Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Nature ; 614(7947): 334-342, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36697826

RESUMO

The liver is bathed in bacterial products, including lipopolysaccharide transported from the intestinal portal vasculature, but maintains a state of tolerance that is exploited by persistent pathogens and tumours1-4. The cellular basis mediating this tolerance, yet allowing a switch to immunity or immunopathology, needs to be better understood for successful immunotherapy of liver diseases. Here we show that a variable proportion of CD8+ T cells compartmentalized in the human liver co-stain for CD14 and other prototypic myeloid membrane proteins and are enriched in close proximity to CD14high myeloid cells in hepatic zone 2. CD14+CD8+ T cells preferentially accumulate within the donor pool in liver allografts, among hepatic virus-specific and tumour-infiltrating responses, and in cirrhotic ascites. CD14+CD8+ T cells exhibit increased turnover, activation and constitutive immunomodulatory features with high homeostatic IL-10 and IL-2 production ex vivo, and enhanced antiviral/anti-tumour effector function after TCR engagement. This CD14+CD8+ T cell profile can be recapitulated by the acquisition of membrane proteins-including the lipopolysaccharide receptor complex-from mononuclear phagocytes, resulting in augmented tumour killing by TCR-redirected T cells in vitro. CD14+CD8+ T cells express integrins and chemokine receptors that favour interactions with the local stroma, which can promote their induction through CXCL12. Lipopolysaccharide can also increase the frequency of CD14+CD8+ T cells in vitro and in vivo, and skew their function towards the production of chemotactic and regenerative cytokines. Thus, bacterial products in the gut-liver axis and tissue stromal factors can tune liver immunity by driving myeloid instruction of CD8+ T cells with immunomodulatory ability.


Assuntos
Linfócitos T CD8-Positivos , Tolerância Imunológica , Receptores de Lipopolissacarídeos , Lipopolissacarídeos , Fígado , Células Mieloides , Humanos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/imunologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Interleucina-2/biossíntese , Interleucina-2/imunologia , Quimiotaxia de Leucócito , Bactérias/imunologia , Intestinos/imunologia , Intestinos/microbiologia
2.
J Virol ; 93(11)2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30867315

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection is associated with aberrant immune activation; however, most model systems for HIV-1 have been used during established infection. Here, we utilize ultrasensitive HIV-1 quantification to delineate early events during the eclipse, burst, and chronic phases of HIV-1 infection in humanized mice. We show that very early in infection, HIV-1 suppresses peripheral type I interferon (IFN) and interferon-stimulated gene (ISG) responses, including the HIV-1 restriction factor IFI44. At the peak of innate immune activation, prior to CD4 T cell loss, HIV-1 infection differentially affects peripheral and lymphoid Toll-like receptor (TLR) expression profiles in T cells and macrophages. This results in a trend toward an altered activation of nuclear factor κB (NF-κB), TANK-binding kinase 1 (TBK1), and interferon regulatory factor 3 (IRF3). The subsequent type I and III IFN responses result in preferential induction of peripheral ISG responses. Following this initial innate immune activation, peripheral expression of the HIV-1 restriction factor SAM domain- and HD domain-containing protein 1 (SAMHD1) returns to levels below those observed in uninfected mice, suggesting that HIV-1 interferes with their basal expression. However, peripheral cells still retain their responsiveness to exogenous type I IFN, whereas splenic cells show a reduction in select ISGs in response to IFN. This demonstrates the highly dynamic nature of very early HIV-1 infection and suggests that blocks to the induction of HIV-1 restriction factors contribute to the establishment of viral persistence.IMPORTANCE Human immunodeficiency virus type 1 (HIV-1) infection is restricted to humans and some nonhuman primates (e.g., chimpanzee and gorilla). Alternative model systems based on simian immunodeficiency virus (SIV) infection of macaques are available but do not recapitulate all aspects of HIV-1 infection and disease. Humanized mice, which contain a human immune system, can be used to study HIV-1, but only limited information on early events and immune responses is available to date. Here, we describe very early immune responses to HIV-1 and demonstrate a suppression of cell-intrinsic innate immunity. Furthermore, we show that HIV-1 infection interacts differently with innate immune responses in blood and lymphoid organs.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Imunidade Inata/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Células HEK293 , HIV-1/imunologia , HIV-1/metabolismo , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Cinética , Macrófagos/virologia , Camundongos , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 com Domínio SAM e Domínio HD/metabolismo
3.
Nature ; 501(7466): 237-41, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23903655

RESUMO

More than 130 million people worldwide chronically infected with hepatitis C virus (HCV) are at risk of developing severe liver disease. Antiviral treatments are only partially effective against HCV infection, and a vaccine is not available. Development of more efficient therapies has been hampered by the lack of a small animal model. Building on the observation that CD81 and occludin (OCLN) comprise the minimal set of human factors required to render mouse cells permissive to HCV entry, we previously showed that transient expression of these two human genes is sufficient to allow viral uptake into fully immunocompetent inbred mice. Here we demonstrate that transgenic mice stably expressing human CD81 and OCLN also support HCV entry, but innate and adaptive immune responses restrict HCV infection in vivo. Blunting antiviral immunity in genetically humanized mice infected with HCV results in measurable viraemia over several weeks. In mice lacking the essential cellular co-factor cyclophilin A (CypA), HCV RNA replication is markedly diminished, providing genetic evidence that this process is faithfully recapitulated. Using a cell-based fluorescent reporter activated by the NS3-4A protease we visualize HCV infection in single hepatocytes in vivo. Persistently infected mice produce de novo infectious particles, which can be inhibited with directly acting antiviral drug treatment, thereby providing evidence for the completion of the entire HCV life cycle in inbred mice. This genetically humanized mouse model opens new opportunities to dissect genetically HCV infection in vivo and provides an important preclinical platform for testing and prioritizing drug candidates and may also have utility for evaluating vaccine efficacy.


Assuntos
Modelos Animais de Doenças , Engenharia Genética , Hepacivirus/fisiologia , Hepatite C/genética , Hepatite C/virologia , Replicação Viral , Animais , Linhagem Celular , Ciclofilina A/genética , Ciclofilina A/metabolismo , Hepacivirus/imunologia , Hepatite C/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ocludina/genética , Ocludina/metabolismo , Fator de Transcrição STAT1/deficiência , Tetraspanina 28/genética , Tetraspanina 28/metabolismo , Viremia/virologia , Vírion/crescimento & desenvolvimento , Vírion/fisiologia
4.
Proc Natl Acad Sci U S A ; 113(9): 2484-9, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26884193

RESUMO

Hepatitis C virus (HCV) is a unique enveloped virus that assembles as a hybrid lipoviral particle by tightly interacting with host lipoproteins. As a result, HCV virions display a characteristic low buoyant density and a deceiving coat, with host-derived apolipoproteins masking viral epitopes. We previously described methods to produce high-titer preparations of HCV particles with tagged envelope glycoproteins that enabled ultrastructural analysis of affinity-purified virions. Here, we performed proteomics studies of HCV isolated from culture media of infected hepatoma cells to define viral and host-encoded proteins associated with mature virions. Using two different affinity purification protocols, we detected four viral and 46 human cellular proteins specifically copurifying with extracellular HCV virions. We determined the C terminus of the mature capsid protein and reproducibly detected low levels of the viral nonstructural protein, NS3. Functional characterization of virion-associated host factors by RNAi identified cellular proteins with either proviral or antiviral roles. In particular, we discovered a novel interaction between HCV capsid protein and the nucleoporin Nup98 at cytosolic lipid droplets that is important for HCV propagation. These results provide the first comprehensive view to our knowledge of the protein composition of HCV and new insights into the complex virus-host interactions underlying HCV infection.


Assuntos
Hepacivirus/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteômica , Proteínas Virais/metabolismo , Vírion/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Hepacivirus/metabolismo , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Morfogênese , Proteínas Virais/química
5.
Immunology ; 154(1): 50-61, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29446074

RESUMO

Humanized mice are increasingly appreciated as an incredibly powerful platform for infectious disease research. The often very narrow species tropism of many viral infections, coupled with the sometimes misleading results from preclinical studies in animal models further emphasize the need for more predictive model systems based on human cells rather than surrogates. Humanized mice represent such a model and have been greatly enhanced with regards to their immune system reconstitution as well as immune functionality in the past years, resulting in their recommendation as a preclinical model by the US Food and Drug Administration. This review aims to give a detailed summary of the generation of human peripheral blood lymphocyte-, CD34+ haematopoietic stem cell- and bone marrow/liver/thymus-reconstituted mice and available improved models (e.g. myeloid- or T-cell-only mice, MISTRG, NSG-SGM3). Additionally, we summarize human-tropic viral infections, for which humanized mice offer a novel approach for the study of disease pathogenesis as well as future perspectives for their use in biomedical, drug and vaccine research.


Assuntos
Viroses/virologia , Vírus/patogenicidade , Imunidade Adaptativa , Animais , Transplante de Medula Óssea , Modelos Animais de Doenças , Predisposição Genética para Doença , Transplante de Células-Tronco Hematopoéticas , Interações Hospedeiro-Patógeno , Humanos , Imunidade Humoral , Imunidade Inata , Camundongos Transgênicos , Fenótipo , Transplante Heterólogo , Viroses/genética , Viroses/imunologia , Vírus/imunologia
6.
Nature ; 492(7427): 118-22, 2012 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-23103874

RESUMO

Human antibodies to human immunodeficiency virus-1 (HIV-1) can neutralize a broad range of viral isolates in vitro and protect non-human primates against infection. Previous work showed that antibodies exert selective pressure on the virus but escape variants emerge within a short period of time. However, these experiments were performed before the recent discovery of more potent anti-HIV-1 antibodies and their improvement by structure-based design. Here we re-examine passive antibody transfer as a therapeutic modality in HIV-1-infected humanized mice. Although HIV-1 can escape from antibody monotherapy, combinations of broadly neutralizing antibodies can effectively control HIV-1 infection and suppress viral load to levels below detection. Moreover, in contrast to antiretroviral therapy, the longer half-life of antibodies led to control of viraemia for an average of 60 days after cessation of therapy. Thus, combinations of potent monoclonal antibodies can effectively control HIV-1 replication in humanized mice, and should be re-examined as a therapeutic modality in HIV-1-infected individuals.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos/imunologia , Modelos Animais de Doenças , Infecções por HIV/virologia , HIV-1/genética , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , HIV-1/isolamento & purificação , Meia-Vida , Humanos , Imunização Passiva , Camundongos , Camundongos Endogâmicos NOD , Fatores de Tempo , Carga Viral/efeitos dos fármacos
7.
Artigo em Inglês | MEDLINE | ID: mdl-28193659

RESUMO

A hepatitis C virus (HCV) epidemic affecting HIV-infected men who have sex with men (MSM) is expanding worldwide. In spite of the improved cure rates obtained with the new direct-acting antiviral drug (DAA) combinations, the high rate of reinfection within this population calls urgently for novel preventive interventions. In this study, we determined in cell culture and ex vivo experiments with human colorectal tissue that lipoquads, G-quadruplex DNA structures fused to cholesterol, are efficient HCV pangenotypic entry and cell-to-cell transmission inhibitors. Thus, lipoquads may be promising candidates for the development of rectally applied gels to prevent HCV transmission.


Assuntos
Antivirais/uso terapêutico , Colesterol/uso terapêutico , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Hepatite C/transmissão , Oligonucleotídeos/uso terapêutico , Internalização do Vírus/efeitos dos fármacos , Linhagem Celular Tumoral , Colesterol/química , Quadruplex G , Células HEK293 , Infecções por HIV , Hepacivirus/crescimento & desenvolvimento , Homossexualidade Masculina , Humanos , Masculino , Oligonucleotídeos/química
8.
Nature ; 474(7350): 208-11, 2011 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-21654804

RESUMO

Hepatitis C virus (HCV) remains a major medical problem. Antiviral treatment is only partially effective and a vaccine does not exist. Development of more effective therapies has been hampered by the lack of a suitable small animal model. Although xenotransplantation of immunodeficient mice with human hepatocytes has shown promise, these models are subject to important challenges. Building on the previous observation that CD81 and occludin comprise the minimal human factors required to render mouse cells permissive to HCV entry in vitro, we attempted murine humanization via a genetic approach. Here we show that expression of two human genes is sufficient to allow HCV infection of fully immunocompetent inbred mice. We establish a precedent for applying mouse genetics to dissect viral entry and validate the role of scavenger receptor type B class I for HCV uptake. We demonstrate that HCV can be blocked by passive immunization, as well as showing that a recombinant vaccinia virus vector induces humoral immunity and confers partial protection against heterologous challenge. This system recapitulates a portion of the HCV life cycle in an immunocompetent rodent for the first time, opening opportunities for studying viral pathogenesis and immunity and comprising an effective platform for testing HCV entry inhibitors in vivo.


Assuntos
Modelos Animais de Doenças , Hepacivirus/fisiologia , Hepatite C/genética , Hepatite C/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Anticorpos Bloqueadores/imunologia , Antígenos CD/genética , Antígenos CD/metabolismo , Células Cultivadas , Claudina-1 , Genótipo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatócitos/citologia , Humanos , Imunização Passiva , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Receptores Virais/genética , Receptores Virais/metabolismo , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Tetraspanina 28 , Transfecção , Tropismo Viral
9.
J Hepatol ; 65(2): 334-43, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27151182

RESUMO

BACKGROUND & AIMS: Human liver chimeric mice are useful models of human hepatitis virus infection, including hepatitis B and C virus infections. Independently, immunodeficient mice reconstituted with CD34(+) hematopoietic stem cells (HSC) derived from fetal liver reliably develop human T and B lymphocytes. Combining these systems has long been hampered by inefficient liver reconstitution of human fetal hepatoblasts. Our study aimed to enhance hepatoblast engraftment in order to create a mouse model with syngeneic human liver and immune cells. METHODS: The effects of human oncostatin-M administration on fetal hepatoblast engraftment into immunodeficient fah(-/-) mice was tested. Mice were then transplanted with syngeneic human hepatoblasts and HSC after which human leukocyte chimerism and functionality were analyzed by flow cytometry, and mice were challenged with HBV. RESULTS: Addition of human oncostatin-M enhanced human hepatoblast engraftment in immunodeficient fah(-/-) mice by 5-100 fold. In contrast to mice singly engrafted with HSC, which predominantly developed human T and B lymphocytes, mice co-transplanted with syngeneic hepatoblasts also contained physiological levels of human monocytes and natural killer cells. Upon infection with HBV, these mice displayed rapid and sustained viremia. CONCLUSIONS: Our study provides a new mouse model with improved human fetal hepatoblast engraftment and an expanded human immune cell repertoire. With further improvements, this model may become useful for studying human immunity against viral hepatitis. LAY SUMMARY: Important human pathogens such as hepatitis B virus, hepatitis C virus and human immunodeficiency virus only infect human cells which complicates the development of mouse models for the study of these pathogens. One way to make mice permissive for human pathogens is the transplantation of human cells into immune-compromised mice. For instance, the transplantation of human liver cells will allow the infection of these so-called "liver chimeric mice" with hepatitis B virus and hepatitis C virus. The co-transplantation of human immune cells into liver chimeric mice will further allow the study of human immune responses to hepatitis B virus or hepatitis C virus. However, for immunological studies it will be crucial that the transplanted human liver and immune cells are derived from the same human donor. In our study we describe the efficient engraftment of human fetal liver cells and immune cells derived from the same donor into mice. We show that liver co-engraftment resulted in an expanded human immune cell repertoire, including monocytes and natural killer cells in the liver. We further demonstrate that these mice could be infected with hepatitis B virus, which lead to an expansion of natural killer cells. In conclusion we have developed a new mouse model that could be useful to study human immune responses to human liver pathogens.


Assuntos
Células Matadoras Naturais , Monócitos , Animais , Hepatite B , Hepatócitos , Humanos , Camundongos , Camundongos SCID
10.
Proc Natl Acad Sci U S A ; 110(41): 16538-43, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24043801

RESUMO

Effective control of HIV-1 infection in humans is achieved using combinations of antiretroviral therapy (ART) drugs. In humanized mice (hu-mice), control of viremia can be achieved using either ART or by immunotherapy using combinations of broadly neutralizing antibodies (bNAbs). Here we show that treatment of HIV-1-infected hu-mice with a combination of three highly potent bNAbs not only resulted in complete viremic control but also led to a reduction in cell-associated HIV-1 DNA. Moreover, lowering the initial viral load by coadministration of ART and immunotherapy enabled prolonged viremic control by a single bNAb after ART was withdrawn. Similarly, a single injection of adeno-associated virus directing expression of one bNAb produced durable viremic control after ART was terminated. We conclude that immunotherapy reduces plasma viral load and cell-associated HIV-1 DNA and that decreasing the initial viral load enables single bNAbs to control viremia in hu-mice.


Assuntos
Antirretrovirais/imunologia , Anticorpos Neutralizantes/imunologia , Infecções por HIV/prevenção & controle , HIV-1/efeitos dos fármacos , HIV-1/imunologia , Imunoterapia/métodos , Animais , Antirretrovirais/farmacologia , Anticorpos Neutralizantes/farmacologia , Primers do DNA/genética , DNA Viral/metabolismo , Dependovirus , Quimioterapia Combinada , Humanos , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA , Carga Viral/efeitos dos fármacos
11.
J Virol ; 88(4): 2205-18, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24335303

RESUMO

Dengue virus (DENV) is the cause of a potentially life-threatening disease that affects millions of people worldwide. The lack of a small animal model that mimics the symptoms of DENV infection in humans has slowed the understanding of viral pathogenesis and the development of therapies and vaccines. Here, we investigated the use of humanized "bone marrow liver thymus" (BLT) mice as a model for immunological studies and assayed their applicability for preclinical testing of antiviral compounds. Human immune system (HIS) BLT-NOD/SCID mice were inoculated intravenously with a low-passage, clinical isolate of DENV-2, and this resulted in sustained viremia and infection of leukocytes in lymphoid and nonlymphoid organs. In addition, DENV infection increased serum cytokine levels and elicited DENV-2-neutralizing human IgM antibodies. Following restimulation with DENV-infected dendritic cells, in vivo-primed T cells became activated and acquired effector function. An adenosine nucleoside inhibitor of DENV decreased the circulating viral RNA when administered simultaneously or 2 days postinfection, simulating a potential treatment protocol for DENV infection in humans. In summary, we demonstrate that BLT mice are susceptible to infection with clinical DENV isolates, mount virus-specific adaptive immune responses, and respond to antiviral drug treatment. Although additional refinements to the model are required, BLT mice are a suitable platform to study aspects of DENV infection and pathogenesis and for preclinical testing of drug and vaccine candidates. IMPORTANCE Infection with dengue virus remains a major medical problem. Progress in our understanding of the disease and development of therapeutics has been hampered by the scarcity of small animal models. Here, we show that humanized mice, i.e., animals engrafted with components of a human immune system, that were infected with a patient-derived dengue virus strain developed clinical symptoms of the disease and mounted virus-specific immune responses. We further show that this mouse model can be used to test preclinically the efficacy of antiviral drugs.


Assuntos
Imunidade Adaptativa/imunologia , Antivirais/farmacologia , Vírus da Dengue/imunologia , Dengue/tratamento farmacológico , Dengue/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Animais , Primers do DNA/genética , Dengue/virologia , Vírus da Dengue/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T/imunologia
12.
Proc Natl Acad Sci U S A ; 109(36): 14610-5, 2012 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-22908290

RESUMO

Dengue virus (DENV) is a global disease threat for which there are no approved antivirals or vaccines. Establishing state-of-the-art screening systems that rely on fluorescent or luminescent reporters may accelerate the development of anti-DENV therapeutics. However, relatively few reporter DENV platforms exist. Here, we show that DENV can be genetically engineered to express a green fluorescent protein or firefly luciferase. Reporter viruses are infectious in vitro and in vivo and are sensitive to antiviral compounds, neutralizing antibodies, and interferons. Bioluminescence imaging was used to follow the dynamics of DENV infection in mice and revealed that the virus localized predominantly to lymphoid and gut-associated tissues. The high-throughput potential of reporter DENV was demonstrated by screening a library of more than 350 IFN-stimulated genes for antiviral activity. Several antiviral effectors were identified, and they targeted DENV at two distinct life cycle steps. These viruses provide a powerful platform for applications ranging from validation of vaccine candidates to antiviral discovery.


Assuntos
Vírus da Dengue/genética , Dengue/fisiopatologia , Genes Reporter , Engenharia Genética/métodos , Proteínas de Fluorescência Verde/genética , Ensaios de Triagem em Larga Escala/métodos , Luciferases de Vaga-Lume/genética , Análise de Variância , Animais , Chlorocebus aethiops , Vírus da Dengue/metabolismo , Citometria de Fluxo , Biblioteca Gênica , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Luciferases de Vaga-Lume/metabolismo , Camundongos , Plasmídeos/genética , Células Vero
13.
Proc Natl Acad Sci U S A ; 109(16): 6205-10, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22492964

RESUMO

Hepatitis C virus (HCV) infects ∼2% of the world's population. It is estimated that there are more than 500,000 new infections annually in Egypt, the country with the highest HCV prevalence. An effective vaccine would help control this expanding global health burden. HCV is highly variable, and an effective vaccine should target conserved T- and B-cell epitopes of the virus. Conserved B-cell epitopes overlapping the CD81 receptor-binding site (CD81bs) on the E2 viral envelope glycoprotein have been reported previously and provide promising vaccine targets. In this study, we isolated 73 human mAbs recognizing five distinct antigenic regions on the virus envelope glycoprotein complex E1E2 from an HCV-immune phage-display antibody library by using an exhaustive-panning strategy. Many of these mAbs were broadly neutralizing. In particular, the mAb AR4A, recognizing a discontinuous epitope outside the CD81bs on the E1E2 complex, has an exceptionally broad neutralizing activity toward diverse HCV genotypes and protects against heterologous HCV challenge in a small animal model. The mAb panel will be useful for the design and development of vaccine candidates to elicit broadly neutralizing antibodies to HCV.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Hepacivirus/imunologia , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos/imunologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito B/imunologia , Genótipo , Células HEK293 , Hepacivirus/genética , Hepatite C/imunologia , Hepatite C/prevenção & controle , Hepatite C/virologia , Anticorpos Anti-Hepatite C/imunologia , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Tetraspanina 28/imunologia , Proteínas do Envelope Viral/genética , Vacinas contra Hepatite Viral/administração & dosagem , Vacinas contra Hepatite Viral/imunologia
14.
Immunology ; 143(2): 202-18, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24766459

RESUMO

Differentiation of CD4(+) T cells into type 1 or type 2 subsets is mediated by the expression of the opposing lineage defining transcription factors T-bet and GATA-3. However, the existence of GATA-3(+) T-bet(+) CD4(+) T cells in mice suggests functional plasticity of these subsets. Little is known about type 1 and type 2 plasticity of human T-cell subsets in vivo. Here, we show that in the xenogeneic environment of humanized mice, which lacks a functional immune-regulatory network, human CD4(+) and, notably, CD8(+) T cells preferentially differentiate into interleukin (IL)-4(+) GATA-3(+) and IL-4(+) interferon-γ(+) GATA-3(+) T-bet(+) subsets. Treatment with recombinant human IL-12 or expansion of IL-12-producing human dendritic cells in vivo reverted this phenotype and led to the down-regulation of GATA-3 expression. These changes also correlated with improved antiviral immune responses in humanized mice. In conclusion, our study shows the capacity of human CD4(+) and CD8(+) T cells for stable co-expression of GATA-3 and T-bet in humanized mice and reveals a critical role for IL-12 in regulating this phenotype.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Fator de Transcrição GATA3/metabolismo , Transplante de Células-Tronco Hematopoéticas , Interleucina-12/metabolismo , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Subpopulações de Linfócitos T/metabolismo , Adenoviridae/patogenicidade , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Linhagem da Célula , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Injeções Intraperitoneais , Interferon gama/metabolismo , Interleucina-12/administração & dosagem , Interleucina-4/metabolismo , Cinética , Fígado/imunologia , Fígado/metabolismo , Fígado/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Transplante Heterólogo , Carga Viral
15.
J Virol ; 87(15): 8282-93, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23698298

RESUMO

Hepatitis C virus (HCV) is believed to initially infect the liver through the basolateral side of hepatocytes, where it engages attachment factors and the coreceptors CD81 and scavenger receptor class B type I (SR-BI). Active transport toward the apical side brings the virus in close proximity of additional entry factors, the tight junction molecules claudin-1 and occludin. HCV is also thought to propagate via cell-to-cell spread, which allows highly efficient virion delivery to neighboring cells. In this study, we compared an adapted HCV genome, clone 2, characterized by superior cell-to cell spread, to its parental genome, J6/JFH-1, with the goal of elucidating the molecular mechanisms of HCV cell-to-cell transmission. We show that CD81 levels on the donor cells influence the efficiency of cell-to-cell spread and CD81 transfer between neighboring cells correlates with the capacity of target cells to become infected. Spread of J6/JFH-1 was blocked by anti-SR-BI antibody or in cells knocked down for SR-BI, suggesting a direct role for this receptor in HCV cell-to-cell transmission. In contrast, clone 2 displayed a significantly reduced dependence on SR-BI for lateral spread. Mutations in E1 and E2 responsible for the enhanced cell-to-cell spread phenotype of clone 2 rendered cell-free virus more susceptible to antibody-mediated neutralization. Our results indicate that although HCV can lose SR-BI dependence for cell-to-cell spread, vulnerability to neutralizing antibodies may limit this evolutionary option in vivo. Combination therapies targeting both the HCV glycoproteins and SR-BI may therefore hold promise for effective control of HCV dissemination.


Assuntos
Hepacivirus/fisiologia , Interações Hospedeiro-Patógeno , Receptores Virais/metabolismo , Receptores Depuradores Classe B/metabolismo , Internalização do Vírus , Técnicas de Silenciamento de Genes , Humanos , Receptores Virais/antagonistas & inibidores , Receptores Virais/genética , Receptores Depuradores Classe B/antagonistas & inibidores , Receptores Depuradores Classe B/genética , Tetraspanina 28/metabolismo
16.
Curr Top Microbiol Immunol ; 369: 49-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23463197

RESUMO

Hepatitis C remains a global epidemic. Approximately 3 % of the world's population suffers from chronic hepatitis C, which is caused by hepatitis C virus (HCV)-a positive sense, single-stranded RNA virus of the Flaviviridae family. HCV has a high propensity for establishing a chronic infection. If untreated chronic HCV carriers can develop severe liver disease including fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Antiviral treatment is only partially effective, costly, and poorly tolerated. A prophylactic or therapeutic vaccine for HCV does not exist. Mechanistic studies of virus-host interactions, HCV immunity, and pathogenesis as well as the development of more effective therapies have been hampered by the lack of a suitable small animal model. Besides humans, chimpanzees are the only species that is naturally susceptible to HCV infection. While experimentation in these large primates has yielded valuable insights, ethical considerations, limited availability, genetic heterogeneity, and cost limit their utility. In search for more tractable small animal models, numerous experimental approaches have been taken to recapitulate parts of the viral life cycle and/or aspects of viral pathogenesis that will be discussed in this review. Exciting new models and improvements in established models hold promise to further elucidate our understanding of chronic HCV infection.


Assuntos
Hepacivirus/fisiologia , Hepatite C/virologia , Modelos Animais , Animais , Hepacivirus/genética , Humanos , Primatas , Roedores , Tupaiidae
17.
Methods ; 59(2): 249-57, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22687621

RESUMO

Approximately 2% of the world's population is chronically infected with hepatitis C virus (HCV). Chronic hepatitis C can culminate in end stage liver disease and liver cancer if the infection is untreated. Current therapy is only partially effective and a vaccine for HCV does not exist. Since the discovery of HCV as the etiologic agent causing hepatitis C several experimental tools have been developed which have improved our understanding of the viral life cycle and the interaction of HCV with human cells. However, it remains challenging to study HCV infection in its native liver environment given its narrow species tropism, limited to humans and chimpanzees. Mice can be rendered susceptible to HCV infection by transplanting human hepatocytes into immunocompromized liver injury strains. Such human liver chimeric mice are useful as a challenge model for human hepatotropic pathogens but their utility is hampered by their inability to mount functional immune responses and practical aspects including high costs, low throughput, and donor-to-donor variability. The barriers that restrict HCV species tropism are incompletely understood. We have previously shown that expression of human CD81 and human OCLN is required for HCV uptake into mouse cells. This led to the construction of a genetically humanized mouse model for HCV infection. Here, we provide a detailed protocol for the generation of these animals and highlight some of its applications for studying HCV biology and preclinical testing of drug and vaccine candidates.


Assuntos
Modelos Animais de Doenças , Técnicas de Transferência de Genes , Hepacivirus/genética , Internalização do Vírus , Adenoviridae/genética , Animais , Hepatite C/genética , Hepatite C/prevenção & controle , Humanos , Camundongos , Camundongos Transgênicos
18.
Mol Ther ; 26(2): 329-330, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29396263
19.
Int J Cancer ; 133(10): 2341-50, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23640782

RESUMO

Lymphoproliferative diseases (LPDs) associated with Epstein-Barr virus (EBV) infection cause significant morbidity and mortality in bone marrow and solid organ transplant recipients. To gain insight into LPD pathogenesis and to identify potential effective therapeutic approaches, we investigated early molecular events leading to B-cell transformation by gene expression profiling of EBV-infected B-cells from tonsils by Affymetrix microarray 72 hr postinfection when the B-cells hyperproliferation phase starts. Cell cycle and apoptosis were the most significantly affected pathways and enriched gene sets. In particular, we found significantly increased expression of cyclin-dependent kinase (CDK)1 and CCNB1 (cyclin B1) and of one of their downstream targets BIRC5 (survivin). Importantly, the strong upregulation of the antiapoptotic protein survivin was confirmed in lymphoblastoid cell lines (LCLs) and 71% of EBV-positive post-transplant EBV-LPD lesions scored positive for survivin. The validity of early transforming events for the identification of therapeutic targets for EBV-LPD was confirmed by the marked antiproliferative effect of the CDK inhibitor flavopiridol on LCLs and by the strong induction of apoptosis by survivin inhibition with YM155 or terameprocol. Our results suggest that targeting of CDKs and/or survivin in post-transplant EBV-LPD by specific inhibitors might be an important approach to control and eliminate EBV-transformed B-cells that should be further considered.


Assuntos
Linfócitos B/metabolismo , Linfócitos B/virologia , Proteína Quinase CDC2/genética , Infecções por Vírus Epstein-Barr/genética , Proteínas Inibidoras de Apoptose/genética , Transtornos Linfoproliferativos/genética , Transplante de Órgãos/efeitos adversos , Apoptose/genética , Proteína Quinase CDC2/metabolismo , Ciclo Celular/genética , Ciclina B1/genética , Ciclina B1/metabolismo , Infecções por Vírus Epstein-Barr/etiologia , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Expressão Gênica , Herpesvirus Humano 4 , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/metabolismo , Survivina , Transformação Genética , Regulação para Cima
20.
Blood ; 117(11): 3076-86, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21252091

RESUMO

Human hematolymphoid mice have become valuable tools for the study of human hematopoiesis and uniquely human pathogens in vivo. Recent improvements in xenorecipient strains allow for long-term reconstitution with a human immune system. However, certain hematopoietic lineages, for example, the myeloid lineage, are underrepresented, possibly because of the limited cross-reactivity of murine and human cytokines. Therefore, we created a nonobese diabetic/severe combined immunodeficiency/interleukin-2 receptor-γ-null (NOD-SCID IL2Rγ(null)) mouse strain that expressed human stem cell factor, granulocyte-macrophage colony-stimulating factor, and interleukin-3, termed NSG-SGM3. Transplantation of CD34(+) human hematopoietic stem cells into NSG-SGM3 mice led to robust human hematopoietic reconstitution in blood, spleen, bone marrow, and liver. Human myeloid cell frequencies, specifically, myeloid dendritic cells, were elevated in the bone marrow of humanized NSG-SGM3 mice compared with nontransgenic NSG recipients. Most significant, however, was the increase in the CD4(+)FoxP3(+) regulatory T-cell population in all compartments analyzed. These CD4(+)FoxP3(+) regulatory T cells were functional, as evidenced by their ability to suppress T-cell proliferation. In conclusion, humanized NSG-SGM3 mice might serve as a useful model to study human regulatory T-cell development in vivo, but this unexpected lineage skewing also highlights the importance of adequate spatiotemporal expression of human cytokines for future xenorecipient strain development.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Subunidade gama Comum de Receptores de Interleucina/deficiência , Interleucina-3/metabolismo , Fator de Células-Tronco/metabolismo , Linfócitos T Reguladores/citologia , Animais , Linhagem da Célula , Proliferação de Células , Hematopoese/imunologia , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Fenótipo , Linfócitos T Reguladores/imunologia , Timo/citologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa