Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39026699

ABSTRACT

Broadly neutralizing antibodies (bNAbs) have shown great promise for prevention and treatment of HIV infection. Breadth of bNAb neutralization, measured in vitro across panels of diverse viral isolates, is often used as a predictor of clinical potential. However, recent prevention studies demonstrate that the clinical efficacy of a broad and potent bNAb (VRC01) is undermined by neutralization resistance of circulating strains. Using HIV-infected humanized mice, we find that therapeutic efficacy of bNAbs delivered as Vectored ImmunoTherapy (VIT) is a function of both the fitness cost and resistance benefit of mutations that emerge during viral escape, which we term 'escapability'. Applying this mechanistic framework, we find that the sequence of the envelope V5-loop alters the resistance benefits of mutants that arise during escape, thereby impacting the therapeutic efficacy of VIT-mediated viral suppression. We also find that an emtricitabine-based antiretroviral drug regimen dramatically enhances the efficacy of VIT, by reducing the fitness of mutants along the escape path. Our findings demonstrate that bNAb escapability is a key determinant to consider in the rational design of antibody regimens with maximal efficacy and illustrates a tractable means of minimizing viral escape from existing bNAbs.

2.
Sci Transl Med ; 14(655): eabn9662, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35895834

ABSTRACT

HIV broadly neutralizing antibodies (bNAbs) are capable of both blocking viral entry and driving innate immune responses against HIV-infected cells through their Fc region. Vaccination or productive infection results in a polyclonal mixture of class-switched immunoglobulin G (IgG) antibodies composed of four subclasses, each encoding distinct Fc regions that differentially engage innate immune functions. Despite evidence that innate immunity contributes to protection, the relative contribution of individual IgG subclasses is unknown. Here, we used vectored immunoprophylaxis in humanized mice to interrogate the efficacy of individual IgG subclasses during prevention of vaginal HIV transmission by VRC07, a potent CD4-binding site-directed bNAb. We find that VRC07 IgG2, which lacks Fc-mediated functionality, exhibited substantially reduced protection in vivo relative to other subclasses. Low concentrations of highly functional VRC07 IgG1 yielded substantial protection against vaginal challenge, suggesting that interventions capable of eliciting modest titers of functional IgG subclasses may provide meaningful benefit against infection.


Subject(s)
HIV Infections , Immunoglobulin G , Animals , Antibodies, Neutralizing , Broadly Neutralizing Antibodies , Female , HIV Antibodies , HIV Infections/prevention & control , Mice , Vagina
4.
Immunity ; 54(2): 235-246.e5, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33357409

ABSTRACT

The interleukin-6 (IL-6) membrane receptor and its circulating soluble form, sIL-6R, can be targeted by antibody therapy to reduce deleterious immune signaling caused by chronic overexpression of the pro-inflammatory cytokine IL-6. This strategy may also hold promise for treating acute hyperinflammation, such as observed in coronavirus disease 2019 (COVID-19), highlighting a need to define regulators of IL-6 homeostasis. We found that conventional dendritic cells (cDCs), defined in mice via expression of the transcription factor Zbtb46, were a major source of circulating sIL-6R and, thus, systemically regulated IL-6 signaling. This was uncovered through identification of a cDC-dependent but T cell-independent modality that naturally adjuvants plasma cell differentiation and antibody responses to protein antigens. This pathway was then revealed as part of a broader biological buffer system in which cDC-derived sIL-6R set the in-solution persistence of IL-6. This control axis may further inform the development of therapeutic agents to modulate pro-inflammatory immune reactions.


Subject(s)
Dendritic Cells/immunology , Interleukin-6/blood , Interleukin-6/immunology , ADAM17 Protein , Animals , Cell Differentiation , Immunity, Humoral , Immunoglobulin M/immunology , Inflammation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-6/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/immunology , Receptors, Interleukin-6/blood , Receptors, Interleukin-6/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 7/immunology
5.
Nat Med ; 26(11): 1776-1787, 2020 11.
Article in English | MEDLINE | ID: mdl-32868878

