Abstract
Mitochondrial dynamics are pivotal for host immune responses upon infection, yet how viruses manipulate these processes to impair host defence and enhance viral fitness remains unclear. Here we show that Kaposi’s sarcoma-associated herpesvirus (KSHV), an oncogenic virus also known as human herpesvirus 8, encodes Bcl-2 (vBcl-2), which reprogrammes mitochondrial architecture. It binds with NM23-H2, a host nucleoside diphosphate (NDP) kinase, to stimulate GTP loading of the dynamin-related protein (DRP1) GTPase, which triggers mitochondrial fission, inhibits mitochondrial antiviral signalling protein (MAVS) aggregation and impairs interferon responses in cell lines. An NM23-H2-binding-defective vBcl-2 mutant fails to evoke fission, leading to defective virion assembly due to activated MAVS–IFN signalling. Notably, we identify two key interferon-stimulated genes restricting vBcl-2-dependent virion morphogenesis. Using a high-throughput drug screening, we discover an inhibitor targeting vBcl-2–NM23-H2 interaction that blocks virion production in vitro. Our study identifies a mechanism in which KSHV manipulates mitochondrial dynamics to allow for virus assembly and shows that targeting the virus–mitochondria interface represents a potential therapeutic strategy.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
The KSHV vBcl-2 gene sequence (gene ID 4961447) was obtained from NCBI. The crystal structures of the human NM23-H2 (PDB 1NSK) and KSHV vBcl-2 (PDB 1K3K) complex were obtained from the Protein Data Bank. The raw RNA sequencing data for BJAB cells expressing vBcl-2 WT (labelled as ‘W’), vBcl-2 E14A (labelled as ‘E’) and empty vector (labelled as ‘V’) have been deposited at NCBI GEO database under accession number GSE292579. RNA sequence reads were aligned to the human genome (UCSC hg38). This database is available at https://www.genome.ucsc.edu/cgi-bin/hgGateway. Source data are provided with this paper.
References
Tiku, V., Tan, M. W. & Dikic, I. Mitochondrial functions in infection and immunity. Trends Cell Biol. 30, 263–275 (2020); erratum 30, 748 (2020).
Nunnari, J. & Suomalainen, A. Mitochondria: in sickness and in health. Cell 148, 1145–1159 (2012).
Mesri, E. A., Cesarman, E. & Boshoff, C. Kaposi’s sarcoma and its associated herpesvirus. Nat. Rev. Cancer 10, 707–719 (2010).
Naimo, E., Zischke, J. & Schulz, T. F. Recent advances in developing treatments of Kaposi’s sarcoma herpesvirus-related diseases. Viruses https://doi.org/10.3390/v13091797 (2021).
Cavallin, L. E., Goldschmidt-Clermont, P. & Mesri, E. A. Molecular and cellular mechanisms of KSHV oncogenesis of Kaposi’s sarcoma associated with HIV/AIDS. PLoS Pathog. 10, e1004154 (2014).
Lange, P. & Damania, B. Kaposi sarcoma-associated herpesvirus (KSHV). Trends Microbiol. 28, 236–237 (2020).
Chang, H. H. & Ganem, D. A unique herpesviral transcriptional program in KSHV-infected lymphatic endothelial cells leads to mTORC1 activation and rapamycin sensitivity. Cell Host Microbe 13, 429–440 (2013).
Brulois, K. F. et al. Construction and manipulation of a new Kaposi’s sarcoma-associated herpesvirus bacterial artificial chromosome clone. J. Virol. 86, 9708–9720 (2012).
Liang, Q. et al. Identification of the essential role of viral Bcl-2 for Kaposi’s sarcoma-associated herpesvirus lytic replication. J. Virol. 89, 5308–5317 (2015).
Gelgor, A. et al. Viral Bcl-2 encoded by the Kaposi’s sarcoma-associated herpesvirus is vital for virus reactivation. J. Virol. 89, 5298–5307 (2015).
Gallo, A., Lampe, M., Gunther, T. & Brune, W. The viral Bcl-2 homologs of Kaposi’s sarcoma-associated herpesvirus and rhesus rhadinovirus share an essential role for viral replication. J. Virol. https://doi.org/10.1128/JVI.01875-16 (2017).
Sarid, R., Sato, T., Bohenzky, R. A., Russo, J. J. & Chang, Y. Kaposi’s sarcoma-associated herpesvirus encodes a functional bcl-2 homologue. Nat. Med. 3, 293–298 (1997).
