Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters











Publication year range
1.
Mol Ther ; 32(10): 3485-3503, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39222637

ABSTRACT

Chimeric antigen receptor (CAR) T cells from allogeneic donors promise "off-the-shelf" availability by overcoming challenges associated with autologous cell manufacturing. However, recipient immunologic rejection of allogeneic CAR-T cells may decrease their in vivo lifespan and limit treatment efficacy. Here, we demonstrate that the immunosuppressants rapamycin and tacrolimus effectively mitigate allorejection of HLA-mismatched CAR-T cells in immunocompetent humanized mice, extending their in vivo persistence to that of syngeneic humanized mouse-derived CAR-T cells. In turn, genetic knockout (KO) of FKBP prolyl isomerase 1A (FKBP1A), which encodes a protein targeted by both drugs, was necessary to confer CD19-specific CAR-T cells (19CAR) robust functional resistance to these immunosuppressants. FKBP1AKO 19CAR-T cells maintained potent in vitro functional profiles and controlled in vivo tumor progression similarly to untreated 19CAR-T cells. Moreover, immunosuppressant treatment averted in vivo allorejection permitting FKBP1AKO 19CAR-T cell-driven B cell aplasia. Thus, we demonstrate that genome engineering enables immunosuppressant treatment to improve the therapeutic potential of universal donor-derived CAR-T cells.


Subject(s)
Immunosuppressive Agents , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Tacrolimus Binding Protein 1A , Animals , Mice , Humans , Immunosuppressive Agents/pharmacology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/genetics , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , Tacrolimus/pharmacology , Graft Rejection/immunology , Graft Rejection/prevention & control , Sirolimus/pharmacology , Transplantation, Homologous , Antigens, CD19/immunology , Antigens, CD19/metabolism
3.
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.

4.
bioRxiv ; 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38496468

ABSTRACT

The recurring emergence of novel respiratory viruses has highlighted our poor understanding of the human immune mechanisms governing the resolution of lung infection in an immunologically naïve context. Using SARS-CoV-2 as a prototypical emerging respiratory virus, we leveraged mice co-engrafted with a genetically matched fetal lung xenograft (fLX) and a human immune system (BLT-L mice) to investigate such mechanisms. While BLT-L mice effectively resolve SARS-CoV-2 infection following acute viral replication in fLX, viral clearance is robustly abrogated through systemic depletion of CD4+, but not CD3+ or CD8+ cells, resulting in persistent infection. Leveraging single-cell transcriptomics to uncover the CD4-expressing subsets driving infection resolution, we identified a novel subset of lung extravascular inflammatory monocytes (ExiMO) with antiviral functions. ExiMO are the dominant CD163-expressing myeloid population emerging in fLX upon acute infection and derive from recruited circulating CD4+ monocytes. They are highly enriched in viral RNA and elicit a robust antiviral response before vanishing from tissues when infection resolves. Notably, systemic CD4+ cell depletion results in impaired recruitment of CD163+ cells into fLX and leads to a state of immune tolerance and chronic infection defined by the absence of ExiMO antiviral responses. Together, our study uncovers ExiMO as major sentinels driving SARS-CoV-2 infection resolution in human lung tissues without pre-existing immunity. This work expands our understanding of lung extravascular monocytes and unravels novel facets of the cellular determinants governing our vulnerability to viral respiratory pathogens.

