2/CC2D1Bmediates dual repression of the serotonin-1A receptor gene. Eur. J. Neu-rosci. 33:214 –223.

20. Hanson PI, Roth R, Lin Y, Heuser JE. 2008. Plasma membrane defor-mation by circular arrays of ESCRT-III protein filaments. J. Cell Biol.180:389 – 402.

21. Henne WM, Buchkovich NJ, Emr SD. 2011. The ESCRT pathway. Dev.Cell 21:77–91.

22. Hurley JH. 2010. The ESCRT complexes. Crit. Rev. Biochem. Mol. Biol.45:463– 487.

23. Jaekel R, Klein T. 2006. The Drosophila Notch inhibitor and tumorsuppressor gene lethal (2) giant discs encodes a conserved regulator ofendosomal trafficking. Dev. Cell 11:655– 669.

24. Jouvenet N, Zhadina M, Bieniasz PD, Simon SM. 2011. Dynamics ofESCRT protein recruitment during retroviral assembly. Nat. Cell Biol.13:394 – 401.

25. Katoh K, Shibata H, Hatta K, Maki M. 2004. CHMP4b is a major bindingpartner of the ALG-2-interacting protein Alix among the three CHMP4isoforms. Arch. Biochem. Biophys. 421:159 –165.

26. Kieffer C, et al. 2008. Two distinct modes of ESCRT-III recognition arerequired for VPS4 functions in lysosomal protein targeting and HIV-1budding. Dev. Cell 15:62–73.

27. Kondo E, Gottlinger HG. 1996. A conserved LXXLF sequence is themajor determinant in p6gag required for the incorporation of humanimmunodeficiency virus type 1 Vpr. J. Virol. 70:159 –164.

28. Langelier C, et al. 2006. Human ESCRT-II complex and its role in humanimmunodeficiency virus type 1 release. J. Virol. 80:9465–9480.

29. Lata S, et al. 2008. Structural basis for autoinhibition of ESCRT-IIICHMP3. J. Mol. Biol. 378:816 – 825.

30. Lata S, et al. 2008. Helical structures of ESCRT-III are disassembled byVPS4. Science 321:1354 –1357.

31. Lee S, Joshi A, Nagashima K, Freed EO, Hurley JH. 2007. Structuralbasis for viral late-domain binding to Alix. Nat. Struct. Mol. Biol. 14:194 –199.

32. Martin-Serrano J, Neil SJ. 2011. Host factors involved in retroviral bud-ding and release. Nat. Rev. Microbiol. 9:519 –531.

33. Martin-Serrano J, Yarovoy A, Perez-Caballero D, Bieniasz PD. 2003.Divergent retroviral late-budding domains recruit vacuolar protein sort-ing factors by using alternative adaptor proteins. Proc. Natl. Acad. Sci.U. S. A. 100:12414 –12419.

34. Martin-Serrano J, Zang T, Bieniasz PD. 2001. HIV-1 and Ebola virusencode small peptide motifs that recruit Tsg101 to sites of particle assem-bly to facilitate egress. Nat. Med. 7:1313–1319.

35. Matsuda A, et al. 2003. Large-scale identification and characterization ofhuman genes that activate NF-kappaB and MAPK signaling pathways.Oncogene 22:3307–3318.

36. McCullough J, Fisher RD, Whitby FG, Sundquist WI, Hill CP. 2008.ALIX-CHMP4 interactions in the human ESCRT pathway. Proc. Natl.Acad. Sci. U. S. A. 105:7687–7691.

37. Morita E, et al. 2010. Human ESCRT-III and VPS4 proteins are requiredfor centrosome and spindle maintenance. Proc. Natl. Acad. Sci. U. S. A.107:12889 –12894.

38. Morita E, et al. 2007. Human ESCRT and ALIX proteins interact withproteins of the midbody and function in cytokinesis. EMBO J. 26:4215–4227.