Hosseinipour, M. C. et al. Viral profiling identifies multiple subtypes of Kaposi’s sarcoma. mBio 5, e01633-14 (2014).
Loh, J. et al. A surface groove essential for viral Bcl-2 function during chronic infection in vivo. PLoS Pathog. 1, e10 (2005).
Pattingre, S. et al. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 122, 927–939 (2005).
Boissan, M., Schlattner, U. & Lacombe, M. L. The NDPK/NME superfamily: state of the art. Lab. Invest. 98, 164–174 (2018).
Myoung, J. & Ganem, D. Generation of a doxycycline-inducible KSHV producer cell line of endothelial origin: maintenance of tight latency with efficient reactivation upon induction. J. Virol. Methods 174, 12–21 (2011).
Veron, M. et al. Nucleoside diphosphate kinase: an old enzyme with new functions? Adv. Exp. Med. Biol. 370, 607–611 (1994).
Bosnar, M. H., Bago, R. & Cetkovic, H. Subcellular localization of Nm23/NDPK A and B isoforms: a reflection of their biological function? Mol. Cell. Biochem. 329, 63–71 (2009).
Sharma, S., Sengupta, A. & Chowdhury, S. NM23/NDPK proteins in transcription regulatory functions and chromatin modulation: emerging trends. Lab. Invest. 98, 175–181 (2018).
Mears, J. A. et al. Conformational changes in Dnm1 support a contractile mechanism for mitochondrial fission. Nat. Struct. Mol. Biol. 18, 20–26 (2011).
Otera, H. et al. Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. J. Cell Biol. 191, 1141–1158 (2010).
Chan, D. C. Fusion and fission: interlinked processes critical for mitochondrial health. Annu. Rev. Genet. 46, 265–287 (2012).
Cassidy-Stone, A. et al. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev. Cell 14, 193–204 (2008).
Wu, X., Zheng, S., Cui, L., Wang, H. & Ng, T. B. Isolation and characterization of a novel ribonuclease from the pink oyster mushroom Pleurotus djamor. J. Gen. Appl. Microbiol. 56, 231–239 (2010).
Burman, J. L. et al. Mitochondrial fission facilitates the selective mitophagy of protein aggregates. J. Cell Biol. 216, 3231–3247 (2017).
Narendra, D. P. et al. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 8, e1000298 (2010).
Boissan, M. et al. Membrane trafficking. Nucleoside diphosphate kinases fuel dynamin superfamily proteins with GTP for membrane remodeling. Science 344, 1510–1515 (2014).
Tiku, V., Tan, M. W. & Dikic, I. Mitochondrial functions in infection and immunity. Trends Cell Biol. 30, 263–275 (2020).
Hou, F. et al. MAVS forms functional prion-like aggregates to activate and propagate antiviral innate immune response. Cell 146, 448–461 (2011).
Nakamura, H. et al. Global changes in Kaposi’s sarcoma-associated virus gene expression patterns following expression of a tetracycline-inducible Rta transactivator. J. Virol. 77, 4205–4220 (2003).
Xu, H. et al. Structural basis for the prion-like MAVS filaments in antiviral innate immunity. eLife 3, e01489 (2014).
Brubaker, S. W., Gauthier, A. E., Mills, E. W., Ingolia, N. T. & Kagan, J. C. A bicistronic MAVS transcript highlights a class of truncated variants in antiviral immunity. Cell 156, 800–811 (2014).
Ward, P. L., Ogle, W. O. & Roizman, B. Assemblons: nuclear structures defined by aggregation of immature capsids and some tegument proteins of herpes simplex virus 1. J. Virol. 70, 4623–4631 (1996).
Sathish, N. & Yuan, Y. Functional characterization of Kaposi’s sarcoma-associated herpesvirus small capsid protein by bacterial artificial chromosome-based mutagenesis. Virology 407, 306–318 (2010).
Serrero, M. C. et al. The interferon-inducible GTPase MxB promotes capsid disassembly and genome release of herpesviruses. eLife https://doi.org/10.7554/eLife.76804 (2022).
Reddi, T. S., Merkl, P. E., Lim, S. Y., Letvin, N. L. & Knipe, D. M. Tripartite motif 22 (TRIM22) protein restricts herpes simplex virus 1 by epigenetic silencing of viral immediate-early genes. PLoS Pathog. 17, e1009281 (2021).
Hauck, P., Chao, B. H., Litz, J. & Krystal, G. W. Alterations in the Noxa/Mcl-1 axis determine sensitivity of small cell lung cancer to the BH3 mimetic ABT-737. Mol. Cancer Ther. 8, 883–892 (2009).
Trott, O. & Olson, A. J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31, 455–461 (2010).
Mills, E. L., Kelly, B. & O’Neill, L. A. J. Mitochondria are the powerhouses of immunity. Nat. Immunol. 18, 488–498 (2017).
Chatel-Chaix, L. et al. Dengue virus perturbs mitochondrial morphodynamics to dampen innate immune responses. Cell Host Microbe 20, 342–356 (2016).
Vilmen, G. et al. BHRF1, a BCL2 viral homolog, disturbs mitochondrial dynamics and stimulates mitophagy to dampen type I IFN induction. Autophagy 17, 1296–1315 (2021).
Gao, S.-J., Deng, J.-H. & Zhou, F.-C. Productive lytic replication of a recombinant Kaposi’s sarcoma-associated herpesvirus in efficient primary infection of primary human endothelial cells. J. Virol. 77, 9738–9749 (2003).
Yang, Y. et al. Autophagic UVRAG promotes UV-induced photolesion repair by activation of the CRL4(DDB2) E3 ligase. Mol. Cell 62, 507–519 (2016).
Choi, Y. J. et al. SERPINB1-mediated checkpoint of inflammatory caspase activation. Nat. Immunol. 20, 276–287 (2019).
Honorato, R. V. et al. The HADDOCK2.4 web server: a leap forward in integrative modelling of biomolecular complexes. Nat. Protoc. 19, 3219–3241 (2024).
Eberhardt, J., Santos-Martins, D., Tillack, A. F. & Forli, S. AutoDock Vina 1.2.0: new docking methods, expanded force field, and Python bindings. J. Chem. Inf. Model. 61, 3891–3898 (2021).
Vangone, A. & Bonvin, A. M. Contacts-based prediction of binding affinity in protein–protein complexes. eLife 4, e07454 (2015).
Xue, L. C., Rodrigues, J. P., Kastritis, P. L., Bonvin, A. M. & Vangone, A. PRODIGY: a web server for predicting the binding affinity of protein–protein complexes. Bioinformatics 32, 3676–3678 (2016).
Brar, K. K. et al. PERK-ATAD3A interaction provides a subcellular safe haven for protein synthesis during ER stress. Science 385, eadp7114 (2024).
Rambold, A. S., Kostelecky, B., Elia, N. & Lippincott-Schwartz, J. Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc. Natl Acad. Sci. USA 108, 10190–10195 (2011).
Nagashima, S. et al. Golgi-derived PI(4)P-containing vesicles drive late steps of mitochondrial division. Science 367, 1366–1371 (2020).
Legland, D., Arganda-Carreras, I. & Andrey, P. MorphoLibJ: integrated library and plugins for mathematical morphology with ImageJ. Bioinformatics 32, 3532–3534 (2016).
Liu, Y. et al. MAVS Cys508 palmitoylation promotes its aggregation on the mitochondrial outer membrane and antiviral innate immunity. Proc. Natl Acad. Sci. USA 121, e2403392121 (2024).
Moschonas, G. D. et al. MX2 forms nucleoporin-comprising cytoplasmic biomolecular condensates that lure viral capsids. Cell Host Microbe 32, 1705–1724.e14 (2024).
Ram, B. M. & Dai, C. Detection of the DNA binding of transcription factors in situ at the single-cell resolution in cultured cells by proximity ligation assay. STAR Protoc. 4, 102692 (2023).
Hnasko, R. (ed.) ELISA. Methods and Protocols (Humana Press, 2015).
Triolo, M., Wade, S., Baker, N. & Khacho, M. Evaluating mitochondrial length, volume, and cristae ultrastructure in rare mouse adult stem cell populations. STAR Protoc. 4, 102107 (2023).
West, A. P. et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 520, 553–557 (2015).
Oh, S. et al. Downregulation of autophagy by Bcl-2 promotes MCF7 breast cancer cell growth independent of its inhibition of apoptosis. Cell Death Differ. 18, 452–464 (2011).
Proust, B. et al. NME6 is a phosphotransfer-inactive, monomeric NME/NDPK family member and functions in complexes at the interface of mitochondrial inner membrane and matrix. Cell Biosci. 11, 195 (2021).
Yang, Y. et al. Central role of autophagic UVRAG in melanogenesis and the suntan response. Proc. Natl Acad. Sci. USA 115, E7728–E7737 (2018).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).
Huang, Q., Petros, A. M., Virgin, H. W., Fesik, S. W. & Olejniczak, E. T. Solution structure of a Bcl-2 homolog from Kaposi sarcoma virus. Proc. Natl Acad. Sci. USA 99, 3428–3433 (2002).
Shikhaliev, K. S., Kryl’skii, D. V., Kovygin, Y. A. & Verezhnikov, V. N. Verezhnikov condensation of benzoxa(thia)zolyl-2-guanidines with dicarbonyl compounds. Russ. J. Gen. Chem. 75, 294–297 (2005).
Grytsai, O., Druhenko, T., Demange, L., Ronco, C. & Benhida, R. Cyanoguanidine as a versatile, eco-friendly and inexpensive reagent for the synthesis of 2-aminobenzoxazoles and 2-guanidinobenzoxazoles. Tetrahedron Lett. 59, 1642–1645 (2018).
Acknowledgements
We thank the Imaging Facility, Genomics Facility, Flow Cytometry Facility, Proteomics and Metabolomics Facility, Bioinformatics Facility, and Molecular Screening and Protein Expression Facility at The Wistar Institute; the Electron Microscopy Resource Laboratory at University of Pennsylvania for TEM sample preparation and processing; Medinoah, Inc. for providing custom synthesis services. Graphic design was created with BioRender.com, for which the authors possess a licence. This work was supported by NIH awards R35GM119721 to J.S.; R01 CA251275 and R01 AI181758 to J.U.J.; R21 DE028256, R01 CA238457, R01 CA140964 and R01 CA262631 to C.L., and the Wistar Science Accelerator Postdoctoral Award to Q.Z.
Author information
Authors and Affiliations
Contributions
C.L. conceptualized the project. Q.Z., R.M., J.C., B.M., S.G., C.P., J.L., D.S., L.Z., J.M.S., Q. Liu, A.V.K., J.S. and C.L. designed the methodology. Q.Z., R.M., J.S.M., J.C., Z.Z., H.G. and C.L. conducted investigations. C.L. and Q.Z. wrote the original paper draft. Q.Z., J.S.M., J.C., D.C.A., P.M.L., M.E.M., J.S., J.M.S. and C.L. reviewed and edited the paper. Q.Z. and C.L. acquired funding. D.C.A., S-J.G., P.F., J.U.J., Q. Liang and C.L. acquired resources. C.L. supervised the project.
Corresponding author
Ethics declarations
Competing interests
C.L., Q.Z. and R.M. are inventors of provisional patent 63/693,000, assigned to The Wistar Institute, and claiming compositions and methods disclosed herein. The other authors declare no competing interests.
Peer review
Peer review information
Nature Microbiology thanks Craig McCormick, Richard Stanton and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 KSHV vBcl-2 targets NM23-H2 independently of inhibition of autophagy and apoptosis.
a, Co-immunoprecipitation (co-IP) of vBcl-2 with NM23-H2 in HEK293T cells co-transfected with HA-vBcl-2 and Flag-NM23-H2. Actin serves as a loading control. b, Co-IP of endogenous NM23-H2 with vBcl-2 in HEK293T cells expressing empty vector (Vec) or HA-vBcl-2. c, Schematic of WT vBcl-2 and deletion mutants, with binding to NM23-H2 determined by co-IP of Flag-NM23-H2 with HA-vBcl-2 in HEK293T cell lysates. BH (Bcl-2 homology) domains and the conserved 84W85G86R residues in BH1 domain are highlighted. TM, transmembrane domain. +, strong binding; -, no binding. d, Co-IP of Flag-tagged NM23-H2 with HA-tagged wild-type (WT) or mutant vBcl-2 in HEK293T cells. The binding results are summarized in c. e, IB analysis of LC3-I/LC3-II and p62 in HEK293T cells stably expressing Vec or HA-vBcl-2 (WT or mutants) with or without Torin 1 (50 nM, 3 h). f, Densitometric quantification of LC3-II/LC3-I (red bars) and p62/actin (blue) from e. n = 3. g, Flow cytometry analysis of apoptosis in HEK293T cells stably expressing Vec or HA-vBcl-2 (WT or mutants) treated with TNF-α (10 ng/ml) and CHX (5 μg/ml) for 4 h. Early apoptotic cells (Annexin V-positive and PI-negative) were quantified (n = 3). Data in a,b,d,e are from one experiment that is representative of three independent experiments. See Source Data for uncropped data of a,b,d,e. Data shown in f,g are mean ± s.d. analyzed by one-way ANOVA followed by Tukey’s post hoc test for comparisons among multiple groups. *, p < 0.05; **, p < 0.01; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 1.
Extended Data Fig. 2 vBcl-2 interaction with NM23-H2 is required for infectious virion production.
a, Experimental schematic to assess vBcl-2 knockout (KO) effects on KSHV reactivation. iSLK-BAC16.vBcl-2 WT and iSLK-BAC16-vBcl-2 KO cells were induced (Dox/NaB, 60-72 h), and supernatants infected naïve SLK cells for 24 h (results in b-d). b, Bright field (BF) and GFP images of SLK cells infected with virus from induced iSLK-BAC16 cells. Scale bars, 50 μm. c,d, Virus titres determined by two-fold serial dilution of supernatants collected 72 h post-reactivation (a) and quantification of GFP-positive SLK cells per well by flow cytometry at 24 h post-infection (c), expressed as infectious units/ml (d). e, Schematic for assessing vBcl-2 KO impact on KSHV de novo infection in HCT116 cells infected with vBcl-2 WT or vBcl-2 KO KSHV at MOI = 10, and supernatant collected were used to infect SLK cells for 24 h (results in f-h). f, GFP images of SLK cells infected in e. Scale bars, 430 μm. g, Flow cytometry analysis of GFP-positive SLK cell percentages and mean fluorescence intensity from e. h, qPCR analysis of viral genome copies in supernatants from e. (n = 3 independent experiments). i, Close-up view of site variations between NM23-H2 (yellow) and -H1 (cyan) in the structural model of NM23-H2 and vBcl-2. The corresponding residues of NM23-H2 and NM23-H1 are shown in stick. The ΔG for the hydrogen (H)-bonding interactions of NM23-H2 Q50 was noted65, which was lost when Q50 was replaced by E50. j, Co-IP of Flag-NM23-H2 (WT/mutant) with HA-vBcl-2 in HEK293T cells. k-m, iSLK-BAC16.vBcl-2 WT cells transduced with ctrl or NM23-H1 shRNA induced as above and supernatants infected SLK cells. GFP/BF images are shown in k, virus titer determined in l (n = 3), and protein expression shown in m. Data in j,m are representative of three independent experiments (see Source Data for uncropped data). Data in d,h,l are mean ± s.d. analyzed by two-tailed Student’s t test and one-way ANOVA with Tukey’s post hoc test. *, p < 0.05; **, p < 0.01; ****, p < 0.0001. ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 2.
Extended Data Fig. 3 KSHV vBcl-2 regulates NM23-H2 activities.
a, IB (top) and densitometric quantification (bottom) of NM23-H2 stability in cells transduced with Vec or HA-vBcl-2 (WT/E14A) and treated with CHX (n = 3). b, Coupled pyruvate kinase-lactate dehydrogenase assay (Methods) showing vBcl-2-driven NDPK activity of recombinant NM23-H2 preincubated with GST-vBcl-2 (WT/E14A) or Vec (n = 3 independent experiments). c, in vitro assay showing that increasing amount of recombinant GST-vBcl-2 (WT or E14A) modulate histidine phosphorylation of purified His-NM23-H2 in the presence of ATP, detected by IB with anti-1-pHis antibody. Coomassie blue staining shows input of each purified proteins as indicated. d, Densitometric quantification of 1-pHis levels in (c) normalized to NM23-H2 input (n = 3 independent experiments). e-g, Representative confocal images of in HeLa cells (e) co-transfected with HA-vBcl-2 and Flag-NM23-H2 showing the distributions of vBcl-2 (green) and NM23-H2 (red) relative to organelle markers (magenta), including PDI (ER), TGN46 (trans-Golgi), TOM20 (mitochondria), and WGA (wheat germ agglutinin; plasma membrane). Scale bars, 10 μm. The percentage of vBcl-2 overlapping with each marker (f) and vBcl-2 colocalization with NM23-H2 (g) were quantified (n = 50 cells pooled from three independent experiments). Data in a,c,e are from one experiment that is representative of three independent experiments. See Source Data for uncropped data of a,c. Data in a,b,d represent mean ± s.d. analyzed by one-way ANOVA followed by Tukey’s post hoc test. Box-plot data in f,g are presented as median (center line), interquartile range (IQR; box, 25th to 75th percentiles), and whiskers extending from minimum to maximum values; and analyzed using Kruskal-Wallis with post hoc Dunn’s test. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Fig. 3.
Extended Data Fig. 4 Characterization of the impacts of vBcl-2 on mitochondrial morphology.
a, Confocal micrographs of mitochondria in iSLK-BAC16.vBcl-2 WT/KO cells induced by Dox/NaB for indicated time and stained with MitoTracker (red) and DAPI (blue). b, Quantification of mitochondrial morphology (tubulated, intermediate, and fragmented) in a (n = 3, with 100 cells/experiment). c-e, Analysis of mitochondrial numbers per cell (c), mean area (d) and perimeter (e) in cells in a (n = 94 cells/group, 3 independent experiments). f,g, Confocal micrographs of mitochondria (red) in cells transduced with indicated shRNAs plus NM23-H2 rescue plasmids post-reactivation (f) with corresponding quantification of fragmentation (g; n = 3, 300 cells/sample). h, IB of the indicated mitochondrial fission and fusion factors in mitochondrial fractions and WCL from iSLK-BAC16.vBcl-2 KO cells stably expressing HA-vBcl-2 (WT/mutants) at 60 h post-reactivation. TOM20, control for mitochondrial fraction. GAPDH, a loading control for WCLs (see Source Data for uncropped WB). i, Confocal micrographs of mitochondria (TOM20, green) in iSLK-BAC16.vBcl-2 KO cells expressing mCherry-vBcl-2 (red) and transduced with Vec, an MFN2-encoding plasmid, or with Ctrl shRNA, DRP1 shRNA, MFF shRNAs at 60 h post- reactivation. j, Percentage of cells in (i) with mitochondrial fission was quantified (n = 3, with 100 cells/experiment). k, Infectious virus titer in cells in (i) with MFF depletion was quantified by infecting SLK cells (top, n = 3) and GFP images are shown (left). Scale bars, 10 μm. Data in a,f,h,i,k are from one experiment that is representative of three independent experiments. Data in b,g,j,k represent mean ± s.d. analyzed by one-way ANOVA followed by Tukey’s post hoc test. Box-plot data in c-e are presented as median (center line), interquartile range (IQR; box, 25th to 75th percentiles), and whiskers extending from minimum to maximum values; and analyzed using Kruskal-Wallis with post hoc Dunn’s test. ***, p < 0.001; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 4.
Extended Data Fig. 5 Impacts of vBcl-2 on mitochondrial function and integrity.
a, Representative confocal micrographs (left) and quantification (right) of PINK1 (magenta) colocalization with TOM20 (green) in iSLK-BAC16.vBcl-2 KO cells expressing mCherry-vBcl-2 (red) at 60 h post-reactivation (n = 100 cells/sample, 3 independent experiments). Scale bars, 10 μm. b, Co-IP of vBcl-2 and endogenous NM23-H2 with DRP1 in HEK293T cells expressing NM23-H2 shRNA or Ctrl shRNA and transduced with Vec or HA-vBcl-2. c, Co-IP of indicated proteins in HEK293T expressing mCherry-vBcl-2 (WT/E14A). The relative amount of HA-NM23-H2 co-immunoprecipitated with DRP1 was normalized to total HA-NM23-H2 in WCL (right; n = 3 independent experiments). d, DRP1 GTPase activity in the presence/absence of purified NM23-H2, vBcl-2 (WT/E14A), and indicated nucleotides (n = 3 independent experiments). Data in a-c are representative of three independent experiments. See Source Data for uncropped data of b,c. Data in a,c,d are mean ± s.d. analyzed by one-way ANOVA with Tukey’s post hoc test. Exact P values are provided in Source Data Extended Data Fig. 5.
Extended Data Fig. 6 KSHV vBcl-2 attenuates MAVS-mediated IFN response.
a-c, RT-qPCR analysis of IFNB1 (a) and ISG15 (c) mRNA levels (normalized to 18S rRNA), and IFN-β production (b) in supernatant measured by ELISA in iSLK-BAC16.vBcl-2 WT and vBcl-2 KO cells induced with Dox/NaB for the indicated time. n = 3. d-f, RT-qPCR analysis of vBcl-2 transcripts (d, n = 3), IFNB1 transcripts (e, n = 3), and ISG15 transcripts (f, n = 3) in TREx BCBL-1-RTA cells transfected with two independent vBcl-2-specific siRNA for 12 h and induced with Dox (1 μg/ml) and Nab (1 mM) for 48 h. g, RT-qPCR of IFNB1 mRNA in HCT116 cells infected with vBcl-2 WT or vBcl-2 KO KSHV at 10 MOI for 60 h (n = 3). h, Representative STED micrographs of MAVS (magenta) and TOM20 (green) in iSLK-BAC16.vBcl-2 WT cells transduced with Ctrl vs. DRP1 shRNAs or NM23-H2 shRNAs at 60 h post-reactivation. Insets highlight MAVS distribution. Arrows indicate MAVS puncta and dotted lines denote clustering. Data are from one experiment that is representative of three independent experiments. i, Diameter of MAVS clusters measured from images in h (n = 100). j. RT-qPCR of ISG15 mRNA expression in h (n = 3). Data in a-g represent mean ± s.d. obtained from three independent experiments performed in triplicates, and analyzed by two-tailed t test (a-c) or one-way ANOVA followed by Tukey’s post hoc test (d-g). Box-plot data in i are presented as median (center line), interquartile range (IQR; box, 25th to 75th percentiles), and whiskers extending from minimum to maximum values; and analyzed using Kruskal-Wallis with post hoc Dunn’s test. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 6.
Extended Data Fig. 7 The NM23-H2 binding-deficient vBcl-2 E14A mutant KSHV is defective in viral capsid assembly and nuclear egress during the late phase of lytic reactivation.
a, RT-qPCR of viral gene expression in iSLK-BAC16.vBcl-2 cells (WT and KO) stably expressing Vec, HA-vBcl-2 (WT/mutants) at indicated time post-reactivation with Dox/NaB (n = 3). b, IB of viral proteins in WCLs from (a) at 48 h post-reactivation. IE/E, immediate early and early genes; L, late genes. c, Relative viral genome copy number changes in cells from (a; n = 3). d, Representative confocal micrographs of ORF65 (red) in iSLK-BAC16.vBcl-2 WT cells expressing Ctrl shRNA, NM23-H2 shRNAs, or DRP1 shRNAs after 60 h Dox/NaB induction. iSLK-BAC16.vBcl-2 KO cells were included as a control. DAPI stains nuclei (blue). Green signals (GFP) mark virus-infected cells. e, Percentage of cells with cytoplasmic ORF65 in (d) was quantified (n = 3, with 100 cells/group). f, IB for ORF65, NM23-H2, and DRP1 expression in cells in (d). g, Confocal micrographs of ORF65 (red) in HCT116 cells infected with vBcl-2 WT or vBcl-2 KO KSHV at 10 MOI for 60 h (left). The percentage of cells with cytoplasmic ORF65 was quantified (right; n = 3, with 100 cells/group). Scale bars, 10 μm. Data in b,d,f,g are from one experiment that is representative of three independent experiments. See Source Data for uncropped data of b,f. Data shown in a,c,e,g represent mean ± s.d. analyzed by Student’s two-tailed t test or one-way ANOVA followed by Tukey’s post hoc test. ***, p < 0.001; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 7.
Extended Data Fig. 8 Identification of IFN-induced antiviral factors that restrict vBcl-2-dependent capsid assembly and nuclear egress.
a, Representative confocal images of ORF65 staining in iSLK-BAC16.vBcl-2 KO cells transduced with ISG-specific shRNA as indicated. Scale bars, 10 μm. b, Quantification of the percentage of cells from (a) with cytoplasmic ORF65 staining (100 cells/experiment, n = 3). c, RT-qPCR validation of ISG knockdown in (a, n = 3). d,e, TRIM22 and MxB suppress infectious KSHV production. iSLK-BAC16.vBcl-2 KO cells were transduced with Ctrl or ISG-specific shRNA and induced with Dox/NaB for 72 h. Supernatants were harvested and infectious virus yield was determined by infecting SLK cells for 24 h. Representative GFP/BF images in SLK cells are shown (d). Scale bars, 430 μm. Virus yield was quantified as the percentage of GFP-positive SLK cells (e). DKD, double knockdown (n = 3). f, RT-qPCR analysis of TRIM22 and MxB mRNA in HCT116 cells infected with vBcl-2 WT or vBcl-2 KO KSHV at MOI = 10 for 60 h (n = 3). g-i, Confocal images of ORF65 staining in iSLK-BAC16.vBcl-2 WT/KO cells expressing TRIM22 and MxB post-reactivation. Percentage of cells with cytoplasmic ORF65 (h; n = 3, 300 cells/sample) and the percentage of cytoplasmic ORF65 per cell (i; n = 49 cells/sample) were quantified. Scale bars, 10 μm. Data in a,d,g are from one experiment that is representative of three independent experiments. Data in b,c,e,f,h represent mean ± s.d. analyzed by Student’s two-tailed t test or by one-way ANOVA followed by Tukey’s post hoc test. Scatter plots in i are analyzed with Kruskal-Wallis with post hoc Dunn’s test. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 8.
Extended Data Fig. 9 VBNI-1 disrupts KSHV vBcl-2 interaction with NM23-H2 and impairs late stage KSHV lytic replication.
a, HTRF assay. His-NM23-H2 is bound by an anti-His-Tb HTRF donor fluorophore. GST-vBcl-2 is bound by an anti-GST-d2 HTRF acceptor. vBcl-2 binding to NM23-H2 produces FRET. The addition of small molecule inhibitor(s) reduces FRET. b, HTRF showing dose-dependent vBcl-2 binding to NM23-H2 (3 nM; red) or NM23-H1(25 nM; blue fitted by nonlinear regression. c, HTRF comparison of vBcl-2 binding to NM23-H2 vs. BID BH3 peptide in the presence of ABT-263; IC50 values indicated (n = 3). d-g, Co-IP of endogenous NM23-H2, Beclin1, and BAK with vBcl-2 in HEK293T cells expressing HA-vBcl-2 after VBNI-1 treatment at increasing doses for 36 h (d,e) or with VBNI-1 (2 μM) at increasing timepoints (f,g). Interactions with indicated binding partners were quantified in (e,g; n = 3). h, The effects of VBNI-1 on NM23-H1/H2 kinase activity measured by Transcreener ADP2-FI assay; inhibition curves with IC50 values shown. i, Docking model (left) shows the electrostatic surface of vBcl-2 harbouring VBNI-1 (sphere). Overlay (right) of vBcl-2-VBNI-1 with vBcl-2-NM23-H2 highlights potential steric clash between VBNI-1 and NM23-H2 interface residue K128. For clarity, the vBcl-2 molecule bound to VBNI-1 is not shown. j, SPR analysis of VBNI-1 interaction with vBcl-2 across concentrations. k, Cytotoxicity of VBNI-1 in SLK (blue) and BJAB (red) cells; median effective concentration (EC50) values indicated. l, Dose-dependent inhibition of KSHV production by VBNI-1 in reactivated iSLK-BAC16.vBcl-2 WT but not KO cells; EC50 values indicated. m-o, Virus production (m,n) and gene expression (o) following VBNI-1 treatment and indicated shRNA knockdowns post-reactivation. GFP/BF images shown in m; quantified in n (n = 3). Scale bars, 430 μm. Data in d,f,j,m are from one experiment that is representative of three independent experiments. See Source Data for uncropped data (d,f). Data in e,g,n,o represent mean ± s.d. analyzed by one-way ANOVA followed by Tukey’s post hoc test. **, p < 0.01; ****, p < 0.0001; ns, not significant. Exact P values are provided in Source Data Extended Data Fig. 9.
Extended Data Fig. 10 Schematic representation of the mechanistic action of vBcl-2 manipulation of mitochondrial dynamics for immune subversion and its therapeutic targeting by VBNI-1.
vBcl-2 interaction with host NM23-H2 allows for activation of DRP1-mediated mitochondrial fission, which reduces MAVS aggregation and dampens the downstream type I IFN response, leading to productive virion assembly and nuclear egress. This virus-host interaction can be specifically antagonized by VBNI-1. VBNI-1 blocks the late phase of viral assembly/egress and thwarts virion production through a mechanism that involves induced expression of the ISGs TRIM22 and MxB. See main text for details.
Supplementary information
Supplementary Information
Supplementary Figs. 1–15, Tables 1–4 and Note.
Supplementary Data 1
Statistical source data for Supplementary Figs. 2–5, 7 and 9–13.
Supplementary Data 2
Chemical structure of VBNI-1.
Source data
Source Data Fig. 1
Statistical source data.
Source Data Fig. 2
Statistical source data.
Source Data Fig. 3
Statistical source data.
Source Data Fig. 4
Statistical source data.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 2
Statistical source data.
Source Data Extended Data Fig. 3
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 5
Statistical source data.
Source Data Extended Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 9
Statistical source data.
Source Data Figs. 1–6 and Source Data Extended Data Figs. 1–5, 7 and 9
Uncropped western blots and/or gels of Figs. 1–6 and Extended Data Figs. 1–5, 7 and 9.
Rights and permissions
Springer Nature or its licensor (e.g. 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.
About this article
Cite this article
Zhu, Q., McElroy, R., Machhar, J.S. et al. Kaposi’s sarcoma-associated herpesvirus induces mitochondrial fission to evade host immune responses and promote viral production. Nat Microbiol 10, 1501–1520 (2025). https://doi.org/10.1038/s41564-025-02018-3
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41564-025-02018-3