Citation: Tian-Jiao Fan, Li Sun, Xian-Guang Yang, Xia Jin, Wei-Wei Sun, Jian-Hua Wang. The Establishment of an In Vivo HIV-1 Infection Model in Humanized B-NSG Mice .VIROLOGICA SINICA, 2020, 35(4) : 417-425.  http://dx.doi.org/10.1007/s12250-019-00181-6

The Establishment of an In Vivo HIV-1 Infection Model in Humanized B-NSG Mice

  • Corresponding author: Wei-Wei Sun, wwsun@ips.ac.cn, ORCID: http://orcid.org/0000-0002-4578-6292
    Jian-Hua Wang, jh_wang@sibs.ac.cn, ORCID: http://orcid.org/0000-0002-6435-9907
  • Received Date: 12 July 2019
    Accepted Date: 02 December 2019
    Published Date: 21 December 2019
    Available online: 01 August 2020
  • Suitable animal models for human immunodeficiency virus type 1 (HIV-1) infection are important for elucidating viral pathogenesis and evaluating antiviral strategies in vivo. The B-NSG (NOD-PrkdcscidIl2rgtm1/Bcge) mice that have severe immune defect phenotype are examined for the suitability of such a model in this study. Human peripheral blood mononuclear cells (PBMCs) were engrafted into B-NSG mice via mouse tail vein injection, and the repopulated human T-lymphocytes were observed at as early as 3-weeks post-transplantation in mouse peripheral blood and several tissues. The humanized mice could be infected by HIV-1, and the infection recapitulated features of T-lymphocyte dynamic observed in HIV-1 infected humans, meanwhile the administration of combination antiretroviral therapy (cART) suppressed viral replication and restored T lymphocyte abnormalities. The establishment of HIV-1 infected humanized B-NSG mice not only provides a model to study virus and T cell interplays, but also can be a useful tool to evaluate antiviral strategies.

  • 加载中
    1. Brainard DM, Seung E, Frahm N, Cariappa A, Bailey CC, Hart WK, Shin HS, Brooks SF, Knight HL, Eichbaum Q, Yang YG, Sykes M, Walker BD, Freeman GJ, Pillai S, Westmoreland SV, Brander C, Luster AD, Tager AM (2009) Induction of robust cellular and humoral virus-specific adaptive immune responses in human immunodeficiency virus-infected humanized BLT mice. J Virol 83:7305–7321
        doi: 10.1128/JVI.02207-08

    2. Brooks DG, Hamer DH, Arlen PA, Gao L, Bristol G, Kitchen CM, Berger EA, Zack JA (2003) Molecular characterization, reactivation, and depletion of latent HIV. Immunity 19:413–423
        doi: 10.1016/S1074-7613(03)00236-X

    3. Chung YS, Son JK, Choi B, Joo SY, Lee YS, Park JB, Moon H, Kim TJ, Kim SH, Hong S, Chang J, Kang MS, Kim SJ (2015) Co-transplantation of human fetal thymus, bone and CD34(+) cells into young adult immunodeficient NOD/SCID IL2Rgamma(null) mice optimizes humanized mice that mount adaptive antibody responses. Clin Immunol 157:156–165
        doi: 10.1016/j.clim.2015.02.005

    4. Covassin L, Jangalwe S, Jouvet N, Laning J, Burzenski L, Shultz LD, Brehm MA (2013) Human immune system development and survival of non-obese diabetic (NOD)-SCID IL2rgamma(null) (NSG) mice engrafted with human thymus and autologous haematopoietic stem cells. Clin Exp Immunol 174:372–388
        doi: 10.1111/cei.12180

    5. Dinoso JB, Rabi SA, Blankson JN, Gama L, Mankowski JL, Siliciano RF, Zink MC, Clements JE (2009) A simian immunodeficiency virus-infected macaque model to study viral reservoirs that persist during highly active antiretroviral therapy. J Virol 83:9247–9257
        doi: 10.1128/JVI.00840-09

    6. Evans DT, Silvestri G (2013) Nonhuman primate models in aids research. Curr Opin HIV AIDS 8:255–261
        doi: 10.1097/COH.0b013e328361cee8

    7. Ganick DJ, Sarnwick RD, Shahidi NT, Manning DD (1980) Inability of intravenously injected monocellular suspensions of human bone marrow to establish in the nude mouse. Int Arch Allergy Appl Immunol 62:330–333
        doi: 10.1159/000232530

    8. Gruell H, Klein F (2017) Progress in HIV-1 antibody research using humanized mice. Curr Opin HIV AIDS 12:285–293
        doi: 10.1097/COH.0000000000000368

    9. Hessell AJ, Haigwood NL (2015) Animal models in HIV-1 protection and therapy. Curr Opin HIV AIDS 10:170–176
        doi: 10.1097/COH.0000000000000152

    10. Honeycutt JB, Wahl A, Archin N, Choudhary S, Margolis D, Garcia JV (2013) HIV-1 infection, response to treatment and establishment of viral latency in a novel humanized T cell-only mouse (TOM) model. Retrovirology 10:121
        doi: 10.1186/1742-4690-10-121

    11. Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, Watanabe T, Akashi K, Shultz LD, Harada M (2005) Development of functional human blood and immune systems in NOD/SCID/IL2 receptor gamma chain(null) mice. Blood 106:1565–1573
        doi: 10.1182/blood-2005-02-0516

    12. Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, Ueyama Y, Koyanagi Y, Sugamura K, Tsuji K, Heike T, Nakahata T (2002) NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood 100:3175–3182
        doi: 10.1182/blood-2001-12-0207

    13. Jiang Q, Zhang L, Wang R, Jeffrey J, Washburn ML, Brouwer D, Barbour S, Kovalev GI, Unutmaz D, Su L (2008) Foxp3 + CD4 + regulatory T cells play an important role in acute HIV-1 infection in humanized Rag2–/–gammac–/– mice in vivo. Blood 112:2858–2868
        doi: 10.1182/blood-2008-03-145946

    14. Kim KC, Choi BS, Kim KC, Park KH, Lee HJ, Cho YK, Kim SI, Kim SS, Oh YK, Kim YB (2016) A simple mouse model for the study of human immunodeficiency virus. AIDS Res Hum Retrovir 32:194–202
        doi: 10.1089/aid.2015.0211

    15. King M, Pearson T, Shultz LD, Leif J, Bottino R, Trucco M, Atkinson MA, Wasserfall C, Herold KC, Woodland RT, Schmidt MR, Woda BA, Thompson MJ, Rossini AA, Greiner DL (2008) A new Hu-PBL model for the study of human islet alloreactivity based on NOD-SCID mice bearing a targeted mutation in the IL-2 receptor gamma chain gene. Clin Immunol 126:303–314
        doi: 10.1016/j.clim.2007.11.001

    16. Kline C, Ndjomou J, Franks T, Kiser R, Coalter V, Smedley J, Piatak M Jr, Mellors JW, Lifson JD, Ambrose Z (2013) Persistence of viral reservoirs in multiple tissues after antiretroviral therapy suppression in a macaque RT-SHIV model. PLoS ONE 8:e84275
        doi: 10.1371/journal.pone.0084275

    17. Kumar N, Chahroudi A, Silvestri G (2016) Animal models to achieve an HIV cure. Curr Opin HIV AIDS 11:432–441
        doi: 10.1097/COH.0000000000000290

    18. Lan P, Tonomura N, Shimizu A, Wang S, Yang YG (2006) Reconstitution of a functional human immune system in immunodeficient mice through combined human fetal thymus/liver and CD34+ cell transplantation. Blood 108:487–492
        doi: 10.1182/blood-2005-11-4388

    19. Li C, Wang HB, Kuang WD, Ren XX, Song ST, Zhu HZ, Li Q, Xu LR, Guo HJ, Wu L, Wang JH (2017) Naf1 regulates HIV-1 latency by suppressing viral promoter-driven gene expression in primary CD4+ T cells. J Virol 91:e01830-16

    20. Lu W, Mehraj V, Vyboh K, Cao W, Li T, Routy JP (2015) CD4:CD8 ratio as a frontier marker for clinical outcome, immune dysfunction and viral reservoir size in virologically suppressed HIV-positive patients. J Int AIDS Soc 18:20052
        doi: 10.7448/IAS.18.1.20052

    21. Marsden MD, Kovochich M, Suree N, Shimizu S, Mehta R, Cortado R, Bristol G, An DS, Zack JA (2012) HIV latency in the humanized BLT mouse. J Virol 86:339–347
        doi: 10.1128/JVI.06366-11

    22. McBride JA, Striker R (2017) Imbalance in the game of T cells: what can the CD4/CD8 T-cell ratio tell us about HIV and health? PLoS Pathog 13:e1006624
        doi: 10.1371/journal.ppat.1006624

    23. McCune JM, Namikawa R, Kaneshima H, Shultz LD, Lieberman M, Weissman IL (1988) The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science 241:1632–1639
        doi: 10.1126/science.2971269

    24. McDermott SP, Eppert K, Lechman ER, Doedens M, Dick JE (2010) Comparison of human cord blood engraftment between immunocompromised mouse strains. Blood 116:193–200
        doi: 10.1182/blood-2010-02-271841

    25. Mosier DE, Gulizia RJ, Baird SM, Wilson DB (1988) Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature 335:256–259
        doi: 10.1038/335256a0

    26. Nixon CC, Mavigner M, Silvestri G, Garcia JV (2017) In vivo models of human immunodeficiency virus persistence and cure strategies. J Infect Dis 215:S142–S151
        doi: 10.1093/infdis/jiw637

    27. Satheesan S, Li H, Burnett JC, Takahashi M, Li S, Wu SX, Synold TW, Rossi JJ, Zhou J (2018) HIV replication and latency in a humanized NSG mouse model during suppressive oral combinational antiretroviral therapy. J Virol 92:e02118-17
        doi: 10.1128/JVI.02118-17

    28. Shultz LD, Schweitzer PA, Christianson SW, Gott B, Schweitzer IB, Tennent B, McKenna S, Mobraaten L, Rajan TV, Greiner DL et al (1995) Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol 154:180–191

    29. Shultz LD, Ishikawa F, Greiner DL (2007) Humanized mice in translational biomedical research. Nat Rev Immunol 7:118–130
        doi: 10.1038/nri2017

    30. Strowig T, Rongvaux A, Rathinam C, Takizawa H, Borsotti C, Philbrick W, Eynon EE, Manz MG, Flavell RA (2011) Transgenic expression of human signal regulatory protein alpha in Rag2–/–gamma(c)–/–mice improves engraftment of human hematopoietic cells in humanized mice. Proc Natl Acad Sci U S A 108:13218–13223
        doi: 10.1073/pnas.1109769108

    31. Sun Z, Denton PW, Estes JD, Othieno FA, Wei BL, Wege AK, Melkus MW, Padgett-Thomas A, Zupancic M, Haase AT, Garcia JV (2007) Intrarectal transmission, systemic infection, and CD4 + T cell depletion in humanized mice infected with HIV-1. J Exp Med 204:705–714
        doi: 10.1084/jem.20062411

    32. Wu X, Liu L, Cheung KW, Wang H, Lu X, Cheung AK, Liu W, Huang X, Li Y, Chen ZW, Chen SM, Zhang T, Wu H, Chen Z (2016) Brain invasion by CD4(+) T cells infected with a transmitted/founder HIV-1BJZS7 during acute stage in humanized mice. J Neuroimmune Pharmacol 11:572–583
        doi: 10.1007/s11481-016-9654-0

    33. Ye C, Wang W, Cheng L, Li G, Wen M, Wang Q, Zhang Q, Li D, Zhou P, Su L (2017) Glycosylphosphatidylinositol-anchored anti-HIV scFv efficiently protects CD4 T cells from HIV-1 infection and deletion in hu-PBL mice. J Virol 91:e10389-16

    34. Zhang L, Su L (2012) Hiv-1 immunopathogenesis in humanized mouse models. Cell Mol Immunol 9:237–244
        doi: 10.1038/cmi.2012.7

  • 加载中

Figures(4)

Article Metrics

Article views(5441) PDF downloads(31) Cited by()

Related
Proportional views

    The Establishment of an In Vivo HIV-1 Infection Model in Humanized B-NSG Mice

      Corresponding author: Wei-Wei Sun, wwsun@ips.ac.cn
      Corresponding author: Jian-Hua Wang, jh_wang@sibs.ac.cn
    • 1. CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
    • 2. University of Chinese Academy of Sciences, Beijing 100039, China
    • 3. College of Life Science, Henan Normal University, Xinxiang 453007, China

    Abstract: Suitable animal models for human immunodeficiency virus type 1 (HIV-1) infection are important for elucidating viral pathogenesis and evaluating antiviral strategies in vivo. The B-NSG (NOD-PrkdcscidIl2rgtm1/Bcge) mice that have severe immune defect phenotype are examined for the suitability of such a model in this study. Human peripheral blood mononuclear cells (PBMCs) were engrafted into B-NSG mice via mouse tail vein injection, and the repopulated human T-lymphocytes were observed at as early as 3-weeks post-transplantation in mouse peripheral blood and several tissues. The humanized mice could be infected by HIV-1, and the infection recapitulated features of T-lymphocyte dynamic observed in HIV-1 infected humans, meanwhile the administration of combination antiretroviral therapy (cART) suppressed viral replication and restored T lymphocyte abnormalities. The establishment of HIV-1 infected humanized B-NSG mice not only provides a model to study virus and T cell interplays, but also can be a useful tool to evaluate antiviral strategies.

    • Suitable human immunodeficiency virus type 1 (HIV-1) infected animal models assist the elucidation of viral pathogenesis and the evaluation of antiviral strategies. Currently, several models based on nonhuman primates (NHP) or humanized mice are available (Kumar et al. 2016). These NHPs, including Northern pig-tailed macaque (Macaca nemestrina) and Indian Rhesus monkey (Macaca mulatta), have been used to establish the acutely and persistently viral infection models by infection with simian immunodeficiency virus (SIV) or chimeric simian/human immunodeficiency virus (SHIV). These models are useful for studying viral transmission, pathogenesis, drugs, antibodies and candidate vaccines (Dinoso et al. 2009; Kline et al. 2013; Hessell and Haigwood 2015). Meanwhile, these models have several noted limitations, such as the inability to precisely examine human specific immune functions because of genetic differences in major histocompatibility complex (MHC) genes, the difficulty to operate sufficient sample size due to their expensive costs, and the mounting ethical concerns of experimentation with large animals (Evans and Silvestri 2013). To avoid the problems encountered in these NHP models, the humanized mouse models offer an alternative. The versatility of inbreeding allows mice to reproduce rapidly and produce strains with a clear genetic background and well defined immune systems (Shultz et al. 2007).

      To construct humanized mice, human PBMCs, stem cells, or lymphoid tissues have been used for transplantation into immunodeficient mice (Zhang and Su 2012). Three decades ago, mice with severe combine immune deficiency (SCID) that lacked of both T cells and B cells had been used for the reconstruction of human immune cells or system. SCID mice transplanted with human PBMCs can construct SCID-hu PBL (human peripheral blood leukocytes) mice (Ganick et al. 1980; Mosier et al. 1988), and those transplanted with human fetal liver and thymus can construct SCID-hu Thy/Liv mice (McCune et al. 1988). However, SCID-hu PBL mice lack human lymphatic organs and have been proved to cause graft versus host disease (GVHD), while SCID-hu Thy/Livmice have low frequency of human cells in their blood and peripheral organs.

      B-NSG (NOD-PrkdcscidIl2rgtm1/Bcge) mice have Nodscid-il2rg null gene background and a longer lifespan than NOD/SCID mice, and those mice have recently been experimented for humanization (Shultz et al. 1995; Ito et al. 2002). These B-NSG mice display severe immune defects, lacking mature T cells, B cells and functional NK cells due to a defective rearrangement of the T cell receptor and immunoglobulin genes. Therefore, these homozygous mice have been shown to be suitable for the transplantation and growth of human PBMCs and hematopoietic stem cells (HSCs) (Ishikawa et al. 2005; Strowig et al. 2011). The humanized B-NSG mice transplanted with bone marrow/liver/thymus (BLT) can afford differentiation of human HSCs into a variety of human cells, including lymphocytes, NK cells, monocytes, macrophages and dendritic cells, and these human cells can be detected in multiple tissues and organs, including blood, bone marrow, lymph nodes, spleen, thymus, liver, lung, digestive tract and reproductive tract (Lan et al. 2006; Brainard et al. 2009; Chung et al. 2015). Whereas, the B-NSG mice-based Hu-thy/liv model can only allow differentiation of human T cells (Honeycutt et al. 2013; Nixon et al. 2017). These humanized mice have been used for infection with HIV-1 to establish the acute and persist infection models to elucidate the processes of viral invasion, immuno-pathogenesis and to evaluate strategies for HIV-1 prevention, treatment and eradication (Brooks et al. 2003; Sun et al. 2007; Jiang et al. 2008; Brainard et al. 2009; Marsden et al. 2012; Zhang and Su 2012; Gruell and Klein 2017; Nixon et al. 2017). Meanwhile, the limitation of these mice models includes expensive experimental cost, delicate surgical procedures, and months of time required for reconstruction.

      In this study, we construct a rapid and convenient humanized mouse model of HIV-1 infection using B-NSG mice which were engrafted with human PBMCs via tail vein injection. We then demonstrated that the humanized mice can be infected by HIV-1, and the viral infection recapitulates the expected changes of T cell subsets in human HIV infection, while cART suppresses viral replication and restores T cell subset abnormalities.

    • NOD-PrkdcscidIl2rgtm1/Bcge (B-NSG) mice were purchased from Beijing Biocytogen and housed in a pathogenfree animal facility at Institut Pasteur of Shanghai. Human peripheral blood buffy coats were purchased from Changhai Hospital, Shanghai, China, and PBMCs were isolated from the buffy coats of healthy donors by Ficoll-density gradient centrifugation as described previously (Li et al. 2017). All procedures were conducted in compliance with a protocol approved by the Institutional Animal Care and Use Committee at Institut Pasteur of Shanghai. All experiments were performed in accordance with relevant guidelines and regulations.

    • 5-weeks old female B-NSG mice were transplanted with human healthy PBMCs (5 × 106 cells/mouse) via tail intravenous injection. Mice were humanized with PBMCs from different donors separately. The peripheral blood from the retro-orbital sinus was collected each week after transplantation and the differentiation of human CD45+ cells were monitored by flow cytometry. For HIV-1 infection, humanized mice with 3-weeks human PBMCs transplantation were selected for infection with HIV-1/JRCSF (CCR5-tropism) (10 ng p24Gag/mouse) by intraperitoneal injection. Viral replication in plasma and tissues of humanized mice was quantified by quantitative real-time PCR (qRT-PCR) to detect the production of gag mRNA.

      At 4-weeks post-infection (w.p.i.), mice were treated with combination antiretroviral therapy (cART). cART regimens are composed of nucleoside reverse transcriptase inhibitor Tenofovir disoproxil fumarate (TDF; 205 mg/kg), nucleoside reverse transcriptase inhibitor Emtricitabine (FTC; 211 mg/kg) and integrase inhibitor Raltegravir (RAL; 80 mg/kg) (Satheesan et al. 2018). Triple combination drugs were dissolved in 3% DMSO, 40% PEG-400, 2% Tween-80 and 56% ddH2O. Mice were administered daily via intraperitoneal injection.

    • Total cellular mRNAs from plasma and tissues were extracted using the QiaAmp Viral RNA Mini Kit (Qiagen, Germany) and then reverse transcribed into cDNA using the ReverTra Ace qPCR RT Master Mix with gDNA Remover Kit (Toyobo, Japan). qRT-PCR was performed on the ABI 7900HT Real-Time PCR system (Applied Biosystems, United states) (50× ROX and 2× Gold Taq-Man Mixture; CMBIO), with an initial denaturation step at 95 ℃ for 10 min, amplification with 40 cycles of denaturation (95 ℃, 15 s) and annealing (60 ℃, 1 min). HIV-1 gag primers were used: forward primer, 5′-GGT GCG AGA GCG TCA GTA TTA AG-3′, reverse primer 5′-AGC TCC CTG CTT GCC CAT A-3′, and probe, 5′-FAM-AAA ATT CGG TTA AGG CCA GGA GGA AAG AATAMRA-3′ (Ye et al. 2017).

    • Peripheral blood was collected from retro-orbit. Spleen, bone marrow and liver were gained at necropsy to make the single cell suspension. Red blood cells were eliminated by using Red Cell Lysis buffer solution (Multi Sciences, Hangzhou, China). Monoclonal antibodies against these specific antigens of human and mouse cells were listed as follows: APC-mouse CD45 (30-F11, eBioscience, United states), FITC-human CD45 (HI30, eBioscience), PE-human CD3 (UCHT1, eBioscience), percpcy5.5-human CD4 (HIB19, eBioscience), Brilliant Violet 421-human CD8 (RPA-T8, BD Biosciences), APC-cy7-human CCR5(2D7, eBioscience), APC-human CXCR4 (12G5; BD Pharmingen, United states). The stained cells were detected using a Fortessa flow cytometer (BD Pharmingen) and analyzed with FlowJo 7.6.1. software (United states).

    • This software of GraphPad Prism version 7.0. (San Diego, California, United states) was used to perform paired t tests to analyze statistically significant differences.

    • Five weeks old female B-NSG mice were transplanted with healthy human PBMCs via tail vein injection (Fig. 1A). The human CD45+ (hCD45+) leucocytes in mouse peripheral blood were monitored by flow cytometry over time. At 4–7 weeks post transplantation, the number of hCD45+ leucocytes reached a peak, and then declined but remained detectable for an additional 4–5 weeks (Fig. 1B). As shown in a representative mouse, hCD45+ cell population in peripheral blood compromises of both CD4+ and CD8+ T cell subsets. Among the 82.4% of hCD45+ cells, the majority were CD3+ T cells (93.8%), of which, CD4+ and CD8+ T-lymphocytes account for 50.2% and 41.8%, respectively (Fig. 1C). The reconstruction of human B cells and myeloid cells was not observed (data not shown). Taken together, these data show the successful reconstruction of human T-lymphocyte subsets in the peripheral blood of B-NSG mice transplanted with human PBMCs.

      Figure 1.  Establishment of humanized B-NSG mice. A Schematic diagram showing the construction of humanized B-NSG mice. 5-weeks old female B-NSG mice were transplanted with human healthy PBMCs (5 × 106 cells/mouse) via tail intraven ous injection, peripheral blood cells were harvested each week after transplantation for detecting mCD45+ and hCD45+ cells with flow cytometry using specific antibodies. B The dynamic of hCD45+ cell population in peripheral blood of humanized mice. Each mouse was humanized with PBMCs from different donors separately, and eight representative mice were shown (n = 8). C The reconstruction of human T-lymphocyte subsets. Peripheral blood cells from one representative mouse with 5-week transplantation were collected, immunostainings were performed using specific antibodies and detected by flow cytometry. SSC-A, side-scattered light area.

    • Next, we examined the susceptibility of these humanized mice for HIV-1 infection. These B-NSG mice with reconstruction of human T-lymphocyte were intraperitoneally (i.p.) injected with HIV-1 (Fig. 2A). Because these reconstructed hCD4+ T cells expressed HIV-1 co-receptor CCR5 (10%–33%), but not CXCR4 (less than 1%) (Fig. 2B), the CCR5-tropic virus HIV-1/JR-CSF was used for infection (Fig. 2A). Viral replication in plasma was monitored longitudinally by qRT-PCR detection for gag gene expression. Results showed a rapid viral replication during the first-week of infection, and viremia reached a steady peak level for the next few weeks (Fig. 2C). The adminstration of a cocktail of TDF/FTC/RAL at 3 w.p.i. markedly suppressed viral replication and reduced the plasma viral load to undetectable level within 1 week of treatment (Fig. 2C). Neither infection nor cART-treatment obviously affected mouse body weight (Fig. 2D). Taken together, these data demonstrate that humanized B-NSG mice can be successfully infected by HIV-1 and viral replication can be suppressed by cART-treatment, and thereby can be used as an in vivo model for studying the establishment of HIV infection and antiretroviral treatment outcome.

      Figure 2.  HIV-1 infection of humanized B-NSG mice and cART treatments. A Schematic illustration of HIV-1 infection of humanized B-NSG mice and cART treatments. B-NSG mice with 3-week transplantation were i.p. infected with HIV-1/JR-CSF (10 ng p24Gag/mouse), and 4 w.p.i, cART regimens (TDF/FTC/RAL) was administered daily by i.p. injection and lasted overtime of surveillance. Viral load and cell subsets from retro-orbital blood sampling were monitored. B Assay of HIV-1 co-receptor expression. Cells were immunostained with specific antibodies and detected with flow cytometry, and the expressions of CXCR4 and CCR5 in hCD3+ hCD4+ hCD45+ population were calculated. C Viral load in plasma of humanized mice was quantified by real-time (RT-) PCR to detect the production of gag mRNA. The shaded area indicates cART-treatment. D Mice weights were monitored.

    • HIV-1 infection of human individuals can induce a rapid CD4+ T cell decline and a reverse of CD4+/CD8+ T cell ratio (Lu et al. 2015), and the cART treatment in most cases can normalize these changes (McBride and Striker 2017). Therefore, we next investigated whether the HIV-1 infected-humanized mice could be used to feature these changes. Results from one representative mouse showed that, after being transplanted with human PBMCs for 3 weeks and then infected with HIV-1/JR-CSF for an additional 4 weeks, hCD4+ T cell frequency reduced from 78.5% (before infection) to 12.1% (after infection), and hCD8+ T cell frequency increased from 19.6% to 80.4%. cART-treatment for an additional 3-weeks recovered hCD4+ T cell frequency to 45.9% and reduced hCD8+ T cell frequency to 38.5% (Fig. 3A). Similar results were obtained from four HIV-1 infected-humanized mice (Fig. 3B). HIV-1 infection reversed blood hCD4+/CD8+ ratio in mice, whereas cART-treatment normalized the ratio (Fig. 3C). The dynamic changes of hCD4+ and hCD8+ T cells over time of infection and treatments were monitored (Fig. 3D). HIV-1 infection induced a typically rapid decline of hCD4+ T cells and a compensatory increase of hCD8+ T cells, and the cART-treatment normalized these parameters (Fig. 3D). To sum up, these data demonstrate that HIV-1 infection of humanized mice can recapitulate typical CD4+ T cell and CD8+ T changes observed in human infections, and such abnormality of T cell dynamics can be reversed by cART-treatment, suggesting such a model could be used to examine HIV-1 pathogenesis in vivo.

      Figure 3.  The dynamic of human T-lymphocytes in HIV-1 infectedhumanized mice. A The longitude detection of hCD4+ and hCD8+ T cell. The peripheral blood cells from one representative mouse were harvested at the different times and further analyzed with flow cytometry. B, C The frequencies of hCD4+ and hCD8+ T cell from four humanized mice were summarized (B) and the hCD4+/CD8+ T cell ratio was calculated (C). D The dynamic changes of hCD4+ and hCD8+ T cells over time of infection and cART-treatment. The shaded area indicates cART-treatment. Data are presented as mean ± standard deviation. *P < 0.05 and **P < 0.01 are considered as significant differences in paired t test.

    • Having demonstrated the recapitulation of T cell dynamic features of HIV infection in human peripheral blood, we went on to investigate whether the B-NSG mice can also be used to outline major features of human HIV-1 infection in tissues and whether primary infection starts and latent reservoir virus may form. The new experimental procedures include harvesting tissue samples in addition to the transplantation, infection, cART treatment steps as performed in previous experiments (Fig. 4A). About 2% of hCD45+ cell subset was observed in liver, whereas 20%– 80% of hCD45+ cell population were observed in spleen and bone marrow (BM) after PBMC transplantation, and these numbers were not altered significantly by HIV-1 infection and cART-treatment (Fig. 4B). By quantifying viral RNA copies at 4 w.p.i., HIV-1 replication was observed in spleen and BM, suggesting a successful infection happened; and viral replication could be suppressed to below the level of detection after 3-weeks cART-treatment (Fig. 4C). Accompanied with a successful virological response to treatment, there was limited reconstruction of CD4+ and CD8+ T cell subsets in liver (less than 2% of the whole cells population), but greater reconstruction in spleen and BM. In spleen, CD3+ and CD8+ T subsets displayed 15%–35% and 10% reconstruction, respectively; in BM, CD3+ and CD8+ T subsets displayed 8% and 4% reconstruction, respectively (Fig. 4D). Similar to that being observed in the peripheral blood, HIV-1 infection for 4-weeks reduced CD3+ T cell but increased CD8+ T-cell frequency, and cART-treatment for 3-weeks partially normalized these cell population (Fig. 4D). Taken together, these results demonstrate that the HIV-1 infection-induced changes of human T-lymphocytes can also be recapitulated in several tissues of HIV-1 infected B-NSG mice.

      Figure 4.  The reconstruction of human T-lymphocytes in tissues of B-NSG mice and viral infection. A Schematic illustration of HIV-1 infection, cART treatment and tissue harvest. B Reconstruction evaluation by detecting hCD45+ cell. Humanized mice were infected by HIV-1 and then treated with cART as above, and the hCD45+ cells in different tissues were detected with flow cytometry. C Viral replication in different tissues was quantified by real-time (RT-) PCR to detect the production of gag mRNA. D The distribution of hCD4+ and hCD8+ T cells in different tissues was evaluated. Data are presented as mean ± standard deviation. *P < 0.05 and ***P < 0.001 are considered as significant differences in paired t test.

    • Humanized mouse models have been widely used to investigate human haematopoiesis, innate and adaptive immunity, autoimmunity, infectious diseases, cancer biology and regenerative medicine (Shultz et al. 2007). In the HIV-1 field, the BLT and Hu-thy/liv mice have previously been used to elucidate viral immuno-pathogenesis and to evaluate strategies for HIV-1 prevention and treatment. These humanized mouse models need to be implanted a piece of fetal liver tissue between two pieces of fetal thymus like sandwiches. Although those models have a more complete thymus development process and can generate an adaptive immune response, the technical and traumatic surgery, the acquisition of human tissue and the over a 2-month rebuilding cycle bring many difficulties.

      The B-NSG mice used in this study have severe immune defect phenotype and are suitable for the transplantation and growth of human PBMCs and HSCs (Ishikawa et al. 2005; Strowig et al. 2011). In this study, we construct a more convenient model of HIV-1 infection using humanized B-NSG mice. At as early as 3-weeks post-transplantation, these mice showed repopulation of human CD45+ cells in peripheral blood, and these human CD45+ cells were mainly comprised of CD4+ and CD8+ T cells. These reconstructed CD4+ T cells mainly expressed the HIV-1 co-receptor CCR5, and the infection of these humanized mice with CCR5-tropic HIV-1/JR-CSF induced viremia within 1-week. HIV-1 infection induced a rapid CD4+ T cell depletion and a CD8+ T cell increase, which recapitulated the major features of acute HIV-1 infection in humans. Therefore, these HIV-1 infected B-NSG mice - provide a model suitable for studying HIV-1 induced CD4+ T cell decline and evaluating blocking strategies for HIV-1 transmission. Moreover, our data show that the administration of cART suppressed viral replication to an undetected level. Therefore, our mouse model may have potential to use to evaluate HIV-1 latency-reversing agents and strategies for latency eradication. Furthermore, the reconstruction of the human CD4+ and CD8+ T cells in spleen and BM also support the usage of this model for studying viral dynamic and host immune responses in tissues.

      In fact, transplanting human PBMCs to construct HIV-1 infected mice models have been reported (Kim et al. 2016; Wu et al. 2016). In the Wu's and Kim's models, the NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ mice were used, and i.p. injection for engraft was adopted. In our study, the B-NSG (NOD-PrkdcscidIl2rgtm1/Bcgen) mice was used, which are more suitable for the transplantation and growth of human PBMCs and HSCs. Further, we used i.v. injection of PBMCs, instead of their i.p. route. The i.v route has advantages for humanization, by which a significantly higher level of human CD45 cells engraftment can be acquired than that of through i.p. route. The i.v. injection can directly put human PBMCs into the circulation system, whereas for i.p. injection, a period of 7–14 days is required for human PBMCs to drain from the peritoneal cavity into the circulation. Thus, i.v. injection has the ability to engraft higher levels of human PBMCs at lower donor cell doses (5 × 106 cells/mouse used in our study), permitting the better reconstruction; further, i.v. injection permits immediate interaction between the human immune system with the allograft (King et al. 2008). Additionally, for cART-treatment to suppress HIV-1 infection, we used i.p. injection of drugs instead of oral administration in Kim's model, guaranteeing the same dose of drugs per mouse per day.

      To suppress HIV-1 infection in our mice model, the cART regimens composed of nucleoside reverse transcriptase inhibitor TDF, nucleoside reverse transcriptase inhibitor FTC and integrase inhibitor RAL were used. The triple combination of TDF/FTC/RAL is currently used for clinical treatment of patients, therefore, to recapitulate patient treatment, this drug-combination has been widely used in the HIV-infected humanized mice models (Honeycutt et al. 2013; Satheesan et al. 2018).

      We appreciate that the humanized mouse model also has deficiencies. For example, the engrafted human PBMCs can only differentiate human T cells, not other cells which may affect T cells' functions. A modified strategy that uses human CD34+ hematopoietic stem cell for injection may provide an opportunity to markedly improve the reconstitution of other elements of the human immune system both in tissues and peripheral blood (McDermott et al. 2010; Covassin et al. 2013).

      In summary, in this study, we construct a rapid and convenient humanized mouse model for HIV-1 infection using B-NSG mice. The establishment of HIV-1 infected humanized B-NSG mice not only provides a model to study virus invasion-induced T cell dynamic changes but also offers an effective tool for evaluating antiviral strategies.

    • This work was supported by Grants to JHW from the National Grant Program on Key Infectious Disease (2018ZX10301101-003-002), the Natural Science Foundation of China (NSFC, 81572001, 81873965), the key project from Chinese Academy of Sciences (QYZDB-SSW-SMC059), and grant to WWS from NSFC (31800152). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    • TJF, LS and WWS conceived the experiments, designed the experimental flow and performed the experiments. TJF performed statistical analyses. TJF, WWS and JHW wrote the manuscript. TJF, XGY and XJ reviewed the manuscript. WWS and JHW supervised the project. All authors read and approved the final manuscript.

    • The authors declare that they have no conflict of interest.

    • All procedures were conducted in compliance with a protocol approved by the Institutional Animal Care and Use Committee at Institut Pasteur of Shanghai. All experiments were performed in accordance with relevant guidelines and regulations.

    Figure (4)  Reference (34) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return