39. Morita E, et al. 2011. ESCRT-III protein requirements for HIV-1 bud-ding. Cell Host Microbe 9:235–242.

40. Muziol T, et al. 2006. Structural basis for budding by the ESCRT-IIIfactor CHMP3. Dev. Cell 10:821– 830.

41. Nakamura A, Arai H, Fujita N. 2009. Centrosomal Aki1 and cohesinfunction in separase-regulated centriole disengagement. J. Cell Biol. 187:607– 614.

42. Nakatani Y, Ogryzko V. 2003. Immunoaffinity purification of mamma-lian protein complexes. Methods Enzymol. 370:430 – 444.

43. Popov S, Popova E, Inoue M, Gottlinger HG. 2008. Human immuno-deficiency virus type 1 Gag engages the Bro1 domain of ALIX/AIP1through the nucleocapsid. J. Virol. 82:1389 –1398.

44. Rogaeva A, Galaraga K, Albert PR. 2007. The Freud-1/CC2D1A family:transcriptional regulators implicated in mental retardation. J. Neurosci.Res. 85:2833–2838.

45. Saksena S, Sun J, Chu T, Emr SD. 2007. ESCRTing proteins in theendocytic pathway. Trends Biochem. Sci. 32:561–573.

46. Saksena S, Wahlman J, Teis D, Johnson AE, Emr SD. 2009. Func-

CC2D1A Regulates CHMP4 Function in HIV Budding

April 2012 Volume 86 Number 7 jvi.asm.org 3755

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 11: Regulation of CHMP4/ESCRT-III Function in Human ...

tional reconstitution of ESCRT-III assembly and disassembly. Cell 136:97–109.

47. Shim S, Kimpler LA, Hanson PI. 2007. Structure/function analysis offour core ESCRT-III proteins reveals common regulatory role for extremeC-terminal domain. Traffic 8:1068 –1079.

48. Strack B, Calistri A, Craig S, Popova E, Gottlinger HG. 2003. AIP1/ALIXis a binding partner for HIV-1 p6 and EIAV p9 functioning in virus bud-ding. Cell 114:689 – 699.

49. Teis D, Saksena S, Emr SD. 2008. Ordered assembly of the ESCRT-IIIcomplex on endosomes is required to sequester cargo during MVB forma-tion. Dev. Cell 15:578 –589.

50. Tsang HT, et al. 2006. A systematic analysis of human CHMP proteininteractions: additional MIT domain-containing proteins bind to mul-tiple components of the human ESCRT III complex. Genomics 88:333–346.

51. Usami Y, Popov S, Gottlinger HG. 2007. Potent rescue of human im-munodeficiency virus type 1 late domain mutants by ALIX/AIP1 dependson its CHMP4 binding site. J. Virol. 81:6614 – 6622.

52. VerPlank L, et al. 2001. Tsg101, a homologue of ubiquitin-conjugating

(E2) enzymes, binds the L domain in HIV type 1 Pr55(Gag). Proc. Natl.Acad. Sci. U. S. A. 98:7724 –7729.

53. von Schwedler UK, et al. 2003. The protein network of HIV budding. Cell114:701–713.

54. Weiss ER, Gottlinger H. 2011. The role of cellular factors in promotingHIV budding. J. Mol. Biol. 410:525–533.

55. Wollert T, Hurley JH. 2010. Molecular mechanism of multivesicularbody biogenesis by ESCRT complexes. Nature 464:864 – 869.

56. Wollert T, Wunder C, Lippincott-Schwartz J, Hurley JH. 2009. Mem-brane scission by the ESCRT-III complex. Nature 458:172–177.

57. Zamborlini A, et al. 2006. Release of autoinhibition converts ESCRT-IIIcomponents into potent inhibitors of HIV-1 budding. Proc. Natl. Acad.Sci. U. S. A. 103:19140 –19145.

58. Zhai Q, et al. 2008. Structural and functional studies of ALIX interactionswith YPX(n)L late domains of HIV-1 and EIAV. Nat. Struct. Mol. Biol.15:43– 49.

59. Zhao M, Li XD, Chen Z. 2010. CC2D1A, a DM14 and C2 domainprotein, activates NF-kappaB through the canonical pathway. J. Biol.Chem. 285:24372–24380.

Usami et al.

3756 jvi.asm.org Journal of Virology

on February 13, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from