5.
Blood Adv ; 7(22): 6898-6912, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37729429

ABSTRACT

Hemogenic endothelial cells (HECs) are specialized cells that undergo endothelial-to-hematopoietic transition (EHT) to give rise to the earliest precursors of hematopoietic progenitors that will eventually sustain hematopoiesis throughout the lifetime of an organism. Although HECs are thought to be primarily limited to the aorta-gonad-mesonephros (AGM) during early development, EHT has been described in various other hematopoietic organs and embryonic vessels. Though not defined as a hematopoietic organ, the lung houses many resident hematopoietic cells, aids in platelet biogenesis, and is a reservoir for hematopoietic stem and progenitor cells (HSPCs). However, lung HECs have never been described. Here, we demonstrate that the fetal lung is a potential source of HECs that have the functional capacity to undergo EHT to produce de novo HSPCs and their resultant progeny. Explant cultures of murine and human fetal lungs display adherent endothelial cells transitioning into floating hematopoietic cells, accompanied by the gradual loss of an endothelial signature. Flow cytometric and functional assessment of fetal-lung explants showed the production of multipotent HSPCs that expressed the EHT and pre-HSPC markers EPCR, CD41, CD43, and CD44. scRNA-seq and small molecule modulation demonstrated that fetal lung HECs rely on canonical signaling pathways to undergo EHT, including TGFß/BMP, Notch, and YAP. Collectively, these data support the possibility that post-AGM development, functional HECs are present in the fetal lung, establishing this location as a potential extramedullary site of de novo hematopoiesis.


Subject(s)
Hemangioblasts , Hematopoiesis , Animals , Mice , Humans , Hematopoietic Stem Cells/metabolism , Cell Differentiation , Endothelium , Hemangioblasts/metabolism
6.
Front Immunol ; 14: 1324618, 2023.
Article in English | MEDLINE | ID: mdl-38274817

ABSTRACT

Glioblastoma (GBM) is the most aggressive and common type of malignant brain tumor diagnosed in adults. Preclinical immunocompetent mouse tumor models generated using mouse tumor cells play a pivotal role in testing the therapeutic efficacy of emerging immune-based therapies for GBMs. However, the clinical translatability of such studies is limited as mouse tumor lines do not fully recapitulate GBMs seen in inpatient settings. In this study, we generated three distinct, imageable human-GBM (hGBM) models in humanized mice using patient-derived GBM cells that cover phenotypic and genetic GBM heterogeneity in primary (invasive and nodular) and recurrent tumors. We developed a pipeline to first enrich the tumor-initiating stem-like cells and then successfully established robust patient-derived GBM tumor engraftment and growth in bone marrow-liver-thymus (BLT) humanized mice. Multiplex immunofluorescence of GBM tumor sections revealed distinct phenotypic features of the patient GBM tumors, with myeloid cells dominating the immune landscape. Utilizing flow cytometry and correlative immunofluorescence, we profiled the immune microenvironment within the established human GBM tumors in the BLT mouse models and showed tumor infiltration of variable human immune cells, creating a unique immune landscape compared with lymphoid organs. These findings contribute substantially to our understanding of GBM biology within the context of the human immune system in humanized mice and lay the groundwork for further translational studies aimed at advancing therapeutic strategies for GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Humans , Mice , Animals , Glioblastoma/therapy , Neoplasm Recurrence, Local/pathology , Disease Models, Animal , Neoplastic Stem Cells/pathology , Tumor Microenvironment
7.
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
8.
Nat Commun ; 13(1): 1103, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35232959

ABSTRACT

The human hematopoietic stem cell harbors remarkable regenerative potential that can be harnessed therapeutically. During early development, hematopoietic stem cells in the fetal liver undergo active expansion while simultaneously retaining robust engraftment capacity, yet the underlying molecular program responsible for their efficient engraftment remains unclear. Here, we profile 26,407 fetal liver cells at both the transcriptional and protein level including ~7,000 highly enriched and functional fetal liver hematopoietic stem cells to establish a detailed molecular signature of engraftment potential. Integration of transcript and linked cell surface marker expression reveals a generalizable signature defining functional fetal liver hematopoietic stem cells and allows for the stratification of enrichment strategies with high translational potential. More precisely, our integrated analysis identifies CD201 (endothelial protein C receptor (EPCR), encoded by PROCR) as a marker that can specifically enrich for engraftment potential. This comprehensive, multi-modal profiling of engraftment capacity connects a critical biological function at a key developmental timepoint with its underlying molecular drivers. As such, it serves as a useful resource for the field and forms the basis for further biological exploration of strategies to retain the engraftment potential of hematopoietic stem cells ex vivo or induce this potential during in vitro hematopoietic stem cell generation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Liver
9.
Eur J Immunol ; 52(3): 447-461, 2022 03.
Article in English | MEDLINE | ID: mdl-34935145

ABSTRACT

Effective function of CD8+ T cells and enhanced innate activation of DCs in response to HIV-1 is linked to protective antiviral immunity in controllers. Manipulation of DC targeting the master regulator TANK-binding Kinase 1 (TBK1) might be useful to acquire controller-like properties. Here, we evaluated the impact of the combination of 2´3´-c´diAM(PS)2 and Poly I:C as potential adjuvants capable of potentiating DC´s abilities to induce polyfunctional HIV-1 specific CD8+ T-cell responses in vitro and in vivo using a humanized BLT mouse model. Adjuvant combination enhanced TBK-1 phosphorylation and IL-12 and IFN-ß expression on DC and increased their ability to activate polyfunctional HIV-1-specific CD8+ T cells in vitro. Moreover, higher proportions of hBLT mice vaccinated with ADJ-DC exhibited less severe CD4+ T-cell depletion following HIV-1 infection compared to control groups. This was associated with infiltration of CD8+ T cells in the white pulp from the spleen, reduced spread of infected p24+ cells to LN, and with preserved abilities of CD8+ T cells from the spleen and blood of vaccinated animals to induce specific polyfunctional responses upon antigen stimulation. Therefore, priming of DC with PolyI:C and STING agonists might be useful for future HIV-1 vaccine studies.


Subject(s)
AIDS Vaccines , HIV-1 , AIDS Vaccines/metabolism , Adjuvants, Immunologic/pharmacology , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Dendritic Cells , HIV Core Protein p24/metabolism , Lymphoid Tissue , Mice , Poly I-C/pharmacology
10.
Nat Commun ; 12(1): 6230, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711828

ABSTRACT

T cells undergo rigorous selection in the thymus to ensure self-tolerance and prevent autoimmunity, with this process requiring innocuous self-antigens (Ags) to be presented to thymocytes. Self-Ags are either expressed by thymic stroma cells or transported to the thymus from the periphery by migratory dendritic cells (DCs); meanwhile, small blood-borne peptides can access the thymic parenchyma by diffusing across the vascular lining. Here we describe an additional pathway of thymic Ag acquisition that enables circulating antigenic macromolecules to access both murine and human thymi. This pathway depends on a subset of thymus-resident DCs, distinct from both parenchymal and circulating migratory DCs, that are positioned in immediate proximity to thymic microvessels where they extend cellular processes across the endothelial barrier into the blood stream. Transendothelial positioning of DCs depends on DC-expressed CX3CR1 and its endothelial ligand, CX3CL1, and disrupting this chemokine pathway prevents thymic acquisition of circulating proteins and compromises negative selection of Ag-reactive thymocytes. Thus, transendothelial DCs represent a mechanism by which the thymus can actively acquire blood-borne Ags to induce and maintain central tolerance.


Subject(s)
Blood/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Autoantigens/immunology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Cell Differentiation , Cell Movement , Chemokine CX3CL1/genetics , Chemokine CX3CL1/immunology , Dendritic Cells/cytology , Endothelial Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Self Tolerance , Thymocytes/cytology , Thymus Gland/cytology
12.
Cell Rep ; 36(9): 109622, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469717

ABSTRACT

HIV-1 entry into host cells leads to one of the following three alternative fates: (1) HIV-1 elimination by restriction factors, (2) establishment of HIV-1 latency, or (3) active viral replication in target cells. Here, we report the development of an improved system for monitoring HIV-1 fate at single-cell and population levels and show the diverse applications of this system to study specific aspects of HIV-1 fate in different cell types and under different environments. An analysis of the transcriptome of infected, primary CD4+ T cells that support alternative fates of HIV-1 identifies differential gene expression signatures in these cells. Small molecules are able to selectively target cells that support viral replication with no significant effect on viral latency. In addition, HIV-1 fate varies in different tissues following infection of humanized mice in vivo. Altogether, these studies indicate that intra- and extra-cellular environments contribute to the fate of HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cellular Microenvironment , HIV Infections/virology , HIV-1/pathogenicity , Animals , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , HEK293 Cells , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/immunology , HIV-1/drug effects , HIV-1/growth & development , HIV-1/immunology , Host-Pathogen Interactions , Humans , Mice, Inbred NOD , Mice, SCID , THP-1 Cells , Transcriptome , Virus Internalization , Virus Latency , Virus Replication
13.
Front Immunol ; 12: 667393, 2021.
Article in English | MEDLINE | ID: mdl-34122425

ABSTRACT

Humanized bone marrow-liver-thymus (HuBLT) mice are a revolutionary small-animal model that has facilitated the study of human immune function and human-restricted pathogens, including human immunodeficiency virus type 1 (HIV-1). These mice recapitulate many aspects of acute and chronic HIV-1 infection, but exhibit weak and variable T-cell responses when challenged with HIV-1, hindering our ability to confidently detect HIV-1-specific responses or vaccine effects. To identify the cause of this, we comprehensively analyzed T-cell development, diversity, and function in HuBLT mice. We found that virtually all HuBLT were well-reconstituted with T cells and had intact TCRß sequence diversity, thymic development, and differentiation to memory and effector cells. However, there was poor CD4+ and CD8+ T-cell responsiveness to physiologic stimuli and decreased TH1 polarization that correlated with deficient reconstitution of innate immune cells, in particular monocytes. HIV-1 infection of HuBLT mice showed that mice with higher monocyte reconstitution exhibited greater CD8+ T cells responses and HIV-1 viral evolution within predicted HLA-restricted epitopes. Thus, T-cell responses to immune challenges are blunted in HuBLT mice due to a deficiency of innate immune cells, and future efforts to improve the model for HIV-1 immune response and vaccine studies need to be aimed at restoring innate immune reconstitution.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Immune Reconstitution , Animals , Biological Evolution , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , HIV Infections/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Viremia
14.
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
15.
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
16.
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
17.
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
18.
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
19.
J Virol ; 93(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30728264

ABSTRACT

Lactobacillus bacteria are potential delivery vehicles for biopharmaceutical molecules because they are well-recognized as safe microorganisms that naturally inhabit the human body. The goal of this study was to employ these lactobacilli to combat human immunodeficiency virus type 1 (HIV-1) infection and transmission. By using a chromosomal integration method, we engineered Lactobacillus acidophilus ATCC 4356 to display human CD4, the HIV-1 receptor, on the cell surface. Since human CD4 can bind to any infectious HIV-1 particles, the engineered lactobacilli can potentially capture HIV-1 of different subtypes and prevent infection. Our data demonstrate that the CD4-carrying bacteria are able to adsorb HIV-1 particles and reduce infection significantly in vitro and also block intrarectal HIV-1 infection in a humanized mouse model in preliminary tests in vivo Our results support the potential of this approach to decrease the efficiency of HIV-1 sexual transmission.IMPORTANCE In the absence of an effective vaccine, alternative approaches to block HIV-1 infection and transmission with commensal bacteria expressing antiviral proteins are being considered. This report provides a proof-of-concept by using Lactobacillus bacteria stably expressing the HIV-1 receptor CD4 to capture and neutralize HIV-1 in vitro and in a humanized mouse model. The stable expression of antiviral proteins, such as CD4, following genomic integration of the corresponding genes into this Lactobacillus strain may contribute to the prevention of HIV-1 sexual transmission.


Subject(s)
CD4 Antigens/metabolism , HIV Infections/prevention & control , HIV-1/metabolism , Lactobacillus acidophilus/metabolism , Animals , CD4 Antigens/genetics , Cell Line , Female , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/genetics , Humans , Lactobacillus acidophilus/genetics , Male , Mice , Mice, Knockout , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL