All Publications


  • Efficacy of Surgical Management of Malignant Bowel Obstruction on Preventing Hospital Readmission - Experience of a Tertiary Care Center Sun, B. J., Ayala, C. I., Einkauf, K. B., Xu, N., Yue, T. M., Lee, B. SPRINGER. 2023: S110
  • Outcomes of Procedural-Based Palliation for Malignant Bowel Obstruction Sun, B. J., Xu, N., Yue, T. M., Einkauf, K. B., Lee, B. SPRINGER. 2023: S103
  • Phenotypic signatures of immune selection in HIV-1 reservoir cells. Nature Sun, W., Gao, C., Hartana, C. A., Osborn, M. R., Einkauf, K. B., Lian, X., Bone, B., Bonheur, N., Chun, T. W., Rosenberg, E. S., Walker, B. D., Yu, X. G., Lichterfeld, M. 2023

    Abstract

    Human immunodeficiency virus 1 (HIV-1) reservoir cells persist lifelong despite antiretroviral treatment1,2 but may be vulnerable to host immune responses that could be exploited in strategies to cure HIV-1. Here we used a single-cell, next-generation sequencing approach for the direct ex vivo phenotypic profiling of individual HIV-1-infected memory CD4+ T cells from peripheral blood and lymph nodes of people living with HIV-1 and receiving antiretroviral treatment for approximately 10 years. We demonstrate that in peripheral blood, cells harbouring genome-intact proviruses and large clones of virally infected cells frequently express ensemble signatures of surface markers conferring increased resistance to immune-mediated killing by cytotoxic T and natural killer cells, paired with elevated levels of expression of immune checkpoint markers likely to limit proviral gene transcription; this phenotypic profile might reduce HIV-1 reservoir cell exposure to and killing by cellular host immune responses. Viral reservoir cells harbouring intact HIV-1 from lymph nodes exhibited a phenotypic signature primarily characterized by upregulation of surface markers promoting cell survival, including CD44, CD28, CD127 and the IL-21 receptor. Together, these results suggest compartmentalized phenotypic signatures of immune selection in HIV-1 reservoir cells, implying that only small subsets of infected cells with optimal adaptation to their anatomical immune microenvironment are able to survive during long-term antiretroviral treatment. The identification of phenotypic markers distinguishing viral reservoir cells may inform future approaches for strategies to cure and eradicate HIV-1.

    View details for DOI 10.1038/s41586-022-05538-8

    View details for PubMedID 36599977

  • Progressive transformation of the HIV-1 reservoir cell profile over two decades of antiviral therapy. Cell host & microbe Lian, X., Seiger, K. W., Parsons, E. M., Gao, C., Sun, W., Gladkov, G. T., Roseto, I. C., Einkauf, K. B., Osborn, M. R., Chevalier, J. M., Jiang, C., Blackmer, J., Carrington, M., Rosenberg, E. S., Lederman, M. M., McMahon, D. K., Bosch, R. J., Jacobson, J. M., Gandhi, R. T., Peluso, M. J., Chun, T. W., Deeks, S. G., Yu, X. G., Lichterfeld, M. 2022

    Abstract

    HIV-1 establishes a life-long reservoir of virally infected cells which cannot be eliminated by antiretroviral therapy (ART). Here, we demonstrate a markedly altered viral reservoir profile of long-term ART-treated individuals, characterized by large clones of intact proviruses preferentially integrated in heterochromatin locations, most prominently in centromeric satellite/micro-satellite DNA. Longitudinal evaluations suggested that this specific reservoir configuration results from selection processes that promote the persistence of intact proviruses in repressive chromatin positions, while proviruses in permissive chromosomal locations are more likely to be eliminated. A bias toward chromosomal integration sites in heterochromatin locations was also observed for intact proviruses in study participants who maintained viral control after discontinuation of antiretroviral therapy. Together, these results raise the possibility that antiviral selection mechanisms during long-term ART may induce an HIV-1 reservoir structure with features of deep latency and, possibly, more limited abilities to drive rebound viremia upon treatment interruptions.

    View details for DOI 10.1016/j.chom.2022.12.002

    View details for PubMedID 36596305

  • Immune correlates of HIV-1 reservoir cell decline in early-treated infants. Cell reports Hartana, C. A., Garcia-Broncano, P., Rassadkina, Y., Lian, X., Jiang, C., Einkauf, K. B., Maswabi, K., Ajibola, G., Moyo, S., Mohammed, T., Maphorisa, C., Makhema, J., Yuki, Y., Martin, M., Bennett, K., Jean-Philippe, P., Viard, M., Hughes, M. D., Powis, K. M., Carrington, M., Lockman, S., Gao, C., Yu, X. G., Kuritzkes, D. R., Shapiro, R., Lichterfeld, M. 2022; 40 (3): 111126

    Abstract

    Initiation of antiretroviral therapy (ART) in infected neonates within hours after birth limits viral reservoir seeding but does not prevent long-term HIV-1 persistence. Here, we report parallel assessments of HIV-1 reservoir cells and innate antiviral immune responses in a unique cohort of 37 infected neonates from Botswana who started ART extremely early, frequently within hours after birth. Decline of genome-intact HIV-1 proviruses occurs rapidly after initiation of ART and is associated with an increase in natural killer (NK) cell populations expressing the cytotoxicity marker CD57 and with a decrease in NK cell subsets expressing the inhibitory marker NKG2A. Immune perturbations in innate lymphoid cells, myeloid dendritic cells, and monocytes detected at birth normalize after rapid institution of antiretroviral therapy but do not notably influence HIV-1 reservoir cell dynamics. These results suggest that HIV-1 reservoir cell seeding and evolution in early-treated neonates is markedly influenced by antiviral NK cell immune responses.

    View details for DOI 10.1016/j.celrep.2022.111126

    View details for PubMedID 35858580

  • Parallel analysis of transcription, integration, and sequence of single HIV-1 proviruses CELL Einkauf, K. B., Osborn, M. R., Gao, C., Sun, W., Sun, X., Lian, X., Parsons, E. M., Gladkov, G. T., Seiger, K. W., Blackmer, J. E., Jiang, C., Yukl, S. A., Rosenberg, E. S., Yu, X. G., Lichterfeld, M. 2022; 185 (2): 266-+

    Abstract

    HIV-1-infected cells that persist despite antiretroviral therapy (ART) are frequently considered "transcriptionally silent," but active viral gene expression may occur in some cells, challenging the concept of viral latency. Applying an assay for profiling the transcriptional activity and the chromosomal locations of individual proviruses, we describe a global genomic and epigenetic map of transcriptionally active and silent proviral species and evaluate their longitudinal evolution in persons receiving suppressive ART. Using genome-wide epigenetic reference data, we show that proviral transcriptional activity is associated with activating epigenetic chromatin features in linear proximity of integration sites and in their inter- and intrachromosomal contact regions. Transcriptionally active proviruses were actively selected against during prolonged ART; however, this pattern was violated by large clones of virally infected cells that may outcompete negative selection forces through elevated intrinsic proliferative activity. Our results suggest that transcriptionally active proviruses are dynamically evolving under selection pressure by host factors.

    View details for DOI 10.1016/j.cell.2021.12.011

    View details for Web of Science ID 000746172100008

    View details for PubMedID 35026153

    View details for PubMedCentralID PMC8809251

  • Signatures of immune selection in intact and defective proviruses distinguish HIV-1 elite controllers. Science translational medicine Lian, X., Gao, C., Sun, X., Jiang, C., Einkauf, K. B., Seiger, K. W., Chevalier, J. M., Yuki, Y., Martin, M., Hoh, R., Peluso, M. J., Carrington, M., Ruiz-Mateos, E., Deeks, S. G., Rosenberg, E. S., Walker, B. D., Lichterfeld, M., Yu, X. G. 2021; 13 (624): eabl4097

    Abstract

    Increasing evidence suggests that durable drug-free control of HIV-1 replication is enabled by effective cellular immune responses that may induce an attenuated viral reservoir configuration with a weaker ability to drive viral rebound. Here, we comprehensively tracked effects of antiviral immune responses on intact and defective proviral sequences from elite controllers (ECs), analyzing both classical escape mutations and HIV-1 chromosomal integration sites as biomarkers of antiviral immune selection pressure. We observed that, within ECs, defective proviruses were commonly located in permissive genic euchromatin positions, which represented an apparent contrast to autologous intact proviruses that were frequently located in heterochromatin regions; this suggests differential immune selection pressure on intact versus defective proviruses in ECs. In comparison to individuals receiving antiretroviral therapy, intact and defective proviruses from ECs showed reduced frequencies of escape mutations in cytotoxic T cell epitopes and antibody contact regions, possibly due to the small and poorly inducible reservoir that may be insufficient to drive effective viral escape in ECs. About 15% of ECs harbored nef deletions in intact proviruses, consistent with increased viral vulnerability to host immunity in the setting of nef dysfunction. Together, these results suggest a distinct signature of immune footprints in proviral sequences from ECs.

    View details for DOI 10.1126/scitranslmed.abl4097

    View details for PubMedID 34910552

  • Loss of Bcl-6-Expressing T Follicular Helper Cells and Germinal Centers in COVID-19. Cell Kaneko, N., Kuo, H. H., Boucau, J., Farmer, J. R., Allard-Chamard, H., Mahajan, V. S., Piechocka-Trocha, A., Lefteri, K., Osborn, M., Bals, J., Bartsch, Y. C., Bonheur, N., Caradonna, T. M., Chevalier, J., Chowdhury, F., Diefenbach, T. J., Einkauf, K., Fallon, J., Feldman, J., Finn, K. K., Garcia-Broncano, P., Hartana, C. A., Hauser, B. M., Jiang, C., Kaplonek, P., Karpell, M., Koscher, E. C., Lian, X., Liu, H., Liu, J., Ly, N. L., Michell, A. R., Rassadkina, Y., Seiger, K., Sessa, L., Shin, S., Singh, N., Sun, W., Sun, X., Ticheli, H. J., Waring, M. T., Zhu, A. L., Alter, G., Li, J. Z., Lingwood, D., Schmidt, A. G., Lichterfeld, M., Walker, B. D., Yu, X. G., Padera, R. F., Pillai, S. 2020; 183 (1): 143-157.e13

    Abstract

    Humoral responses in coronavirus disease 2019 (COVID-19) are often of limited durability, as seen with other human coronavirus epidemics. To address the underlying etiology, we examined post mortem thoracic lymph nodes and spleens in acute SARS-CoV-2 infection and observed the absence of germinal centers and a striking reduction in Bcl-6+ germinal center B cells but preservation of AID+ B cells. Absence of germinal centers correlated with an early specific block in Bcl-6+ TFH cell differentiation together with an increase in T-bet+ TH1 cells and aberrant extra-follicular TNF-α accumulation. Parallel peripheral blood studies revealed loss of transitional and follicular B cells in severe disease and accumulation of SARS-CoV-2-specific "disease-related" B cell populations. These data identify defective Bcl-6+ TFH cell generation and dysregulated humoral immune induction early in COVID-19 disease, providing a mechanistic explanation for the limited durability of antibody responses in coronavirus infections, and suggest that achieving herd immunity through natural infection may be difficult.

    View details for DOI 10.1016/j.cell.2020.08.025

    View details for PubMedID 32877699

    View details for PubMedCentralID PMC7437499

  • Distinct viral reservoirs in individuals with spontaneous control of HIV-1. Nature Jiang, C., Lian, X., Gao, C., Sun, X., Einkauf, K. B., Chevalier, J. M., Chen, S. M., Hua, S., Rhee, B., Chang, K., Blackmer, J. E., Osborn, M., Peluso, M. J., Hoh, R., Somsouk, M., Milush, J., Bertagnolli, L. N., Sweet, S. E., Varriale, J. A., Burbelo, P. D., Chun, T. W., Laird, G. M., Serrao, E., Engelman, A. N., Carrington, M., Siliciano, R. F., Siliciano, J. M., Deeks, S. G., Walker, B. D., Lichterfeld, M., Yu, X. G. 2020; 585 (7824): 261-267

    Abstract

    Sustained, drug-free control of HIV-1 replication is naturally achieved in less than 0.5% of infected individuals (here termed 'elite controllers'), despite the presence of a replication-competent viral reservoir1. Inducing such an ability to spontaneously maintain undetectable plasma viraemia is a major objective of HIV-1 cure research, but the characteristics of proviral reservoirs in elite controllers remain to be determined. Here, using next-generation sequencing of near-full-length single HIV-1 genomes and corresponding chromosomal integration sites, we show that the proviral reservoirs of elite controllers frequently consist of oligoclonal to near-monoclonal clusters of intact proviral sequences. In contrast to individuals treated with long-term antiretroviral therapy, intact proviral sequences from elite controllers were integrated at highly distinct sites in the human genome and were preferentially located in centromeric satellite DNA or in Krüppel-associated box domain-containing zinc finger genes on chromosome 19, both of which are associated with heterochromatin features. Moreover, the integration sites of intact proviral sequences from elite controllers showed an increased distance to transcriptional start sites and accessible chromatin of the host genome and were enriched in repressive chromatin marks. These data suggest that a distinct configuration of the proviral reservoir represents a structural correlate of natural viral control, and that the quality, rather than the quantity, of viral reservoirs can be an important distinguishing feature for a functional cure of HIV-1 infection. Moreover, in one elite controller, we were unable to detect intact proviral sequences despite analysing more than 1.5 billion peripheral blood mononuclear cells, which raises the possibility that a sterilizing cure of HIV-1 infection, which has previously been observed only following allogeneic haematopoietic stem cell transplantation2,3, may be feasible in rare instances.

    View details for DOI 10.1038/s41586-020-2651-8

    View details for PubMedID 32848246

    View details for PubMedCentralID PMC7837306

  • Early antiretroviral therapy in neonates with HIV-1 infection restricts viral reservoir size and induces a distinct innate immune profile. Science translational medicine Garcia-Broncano, P., Maddali, S., Einkauf, K. B., Jiang, C., Gao, C., Chevalier, J., Chowdhury, F. Z., Maswabi, K., Ajibola, G., Moyo, S., Mohammed, T., Ncube, T., Makhema, J., Jean-Philippe, P., Yu, X. G., Powis, K. M., Lockman, S., Kuritzkes, D. R., Shapiro, R., Lichterfeld, M. 2019; 11 (520)

    Abstract

    Neonatal HIV-1 infection is associated with rapidly progressive and frequently fatal immune deficiency if left untreated. Immediate institution of antiretroviral therapy (ART), ideally within hours after birth, may restrict irreversible damage to the developing neonatal immune system and possibly provide opportunities for facilitating drug-free viral control during subsequent treatment interruptions. However, the virological and immunological effects of ART initiation within hours after delivery have not been systematically investigated. We examined a unique cohort of neonates with HIV-1 infection from Botswana who started ART shortly after birth and were followed longitudinally for about 2 years in comparison to control infants started on treatment during the first year after birth. We demonstrate multiple clear benefits of rapid antiretroviral initiation, including an extremely small reservoir of intact proviral sequences, a reduction in abnormal T cell immune activation, a more polyfunctional HIV-1-specific T cell response, and an innate immune profile that displays distinct features of improved antiviral activity and is associated with intact proviral reservoir size. Together, these data offer rare insight into the evolutionary dynamics of viral reservoir establishment in neonates and provide strong empirical evidence supporting the immediate initiation of ART for neonates with HIV-1 infection.

    View details for DOI 10.1126/scitranslmed.aax7350

    View details for PubMedID 31776292

    View details for PubMedCentralID PMC8397898

  • HIV-1 DNA sequence diversity and evolution during acute subtype C infection. Nature communications Lee, G. Q., Reddy, K., Einkauf, K. B., Gounder, K., Chevalier, J. M., Dong, K. L., Walker, B. D., Yu, X. G., Ndung'u, T., Lichterfeld, M. 2019; 10 (1): 2737

    Abstract

    Little is known about the genotypic make-up of HIV-1 DNA genomes during the earliest stages of HIV-1 infection. Here, we use near-full-length, single genome next-generation sequencing to longitudinally genotype and quantify subtype C HIV-1 DNA in four women identified during acute HIV-1 infection in Durban, South Africa, through twice-weekly screening of high-risk participants. In contrast to chronically HIV-1-infected patients, we found that at the earliest phases of infection in these four participants, the majority of viral DNA genomes are intact, lack APOBEC-3G/F-associated hypermutations, have limited genome truncations, and over one year show little indication of cytotoxic T cell-driven immune selections. Viral sequence divergence during acute infection is predominantly fueled by single-base substitutions and is limited by treatment initiation during the earliest stages of disease. Our observations provide rare longitudinal insights of HIV-1 DNA sequence profiles during the first year of infection to inform future HIV cure research.

    View details for DOI 10.1038/s41467-019-10659-2

    View details for PubMedID 31227699

    View details for PubMedCentralID PMC6588551

  • Intact HIV-1 proviruses accumulate at distinct chromosomal positions during prolonged antiretroviral therapy. The Journal of clinical investigation Einkauf, K. B., Lee, G. Q., Gao, C., Sharaf, R., Sun, X., Hua, S., Chen, S. M., Jiang, C., Lian, X., Chowdhury, F. Z., Rosenberg, E. S., Chun, T. W., Li, J. Z., Yu, X. G., Lichterfeld, M. 2019; 129 (3): 988-998

    Abstract

    Chromosomal integration of genome-intact HIV-1 sequences into the host genome creates a reservoir of virally infected cells that persists throughout life, necessitating indefinite antiretroviral suppression therapy. During effective antiviral treatment, the majority of these proviruses remain transcriptionally silent, but mechanisms responsible for viral latency are insufficiently clear. Here, we used matched integration site and proviral sequencing (MIP-Seq), an experimental approach involving multiple displacement amplification of individual proviral species, followed by near-full-length HIV-1 next-generation sequencing and corresponding chromosomal integration site analysis to selectively map the chromosomal positions of intact and defective proviruses in 3 HIV-1-infected individuals undergoing long-term antiretroviral therapy. Simultaneously, chromatin accessibility and gene expression in autologous CD4+ T cells were analyzed by assays for transposase-accessible chromatin using sequencing (ATAC-Seq) and RNA-Seq. We observed that in comparison to proviruses with defective sequences, intact HIV-1 proviruses were enriched for non-genic chromosomal positions and more frequently showed an opposite orientation relative to host genes. In addition, intact HIV-1 proviruses were preferentially integrated in either relative proximity to or increased distance from active transcriptional start sites and to accessible chromatin regions. These studies strongly suggest selection of intact proviruses with features of deeper viral latency during prolonged antiretroviral therapy, and may be informative for targeting the genome-intact viral reservoir.

    View details for DOI 10.1172/JCI124291

    View details for PubMedID 30688658

    View details for PubMedCentralID PMC6391088

  • Effect of analytical treatment interruption and reinitiation of antiretroviral therapy on HIV reservoirs and immunologic parameters in infected individuals. PLoS pathogens Clarridge, K. E., Blazkova, J., Einkauf, K., Petrone, M., Refsland, E. W., Justement, J. S., Shi, V., Huiting, E. D., Seamon, C. A., Lee, G. Q., Yu, X. G., Moir, S., Sneller, M. C., Lichterfeld, M., Chun, T. W. 2018; 14 (1): e1006792

    Abstract

    Therapeutic strategies aimed at achieving antiretroviral therapy (ART)-free HIV remission in infected individuals are under active investigation. Considering the vast majority of HIV-infected individuals experience plasma viral rebound upon cessation of therapy, clinical trials evaluating the efficacy of curative strategies would likely require inclusion of ART interruption. However, it is unclear what impact short-term analytical treatment interruption (ATI) and subsequent reinitiation of ART have on immunologic and virologic parameters of HIV-infected individuals. Here, we show a significant increase of HIV burden in the CD4+ T cells of infected individuals during ATI that was correlated with the level of plasma viral rebound. However, the size of the HIV reservoirs as well as immune parameters, including markers of exhaustion and activation, returned to pre-ATI levels 6-12 months after the study participants resumed ART. Of note, the proportions of near full-length, genome-intact and structurally defective HIV proviral DNA sequences were similar prior to ATI and following reinitiation of ART. In addition, there was no evidence of emergence of antiretroviral drug resistance mutations within intact HIV proviral DNA sequences following reinitiation of ART. These data demonstrate that short-term ATI does not necessarily lead to expansion of the persistent HIV reservoir nor irreparable damages to the immune system in the peripheral blood, warranting the inclusion of ATI in future clinical trials evaluating curative strategies.

    View details for DOI 10.1371/journal.ppat.1006792

    View details for PubMedID 29324842

    View details for PubMedCentralID PMC5764487

  • Transcriptional Changes during Naturally Acquired Zika Virus Infection Render Dendritic Cells Highly Conducive to Viral Replication. Cell reports Sun, X., Hua, S., Chen, H. R., Ouyang, Z., Einkauf, K., Tse, S., Ard, K., Ciaranello, A., Yawetz, S., Sax, P., Rosenberg, E. S., Lichterfeld, M., Yu, X. G. 2017; 21 (12): 3471-3482

    Abstract

    Although dendritic cells are among the human cell population best equipped for cell-intrinsic antiviral immune defense, they seem highly susceptible to infection with the Zika virus (ZIKV). Using highly purified myeloid dendritic cells isolated from individuals with naturally acquired acute infection, we here show that ZIKV induces profound perturbations of transcriptional signatures relative to healthy donors. Interestingly, we noted a remarkable downregulation of antiviral interferon-stimulated genes and innate immune sensors, suggesting that ZIKV can actively suppress interferon-dependent immune responses. In contrast, several host factors known to support ZIKV infection were strongly upregulated during natural ZIKV infection; these transcripts included AXL, the main entry receptor for ZIKV; SOCS3, a negative regulator of ISG expression; and IDO-1, a recognized inducer of regulatory T cell responses. Thus, during in vivo infection, ZIKV can transform the transcriptome of dendritic cells in favor of the virus to render these cells highly conducive to ZIKV infection.

    View details for DOI 10.1016/j.celrep.2017.11.087

    View details for PubMedID 29262327

    View details for PubMedCentralID PMC5751936

  • Extensive virologic and immunologic characterization in an HIV-infected individual following allogeneic stem cell transplant and analytic cessation of antiretroviral therapy: A case study. PLoS medicine Cummins, N. W., Rizza, S., Litzow, M. R., Hua, S., Lee, G. Q., Einkauf, K., Chun, T. W., Rhame, F., Baker, J. V., Busch, M. P., Chomont, N., Dean, P. G., Fromentin, R., Haase, A. T., Hampton, D., Keating, S. M., Lada, S. M., Lee, T. H., Natesampillai, S., Richman, D. D., Schacker, T. W., Wietgrefe, S., Yu, X. G., Yao, J. D., Zeuli, J., Lichterfeld, M., Badley, A. D. 2017; 14 (11): e1002461

    Abstract

    Notwithstanding 1 documented case of HIV-1 cure following allogeneic stem cell transplantation (allo-SCT), several subsequent cases of allo-SCT in HIV-1 positive individuals have failed to cure HIV-1 infection. The aim of our study was to describe changes in the HIV reservoir in a single chronically HIV-infected patient on suppressive antiretroviral therapy who underwent allo-SCT for treatment of acute lymphoblastic leukemia.We prospectively collected peripheral blood mononuclear cells (PBMCs) by leukapheresis from a 55-year-old man with chronic HIV infection before and after allo-SCT to measure the size of the HIV-1 reservoir and characterize viral phylogeny and phenotypic changes in immune cells. At day 784 post-transplant, when HIV-1 was undetectable by multiple measures-including PCR measurements of both total and integrated HIV-1 DNA, replication-competent virus measurement by large cell input quantitative viral outgrowth assay, and in situ hybridization of colon tissue-the patient consented to an analytic treatment interruption (ATI) with frequent clinical monitoring. He remained aviremic off antiretroviral therapy until ATI day 288, when a low-level virus rebound of 60 HIV-1 copies/ml occurred, which increased to 1,640 HIV-1 copies/ml 5 days later, prompting reinitiation of ART. Rebounding plasma HIV-1 sequences were phylogenetically distinct from proviral HIV-1 DNA detected in circulating PBMCs before transplantation. The main limitations of this study are the insensitivity of reservoir measurements, and the fact that it describes a single case.allo-SCT led to a significant reduction in the size of the HIV-1 reservoir and a >9-month-long ART-free remission from HIV-1 replication. Phylogenetic analyses suggest that the origin of rebound virus was distinct from the viruses identified pre-transplant in the PBMCs.

    View details for DOI 10.1371/journal.pmed.1002461

    View details for PubMedID 29182633

    View details for PubMedCentralID PMC5705162

  • Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. The Journal of clinical investigation Lee, G. Q., Orlova-Fink, N., Einkauf, K., Chowdhury, F. Z., Sun, X., Harrington, S., Kuo, H. H., Hua, S., Chen, H. R., Ouyang, Z., Reddy, K., Dong, K., Ndung'u, T., Walker, B. D., Rosenberg, E. S., Yu, X. G., Lichterfeld, M. 2017; 127 (7): 2689-2696

    Abstract

    HIV-1 causes a chronic, incurable disease due to its persistence in CD4+ T cells that contain replication-competent provirus, but exhibit little or no active viral gene expression and effectively resist combination antiretroviral therapy (cART). These latently infected T cells represent an extremely small proportion of all circulating CD4+ T cells but possess a remarkable long-term stability and typically persist throughout life, for reasons that are not fully understood. Here we performed massive single-genome, near-full-length next-generation sequencing of HIV-1 DNA derived from unfractionated peripheral blood mononuclear cells, ex vivo-isolated CD4+ T cells, and subsets of functionally polarized memory CD4+ T cells. This approach identified multiple sets of independent, near-full-length proviral sequences from cART-treated individuals that were completely identical, consistent with clonal expansion of CD4+ T cells harboring intact HIV-1. Intact, near-full-genome HIV-1 DNA sequences that were derived from such clonally expanded CD4+ T cells constituted 62% of all analyzed genome-intact sequences in memory CD4 T cells, were preferentially observed in Th1-polarized cells, were longitudinally detected over a duration of up to 5 years, and were fully replication- and infection-competent. Together, these data suggest that clonal proliferation of Th1-polarized CD4+ T cells encoding for intact HIV-1 represents a driving force for stabilizing the pool of latently infected CD4+ T cells.

    View details for DOI 10.1172/JCI93289

    View details for PubMedID 28628034

    View details for PubMedCentralID PMC5490740