ABSTRACT

An effective strategy to cure HIV will likely require a potent and sustained antiviral T cell response. Here we explored the utility of chimeric antigen receptor (CAR) T cells, expressing the CD4 ectodomain to confer specificity for the HIV envelope, to mitigate HIV-induced pathogenesis in bone marrow, liver, thymus (BLT) humanized mice. CAR T cells expressing the 4-1BB/CD3-ζ endodomain were insufficient to prevent viral rebound and CD4+ T cell loss after the discontinuation of antiretroviral therapy. Through iterative improvements to the CAR T cell product, we developed Dual-CAR T cells that simultaneously expressed both 4-1BB/CD3-ζ and CD28/CD3-ζ endodomains. Dual-CAR T cells exhibited expansion kinetics that exceeded 4-1BB-, CD28- and third-generation costimulated CAR T cells, elicited effector functions equivalent to CD28-costimulated CAR T cells and prevented HIV-induced CD4+ T cell loss despite persistent viremia. Moreover, when Dual-CAR T cells were protected from HIV infection through expression of the C34-CXCR4 fusion inhibitor, these cells significantly reduced acute-phase viremia, as well as accelerated HIV suppression in the presence of antiretroviral therapy and reduced tissue viral burden. Collectively, these studies demonstrate the enhanced therapeutic potency of a novel Dual-CAR T cell product with the potential to effectively treat HIV infection.


Subject(s)
CD4 Antigens/immunology , HIV Infections/therapy , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Bone Marrow/immunology , Bone Marrow/virology , CD3 Complex/antagonists & inhibitors , CD4 Antigens/administration & dosage , Gene Expression Regulation/immunology , HIV Envelope Protein gp41/antagonists & inhibitors , HIV Envelope Protein gp41/immunology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Humans , Liver/immunology , Liver/virology , Mice , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Protein Domains/immunology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Receptors, Chimeric Antigen/administration & dosage , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/virology , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
6.
J Virol ; 93(20)2019 10 15.
Article in English | MEDLINE | ID: mdl-31375576

ABSTRACT

BLT (bone marrow-liver-thymus) humanized mice, which reconstitute a functional human immune system, develop prototypic human virus-specific CD8+ T cell responses following infection with human immunodeficiency virus type 1 (HIV-1). We explored the utility of the BLT model for HIV-1 vaccine development by immunizing BLT mice against the conserved viral Gag protein, utilizing a rapid prime-boost protocol of poly(lactic-co-glycolic) acid microparticles and a replication-defective herpes simplex virus (HSV) recombinant vector. After HIV-1 challenge, the mice developed broad, proteome-wide gamma interferon-positive (IFN-γ+) T cell responses against HIV-1 that reached magnitudes equivalent to what is observed in HIV-1-infected individuals. The functionality of these responses was underscored by the consistent emergence of escape mutations in multiple CD8+ T cell epitopes during the course of infection. Although prechallenge vaccine-induced responses were largely undetectable, the Gag immunization increased both the magnitude and the kinetics of anamnestic Gag-specific T cell responses following HIV-1 infection, and the magnitude of these postchallenge Gag-specific responses was inversely correlated with acute HIV-1 viremia. Indeed, Gag immunization was associated with a modest but significant 0.5-log reduction in HIV-1 viral load when analyzed across four experimental groups of BLT mice. Notably, the HSV vector induced elevated plasma concentrations of polarizing cytokines and chemotactic factors, including interleukin-12p70 (IL-12p70) and MIP-1α, which were positively correlated with the magnitude of Gag-specific responses. Overall, these results support the ability of BLT mice to recapitulate human pathogen-specific T cell responses and to respond to immunization; however, additional improvements to the model are required to develop a robust system for testing HIV-1 vaccine efficacy.IMPORTANCE Advances in the development of humanized mice have raised the possibility of a small-animal model for preclinical testing of an HIV-1 vaccine. Here, we describe the capacity of BLT humanized mice to mount broadly directed HIV-1-specific human T cell responses that are functionally active, as indicated by the rapid emergence of viral escape mutations. Although immunization of BLT mice with the conserved viral Gag protein did not result in detectable prechallenge responses, it did increase the magnitude and kinetics of postchallenge Gag-specific T cell responses, which was associated with a modest but significant reduction in acute HIV-1 viremia. Additionally, the BLT model revealed immunization-associated increases in the plasma concentrations of immunomodulatory cytokines and chemokines that correlated with more robust T cell responses. These data support the potential utility of the BLT humanized mouse for HIV-1 vaccine development but suggest that additional improvements to the model are warranted.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viremia , gag Gene Products, Human Immunodeficiency Virus/immunology , Acute Disease , Animals , Biological Evolution , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Disease Models, Animal , HIV Infections/metabolism , Host-Pathogen Interactions , Humans , Immunization , Mice , Mice, Transgenic , T-Lymphocytes/metabolism , Viral Load
7.
Nat Biotechnol ; 37(3): 293-302, 2019 03.
Article in English | MEDLINE | ID: mdl-30742125

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for multiple disorders, but deficiency and dysregulation of T cells limit its utility. Here we report a biomaterial-based scaffold that mimics features of T cell lymphopoiesis in the bone marrow. The bone marrow cryogel (BMC) releases bone morphogenetic protein-2 to recruit stromal cells and presents the Notch ligand Delta-like ligand-4 to facilitate T cell lineage specification of mouse and human hematopoietic progenitor cells. BMCs subcutaneously injected in mice at the time of HSCT enhanced T cell progenitor seeding of the thymus, T cell neogenesis and diversification of the T cell receptor repertoire. Peripheral T cell reconstitution increased ~6-fold in mouse HSCT and ~2-fold in human xenogeneic HSCT. Furthermore, BMCs promoted donor CD4+ regulatory T cell generation and improved survival after allogeneic HSCT. In comparison to adoptive transfer of T cell progenitors, BMCs increased donor chimerism, T cell generation and antigen-specific T cell responses to vaccination. BMCs may provide an off-the-shelf approach for enhancing T cell regeneration and mitigating graft-versus-host disease in HSCT.


Subject(s)
Bone Marrow Transplantation , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Regulatory/immunology , Tissue Scaffolds , Adoptive Transfer/methods , Animals , Bone Marrow , Chimerism , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Mice , T-Lymphocytes, Regulatory/cytology , Transplantation, Heterologous/methods , Transplantation, Homologous
8.
Cell Rep ; 26(6): 1409-1418.e5, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30726727

ABSTRACT

Lung interstitial CD4+ T cells are critical for protection against pulmonary infections, but the fate of this population during HIV-1 infection is not well described. We studied CD4+ T cells in the setting of HIV-1 infection in human lung tissue, humanized mice, and a Mycobacterium tuberculosis (Mtb)/simian immunodeficiency virus (SIV) nonhuman primate co-infection model. Infection with a CCR5-tropic strain of HIV-1 or SIV results in severe and rapid loss of lung interstitial CD4+ T cells but not blood or lung alveolar CD4+ T cells. This is accompanied by high HIV-1 production in these cells in vitro and in vivo. Importantly, during early SIV infection, loss of lung interstitial CD4+ T cells is associated with increased dissemination of pulmonary Mtb infection. We show that lung interstitial CD4+ T cells serve as an efficient target for HIV-1 and SIV infection that leads to their early depletion and an increased risk of disseminated tuberculosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Coinfection/immunology , HIV Infections/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Coinfection/pathology , Female , HEK293 Cells , HIV Infections/pathology , HIV-1/pathogenicity , Humans , Lung/immunology , Lung/microbiology , Lung/pathology , Lung/virology , Macaca mulatta , Mice , Mycobacterium tuberculosis/pathogenicity , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/pathogenicity , Tuberculosis, Pulmonary/pathology
9.
Cell Host Microbe ; 25(1): 73-86.e5, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30629922

ABSTRACT

HIV-1 primarily infects T lymphocytes and uses these motile cells as migratory vehicles for effective dissemination in the host. Paradoxically, the virus at the same time disrupts multiple cellular processes underlying lymphocyte motility, seemingly counterproductive to rapid systemic infection. Here we show by intravital microscopy in humanized mice that perturbation of the actin cytoskeleton via the lentiviral protein Nef, and not changes to chemokine receptor expression or function, is the dominant cause of dysregulated infected T cell motility in lymphoid tissue by preventing stable cellular polarization required for fast migration. Accordingly, disrupting the Nef hydrophobic patch that facilitates actin cytoskeletal perturbation initially accelerates systemic viral dissemination after female genital transmission. However, the same feature of Nef was subsequently critical for viral persistence in immune-competent hosts. Therefore, a highly conserved activity of lentiviral Nef proteins has dual effects and imposes both fitness costs and benefits on the virus at different stages of infection.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Movement , HIV Infections/transmission , HIV-1/physiology , HIV-1/pathogenicity , Mucous Membrane/metabolism , Actins/metabolism , Animals , Chemokines/metabolism , Disease Models, Animal , Female , HEK293 Cells , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Human Immunodeficiency Virus Proteins/metabolism , Humans , Lymphocytes/virology , Mice , Mucous Membrane/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Regulatory and Accessory Proteins/metabolism , Viremia , nef Gene Products, Human Immunodeficiency Virus/immunology , nef Gene Products, Human Immunodeficiency Virus/metabolism , p21-Activated Kinases/metabolism
10.
J Infect Dis ; 218(3): 471-475, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29617845

ABSTRACT

Background: Small-molecule CD4-mimetic compounds (CD4mc) inhibit human immunodeficiency virus (HIV-1) entry by blocking binding to the CD4 receptor and by premature triggering of the viral envelope glycoprotein (Env) spike. Methods: The efficacy of a CD4mc in protecting bone marrow-liver-thymus (BLT) humanized mice from vaginal HIV-1 challenge was evaluated. Results: Intravaginal application of the CD4mc JP-III-48, either before or simultaneously with virus challenge, protected BLT humanized mice from HIV-1JR-CSF infection in a dose- dependent manner. Conclusion: The direct antiviral effects of a CD4mc prevent HIV-1 infection in a murine model of sexual transmission.


Subject(s)
Biomimetics , CD4 Antigens/administration & dosage , HIV Fusion Inhibitors/administration & dosage , HIV Infections/prevention & control , HIV-1/drug effects , Administration, Intravaginal , Animals , Bone Marrow , Disease Models, Animal , Female , Liver , Mice, SCID , Thymus Gland , Treatment Outcome
11.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29321310

ABSTRACT

Broadly neutralizing antibodies (bNAbs) are being explored for HIV-1 prevention and cure strategies. However, administration of purified bNAbs poses challenges in resource-poor settings, where the HIV-1 disease burden is greatest. In vivo vector-based production of bNAbs represents an alternative strategy. We investigated adenovirus serotype 5 (Ad5) and adeno-associated virus serotype 1 (AAV1) vectors to deliver the HIV-1-specific bNAb PGT121 in wild-type and immunocompromised C57BL/6 mice as well as in HIV-1-infected bone marrow-liver-thymus (BLT) humanized mice. Ad5.PGT121 and AAV1.PGT121 produced functional antibody in vivo Ad5.PGT121 produced PGT121 rapidly within 6 h, whereas AAV1.PGT121 produced detectable PGT121 in serum by 72 h. Serum PGT121 levels were rapidly reduced by the generation of anti-PGT121 antibodies in immunocompetent mice but were durably maintained in immunocompromised mice. In HIV-1-infected BLT humanized mice, Ad5.PGT121 resulted in a greater reduction of viral loads than did AAV1.PGT121. Ad5.PGT121 also led to more-sustained virologic control than purified PGT121 IgG. Ad5.PGT121 afforded more rapid, robust, and durable antiviral efficacy than AAV1.PGT121 and purified PGT121 IgG in HIV-1-infected humanized mice. Further evaluation of vector delivery of HIV-1 bNAbs is warranted, although approaches to prevent the generation of antiantibody responses may also be required.IMPORTANCE Broadly neutralizing antibodies (bNAbs) are being explored for HIV-1 prevention and cure strategies, but delivery of purified antibodies may prove challenging. We investigated adenovirus serotype 5 (Ad5) and adeno-associated virus serotype 1 (AAV1) vectors to deliver the HIV-1-specific bNAb PGT121. Ad5.PGT121 afforded more rapid, robust, and durable antiviral efficacy than AAV1.PGT121 and purified PGT121 IgG in HIV-1-infected humanized mice.


Subject(s)
Adenoviridae , Genetic Therapy/methods , Genetic Vectors , HIV Infections , HIV-1 , Transduction, Genetic/methods , Animals , Female , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/therapy , HIV-1/genetics , HIV-1/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout
12.
JCI Insight ; 2(7): e88533, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28405607

ABSTRACT

HIV vaginal transmission accounts for the majority of newly acquired heterosexual infections. However, the mechanism by which HIV spreads from the initial site of viral entry at the mucosal surface of the female genital tract to establish a systemic infection of lymphoid and peripheral tissues is not known. Once the virus exits the mucosa it rapidly spreads to all tissues, leading to CD4+ T cell depletion and the establishment of a viral reservoir that cannot be eliminated with current treatments. Understanding the molecular and cellular requirements for viral dissemination from the genital tract is therefore of great importance, as it could reveal new strategies to lengthen the window of opportunity to target the virus at its entry site in the mucosa where it is the most vulnerable and thus prevent systemic infection. Using HIV vaginal infection of humanized mice as a model of heterosexual transmission, we demonstrate that blocking the ability of leukocytes to respond to chemoattractants prevented HIV from leaving the female genital tract. Furthermore, blocking lymphocyte egress from lymph nodes prevented viremia and infection of the gut. Leukocyte trafficking therefore plays a major role in viral dissemination, and targeting the chemoattractant molecules involved can prevent the establishment of a systemic infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/transmission , HIV-1/pathogenicity , Receptors, CCR7/immunology , Receptors, Lysosphingolipid/immunology , Vagina/virology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Chemokine CCL19/immunology , Chemokine CCL21/immunology , Disease Models, Animal , Female , Humans , Leukocytes/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Sphingosine-1-Phosphate Receptors , Vagina/immunology
13.
J Infect Dis ; 214(4): 612-6, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27357340

ABSTRACT

Humanized mice reconstituted with a human immune system can be mucosally infected with human immunodeficiency virus (HIV), opening up the possibility of studying HIV transmission in a small-animal model. Here we report that passive immunization with the broadly neutralizing antibody b12 protected humanized mice against repetitive intravaginal infection in a dose-dependent manner. In addition, treatment with the antibody PGT126, which is more potent in vitro, was more efficacious in vivo and provided sterilizing protection. Our results demonstrate that humanized mice can be used as a small-animal model to study the efficacy and mechanism of broadly neutralizing antibody protection against HIV acquisition.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Disease Models, Animal , HIV Antibodies/administration & dosage , HIV Infections/prevention & control , Immunization, Passive/methods , Animals , Dose-Response Relationship, Immunologic , Female , Mice , Mice, SCID , Treatment Outcome
14.
Science ; 348(6241): aaa8205, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26089520

ABSTRACT

Genital Chlamydia trachomatis (Ct) infection induces protective immunity that depends on interferon-γ-producing CD4 T cells. By contrast, we report that mucosal exposure to ultraviolet light (UV)-inactivated Ct (UV-Ct) generated regulatory T cells that exacerbated subsequent Ct infection. We show that mucosal immunization with UV-Ct complexed with charge-switching synthetic adjuvant particles (cSAPs) elicited long-lived protection in conventional and humanized mice. UV-Ct-cSAP targeted immunogenic uterine CD11b(+)CD103(-) dendritic cells (DCs), whereas UV-Ct accumulated in tolerogenic CD11b(-)CD103(+) DCs. Regardless of vaccination route, UV-Ct-cSAP induced systemic memory T cells, but only mucosal vaccination induced effector T cells that rapidly seeded uterine mucosa with resident memory T cells (T(RM) cells). Optimal Ct clearance required both T(RM) seeding and subsequent infection-induced recruitment of circulating memory T cells. Thus, UV-Ct-cSAP vaccination generated two synergistic memory T cell subsets with distinct migratory properties.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Immunologic Memory , Th1 Cells/immunology , Uterus/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, CD/immunology , Bacterial Vaccines/administration & dosage , CD11 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Chlamydia trachomatis/radiation effects , Dendritic Cells/immunology , Female , Integrin alpha Chains/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucous Membrane/immunology , Nanoparticles/administration & dosage , T-Lymphocyte Subsets/immunology , Ultraviolet Rays , Vaccination/methods , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
15.
Nat Commun ; 6: 7562, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26108174

ABSTRACT

Initial events after exposure determine HIV-1 disease progression, underscoring a critical need to understand host mechanisms that interfere with initial viral replication. Although associated with chronic HIV-1 control, it is not known whether interleukin-21 (IL-21) contributes to early HIV-1 immunity. Here we take advantage of tractable primary human lymphoid organ aggregate cultures to show that IL-21 directly suppresses HIV-1 replication, and identify microRNA-29 (miR-29) as an antiviral factor induced by IL-21 in CD4 T cells. IL-21 promotes transcription of all miR-29 species through STAT3, whose binding to putative regulatory regions within the MIR29 gene is enriched by IL-21 signalling. Notably, exogenous IL-21 limits early HIV-1 infection in humanized mice, and lower viremia in vivo is associated with higher miR-29 expression. Together, these findings reveal a novel antiviral IL-21-miR-29 axis that promotes CD4 T-cell-intrinsic resistance to HIV-1 infection, and suggest a role for IL-21 in initial HIV-1 control in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , HIV Infections/metabolism , HIV-1/physiology , Interleukins/pharmacology , MicroRNAs/metabolism , Animals , Antibodies, Monoclonal, Humanized , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , Spleen/cytology , Tissue Culture Techniques
16.
Nature ; 490(7419): 283-7, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22854780

ABSTRACT

After host entry through mucosal surfaces, human immunodeficiency virus-1 (HIV-1) disseminates to lymphoid tissues to establish a generalized infection of the immune system. The mechanisms by which this virus spreads among permissive target cells locally during the early stages of transmission and systemically during subsequent dissemination are not known. In vitro studies suggest that the formation of virological synapses during stable contacts between infected and uninfected T cells greatly increases the efficiency of viral transfer. It is unclear, however, whether T-cell contacts are sufficiently stable in vivo to allow for functional synapse formation under the conditions of perpetual cell motility in epithelial and lymphoid tissues. Here, using multiphoton intravital microscopy, we examine the dynamic behaviour of HIV-infected T cells in the lymph nodes of humanized mice. We find that most productively infected T cells migrate robustly, resulting in their even distribution throughout the lymph node cortex. A subset of infected cells formed multinucleated syncytia through HIV envelope-dependent cell fusion. Both uncoordinated motility of syncytia and adhesion to CD4(+) lymph node cells led to the formation of long membrane tethers, increasing cell lengths to up to ten times that of migrating uninfected T cells. Blocking the egress of migratory T cells from the lymph nodes into efferent lymph vessels, and thus interrupting T-cell recirculation, limited HIV dissemination and strongly reduced plasma viraemia. Thus, we have found that HIV-infected T cells are motile, form syncytia and establish tethering interactions that may facilitate cell-to-cell transmission through virological synapses. Migration of T cells in lymph nodes therefore spreads infection locally, whereas their recirculation through tissues is important for efficient systemic viral spread, suggesting new molecular targets to antagonize HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , HIV/immunology , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Female , Giant Cells , HIV Infections/transmission , Humans , Lymph Nodes/virology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic
17.
Sci Transl Med ; 4(143): 143ra98, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22814851

ABSTRACT

The development of mouse/human chimeras through the engraftment of human immune cells and tissues into immunodeficient mice, including the recently described humanized BLT (bone marrow, liver, thymus) mouse model, holds great promise to facilitate the in vivo study of human immune responses. However, little data exist regarding the extent to which cellular immune responses in humanized mice accurately reflect those seen in humans. We infected humanized BLT mice with HIV-1 as a model pathogen and characterized HIV-1-specific immune responses and viral evolution during the acute phase of infection. HIV-1-specific CD8(+) T cell responses in these mice were found to closely resemble those in humans in terms of their specificity, kinetics, and immunodominance. Viral sequence evolution also revealed rapid and highly reproducible escape from these responses, mirroring the adaptations to host immune pressures observed during natural HIV-1 infection. Moreover, mice expressing the protective HLA-B*57 allele exhibited enhanced control of viral replication and restricted the same CD8(+) T cell responses to conserved regions of HIV-1 Gag that are critical to its control of HIV-1 in humans. These data reveal that the humanized BLT mouse model appears to accurately recapitulate human pathogen-specific cellular immunity and the fundamental immunological mechanisms required to control a model human pathogen, aspects critical to the use of a small-animal model for human pathogens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV-1/immunology , Animals , HIV Infections/immunology , HLA-B Antigens/metabolism , Humans , Immunity, Cellular/immunology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL