Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37461447

RESUMEN

Standard preclinical human tumor models lack a human tumor stroma. However, as stroma contributes to therapeutic resistance, the lack of human stroma may make current models less stringent for testing new therapies. To address this, using patient-derived tumor cells, patient derived cancer-associated mesenchymal stem/progenitor cells, and human endothelial cells, we created a Human Stroma-Patient Derived Xenograft (HS-PDX) tumor model. HS-PDX, compared to the standard PDX model, demonstrate greater resistance to targeted therapy and chemotherapy, and better reflect patient response to therapy. Furthermore, HS-PDX can be grown in mice with humanized bone marrow to create humanized immune stroma patient-derived xenograft (HIS-PDX) models. The HIS-PDX model contains human connective tissues, vascular and immune cell infiltrates. RNA sequencing analysis demonstrated a 94-96% correlation with primary human tumor. Using this model, we demonstrate the impact of human tumor stroma on response to CAR-T cell therapy and immune checkpoint inhibitor therapy. We show an immunosuppressive role for human tumor stroma and that this model can be used to identify immunotherapeutic combinations to overcome stromally mediated immunosuppression. Combined, our data confirm a critical role for human stoma in therapeutic response and indicate that HIS-PDX can be an important tool for preclinical drug testing. Statement of Significance: We developed a tumor model with human stromal, vascular, and immune cells. This model mirrors patient response to chemotherapy, targeted therapy, and immunotherapy, and can be used to study therapy resistance.

2.
Front Immunol ; 13: 881607, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35669780

RESUMEN

Gamma-delta (γδ) T cells recognize antigens in a major histocompatibility complex (MHC) independent and have cytotoxic capability. Human immunodeficiency virus (HIV) infection reduces the proportion of the Vδ2 cell subset compared to the Vδ1 cell subset of γδ T cells in the blood in most infected individuals, except for elite controllers. The capacity of Vδ2 T cells to kill HIV-infected targets has been demonstrated in vitro, albeit in vivo confirmatory studies are lacking. Here, we provide the first characterization of γδ T cell-HIV interactions in bone marrow-liver-thymus (BLT) humanized mice and examined the immunotherapeutic potential of Vδ2 T cells in controlling HIV replication in vivo. We demonstrate a reduced proportion of Vδ2 T cells and an increased proportion of Vδ1 T cells in HIV-infected BLT humanized mice, like in HIV-positive individuals. HIV infection in BLT humanized mice also impaired the ex vivo expansion of Vδ2 T cells, like in HIV-positive individuals. Adoptive transfer of activated Vδ2 T cells did not control HIV replication during cell-associated HIV transmission in BLT humanized mice but instead exacerbated viremia, suggesting that Vδ2 T cells may serve as early targets for HIV replication. Our findings demonstrate that BLT humanized mice can model γδ T cell-HIV interactions in vivo.


Asunto(s)
Infecciones por VIH , Linfocitos Intraepiteliales , Animales , Médula Ósea , Modelos Animales de Enfermedad , Humanos , Hígado , Ratones , Receptores de Antígenos de Linfocitos T gamma-delta
4.
Sci Total Environ ; : 142830, 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33071142

RESUMEN

This article has been withdrawn at the request of the authors and the editors. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.

5.
Sci Rep ; 10(1): 14598, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32884084

RESUMEN

The human skin is a significant barrier for protection against pathogen transmission. Rodent models used to investigate human-specific pathogens that target the skin are generated by introducing human skin grafts to immunocompromised rodent strains. Infection-induced immunopathogenesis has been separately studied in humanized rodent models developed with human lymphoid tissue and hematopoietic stem cell transplants. Successful co-engraftment of human skin, autologous lymphoid tissues, and autologous immune cells in a rodent model has not yet been achieved, though it could provide a means of studying the human immune response to infection in the human skin. Here, we introduce the human Skin and Immune System (hSIS)-humanized NOD-scid IL2Rγnull (NSG) mouse and Sprague-Dawley-Rag2tm2hera Il2rγtm1hera (SRG) rat models, co-engrafted with human full-thickness fetal skin, autologous fetal lymphoid tissues, and autologous fetal liver-derived hematopoietic stem cells. hSIS-humanized rodents demonstrate the development of human full-thickness skin, along with autologous lymphoid tissues, and autologous immune cells. These models also support human skin infection following intradermal inoculation with community-associated methicillin-resistant Staphylococcus aureus. The co-engraftment of these human skin and immune system components into a single humanized rodent model could provide a platform for studying human skin infections.


Asunto(s)
Células Sanguíneas/inmunología , Tejido Linfoide/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Trasplante de Piel , Piel/inmunología , Infecciones Estafilocócicas/inmunología , Replicación Viral/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Piel/patología , Infecciones Estafilocócicas/terapia , Trasplante Autólogo
6.
Retrovirology ; 17(1): 8, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32276640

RESUMEN

The development of safe and effective combination antiretroviral therapies for human immunodeficiency virus (HIV) infection over the past several decades has significantly reduced HIV-associated morbidity and mortality. Additionally, antiretroviral drugs have provided an effective means of protection against HIV transmission. Despite these advances, significant limitations exist; namely, the inability to eliminate HIV reservoirs, the inability to reverse lymphoid tissues damage, and the lack of an effective vaccine for preventing HIV transmission. Evaluation of the safety and efficacy of therapeutics and vaccines for eliminating HIV reservoirs and preventing HIV transmission requires robust in vivo models. Since HIV is a human-specific pathogen, that targets hematopoietic lineage cells and lymphoid tissues, in vivo animal models for HIV-host interactions require incorporation of human hematopoietic lineage cells and lymphoid tissues. In this review, we will discuss the construction of mouse models with human lymphoid tissues and/or hematopoietic lineage cells, termed, human immune system (HIS)-humanized mice. These HIS-humanized mouse models can support the development of functional human innate and adaptive immune cells, along with primary (thymus) and secondary (spleen) lymphoid tissues. We will discuss applications of HIS-humanized mouse models in evaluating the safety and efficacy of therapeutics against HIV reservoirs and associated immunopathology, and delineate the human immune response elicited by candidate HIV vaccines. In addition to focusing on how these HIS-humanized mouse models have already furthered our understanding of HIV and contributed to HIV therapeutics development, we discuss how emerging HIS-humanized rat models could address the limitations of HIS-mouse models.


Asunto(s)
Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/prevención & control , VIH-1/efectos de los fármacos , Animales , Antirretrovirales/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Ratones , Ratas , Bazo/patología
7.
J Virol ; 94(8)2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-31969438

RESUMEN

As a long-acting formulation of the nonnucleoside reverse transcriptase inhibitor rilpivirine (RPV LA) has been proposed for use as preexposure prophylaxis (PrEP) and the prevalence of transmitted RPV-resistant viruses can be relatively high, we evaluated the efficacy of RPV LA to inhibit vaginal transmission of RPV-resistant HIV-1 in humanized mice. Vaginal challenges of wild-type (WT), Y181C, and Y181V HIV-1 were performed in mice left untreated or after RPV PrEP. Plasma viremia was measured for 7 to 10 weeks, and single-genome sequencing was performed on plasma HIV-1 RNA in mice infected during PrEP. RPV LA significantly prevented vaginal transmission of WT HIV-1 and Y181C HIV-1, which is 3-fold resistant to RPV. However, it did not prevent transmission of Y181V HIV-1, which has 30-fold RPV resistance in the viruses used for this study. RPV LA did delay WT HIV-1 dissemination in infected animals until genital and plasma RPV concentrations waned. Animals that became infected despite RPV LA PrEP did not acquire new RPV-resistant mutations above frequencies in untreated mice or untreated people living with HIV-1, and the mutations detected conferred low-level resistance. These data suggest that high, sustained concentrations of RPV were required to inhibit vaginal transmission of HIV-1 with little or no resistance to RPV but could not inhibit virus with high resistance. HIV-1 did not develop high-level or high-frequency RPV resistance in the majority of mice infected after RPV LA treatment. However, the impact of low-frequency RPV resistance on virologic outcome during subsequent antiretroviral therapy still is unclear.IMPORTANCE The antiretroviral drug rilpivirine was developed into a long-acting formulation (RPV LA) to improve adherence for preexposure prophylaxis (PrEP) to prevent HIV-1 transmission. A concern is that RPV LA will not inhibit transmission of drug-resistant HIV-1 and may select for drug-resistant virus. In female humanized mice, we found that RPV LA inhibited vaginal transmission of WT or 3-fold RPV-resistant HIV-1 but not virus with 30-fold RPV resistance. In animals that became infected despite RPV LA PrEP, WT HIV-1 dissemination was delayed until genital and plasma RPV concentrations waned. RPV resistance was detected at similar low frequencies in untreated and PrEP-treated mice that became infected. These results indicate the importance of maintaining RPV at a sustained threshold after virus exposure to prevent dissemination of HIV-1 after vaginal infection and low-frequency resistance mutations conferred low-level resistance, suggesting that RPV resistance is difficult to develop after HIV-1 infection during RPV LA PrEP.


Asunto(s)
Fármacos Anti-VIH/farmacología , Infecciones por VIH/prevención & control , Infecciones por VIH/transmisión , VIH-1/efectos de los fármacos , Profilaxis Pre-Exposición/métodos , Rilpivirina/farmacología , Vagina/virología , Animales , Modelos Animales de Enfermedad , Farmacorresistencia Viral/efectos de los fármacos , Femenino , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , Ratones , Mutación , Inhibidores de la Transcriptasa Inversa/farmacología , Replicación Viral/efectos de los fármacos , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
8.
Curr HIV Res ; 18(1): 19-28, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31870268

RESUMEN

The main advantage of animal models of infectious diseases over in vitro studies is the gain in the understanding of the complex dynamics between the immune system and the pathogen. While small animal models have practical advantages over large animal models, it is crucial to be aware of their limitations. Although the small animal model at least needs to be susceptible to the pathogen under study to obtain meaningful data, key elements of pathogenesis should also be reflected when compared to humans. Well-designed small animal models for HIV, hepatitis viruses and tuberculosis require, additionally, a thorough understanding of the similarities and differences in the immune responses between humans and small animals and should incorporate that knowledge into the goals of the study. To discuss these considerations, the NIAID hosted a workshop on 'Small Animal Models for HIV, Hepatitis B, and Tuberculosis' on May 30, 2019. Highlights of the workshop are outlined below.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por VIH/patología , VIH-1/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B/patología , Mycobacterium tuberculosis/inmunología , Tuberculosis/patología , Animales , Coinfección/microbiología , Cobayas , Infecciones por VIH/inmunología , Hepatitis B/inmunología , Humanos , Macaca mulatta , Marmota , Ratones , National Institute of Allergy and Infectious Diseases (U.S.) , Conejos , Tuberculosis/inmunología , Estados Unidos
9.
Mol Carcinog ; 58(9): 1612-1622, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31062422

RESUMEN

Considerable progress has been made during the past 20 years towards elucidating the role of peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) in skin cancer. In 1999, the original notion that PPARß/δ was involved with epithelial cell function was postulated based on a correlation between PPARß/δ expression and the induction of messenger RNAs encoding proteins that mediate terminal differentiation in keratinocytes. Subsequent studies definitively revealed that PPARß/δ could induce terminal differentiation and inhibit proliferation of keratinocytes. Molecular mechanisms have since been discovered to explain how this nuclear receptor can be targeted for preventing and treating skin cancer. This includes the regulation of terminal differentiation, mitotic signaling, endoplasmic reticulum stress, and cellular senescence. Interestingly, the effects of activating PPARß/δ can preferentially target keratinocytes with genetic mutations associated with skin cancer. This review provides the history and current understanding of how PPARß/δ can be targeted for both nonmelanoma skin cancer and melanoma and postulates how future approaches that modulate PPARß/δ signaling may be developed for the prevention and treatment of these diseases.


Asunto(s)
PPAR delta/metabolismo , PPAR-beta/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Humanos , Queratinocitos/metabolismo , Melanoma/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/fisiología
10.
JCI Insight ; 3(18)2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30232273

RESUMEN

A major pathogenic feature associated with HIV infection is lymphoid fibrosis, which persists during antiretroviral therapy (ART). Lymphoid tissues play critical roles in the generation of antigen-specific immune response, and fibrosis disrupts the stromal network of lymphoid tissues, resulting in impaired immune cell trafficking and function, as well as immunodeficiency. Developing an animal model for investigating the impact of HIV infection-induced lymphoid tissue fibrosis on immunodeficiency and immune cell impairment is critical for therapeutics development and clinical translation. Said model will enable in vivo mechanistic studies, thus complementing the well-established surrogate model of SIV infection-induced lymphoid tissue fibrosis in macaques. We developed a potentially novel human immune system-humanized mouse model by coengrafting autologous fetal thymus, spleen, and liver organoids under the kidney capsule, along with i.v. injection of autologous fetal liver-derived hematopoietic stem cells, thus termed the BM-liver-thymus-spleen (BLTS) humanized mouse model. BLTS humanized mouse model supports development of human immune cells and human lymphoid organoids (human thymus and spleen organoids). HIV infection in BLTS humanized mice results in progressive fibrosis in human lymphoid tissues, which was associated with immunodeficiency in the lymphoid tissues, and lymphoid tissue fibrosis persists during ART, thus recapitulating clinical outcomes.


Asunto(s)
Fibrosis/inmunología , Infecciones por VIH/inmunología , Hígado/inmunología , Tejido Linfoide/inmunología , Bazo/inmunología , Timo/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Trasplante de Tejido Fetal , Fibrosis/patología , Infecciones por VIH/tratamiento farmacológico , Células Madre Hematopoyéticas , Humanos , Hígado/patología , Trasplante de Hígado , Tejido Linfoide/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Organogénesis , Bazo/patología , Bazo/trasplante , Timo/patología , Timo/trasplante , Trasplante Heterólogo
11.
Sci Rep ; 6: 39520, 2016 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-28000758

RESUMEN

The immuno-pathogenic mechanisms of chronic hepatitis C virus (HCV) infection remain to be elucidated and pose a major hurdle in treating or preventing chronic HCV-induced advanced liver diseases such as cirrhosis. Macrophages are a major component of the inflammatory milieu in chronic HCV-induced liver disease, and are generally derived from circulating inflammatory monocytes; however very little is known about their role in liver diseases. To investigate the activation and role of macrophages in chronic HCV-induced liver fibrosis, we utilized a recently developed humanized mouse model with autologous human immune and liver cells, human liver and blood samples and cell culture models of monocyte/macrophage and/or hepatic stellate cell activation. We showed that M2 macrophage activation was associated with liver fibrosis during chronic HCV infection in the livers of both humanized mice and patients, and direct-acting antiviral therapy attenuated M2 macrophage activation and associated liver fibrosis. We demonstrated that supernatant from HCV-infected liver cells activated human monocytes/macrophages with M2-like phenotypes. Importantly, HCV-activated monocytes/macrophages promoted hepatic stellate cell activation. These results suggest a critical role for M2 macrophage induction in chronic HCV-associated immune dysregulation and liver fibrosis.


Asunto(s)
Hepatitis C Crónica/patología , Cirrosis Hepática/patología , Activación de Macrófagos , Macrófagos/citología , Animales , Antivirales/farmacología , Enfermedad Crónica , Modelos Animales de Enfermedad , Hepacivirus , Células Estrelladas Hepáticas/citología , Hepatitis C Crónica/complicaciones , Humanos , Sistema Inmunológico , Inflamación , Hígado/fisiopatología , Hígado/virología , Cirrosis Hepática/complicaciones , Ratones
12.
Antiviral Res ; 121: 1-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26099683

RESUMEN

Despite the availability of a preventive vaccine, chronic hepatitis B virus (HBV) infection-induced liver diseases continue to be a major global public health problem. HBV naturally infects only humans and chimpanzees. This narrow host range has hindered our ability to study the characteristics of the virus and how it interacts with its host. It is thus important to establish small animal models to study HBV infection, persistence, clearance and the immunopathogenesis of chronic hepatitis B. In this review, we briefly summarize currently available animal models for HBV research, then focus on mouse models, especially the recently developed humanized mice that can support HBV infection and immunopathogenesis in vivo. This article is part of a symposium in Antiviral Research on "From the discovery of the Australia antigen to the development of new curative therapies for hepatitis B: an unfinished story."


Asunto(s)
Modelos Animales de Enfermedad , Hepatitis B/tratamiento farmacológico , Hepatitis B/patología , Animales , Hepatitis B/virología , Humanos , Ratones SCID , Ratones Transgénicos
13.
Korean J Parasitol ; 52(6): 695-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25548425

RESUMEN

Chronic Opisthorchis viverrini-induced hepatobiliary disease is associated with significant leukocyte infiltration, including activated macrophages; however, the polarization of infiltrating macrophages remains to be fully characterized. In this study, we characterized macrophage polarization and phenotype in chronic O. viverrini-induced hepatobiliary disease in humans and hamsters using gene expression and histochemical analysis. Chronic O. viverrini infection and associated hepatobiliary diseases were associated with iron loaded M2-like macrophages in both humans and hamsters. This study provides suggestive evidence that iron loaded M2-like macrophages promote hepatobiliary disease in chronic O. viverrini infection.


Asunto(s)
Hierro/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/parasitología , Macrófagos/inmunología , Opistorquiasis/complicaciones , Opistorquiasis/patología , Opisthorchis/aislamiento & purificación , Animales , Cricetinae , Perfilación de la Expresión Génica , Histocitoquímica , Humanos , Inmunohistoquímica , Macrófagos/metabolismo , Mesocricetus
14.
Methods Mol Biol ; 1213: 379-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25173399

RESUMEN

Several human hepatotropic pathogens including chronic hepatitis C virus (HCV) have narrow species restriction, thus hindering research and therapeutics development against these pathogens. Developing a rodent model that accurately recapitulates hepatotropic pathogens infection, human immune response, chronic hepatitis, and associated immunopathogenesis is essential for research and therapeutics development. Here, we describe the recently developed AFC8 humanized liver- and immune system-mouse model for studying chronic hepatitis C virus and associated human immune response, chronic hepatitis, and liver fibrosis.


Asunto(s)
Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatitis C/virología , Animales , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hígado/citología , Hígado/inmunología , Hígado/patología , Hígado/virología , Regeneración Hepática , Ratones , Ratones Transgénicos , Trasplante de Células Madre , Células Madre/citología , Células Madre/metabolismo
15.
Carcinogenesis ; 35(7): 1602-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24639079

RESUMEN

Whether peroxisome proliferator-activated receptor ß/δ (PPARß/δ) reduces skin tumorigenesis by altering aryl hydrocarbon receptor (AHR)-dependent activities was examined. Polycyclic aromatic hydrocarbons (PAH) increased expression of cytochrome P4501A1 (CYP1A1), CYP1B1 and phase II xenobiotic metabolizing enzymes in wild-type skin and keratinocytes. Surprisingly, this effect was not found in Pparß/δ-null skin and keratinocytes. Pparß/δ-null keratinocytes exhibited decreased AHR occupancy and histone acetylation on the Cyp1a1 promoter in response to a PAH compared with wild-type keratinocytes. Bisulfite sequencing of the Cyp1a1 promoter and studies using a DNA methylation inhibitor suggest that PPARß/δ promotes demethylation of the Cyp1a1 promoter. Experiments with human HaCaT keratinocytes stably expressing shRNA against PPARß/δ also support this conclusion. Consistent with the lower AHR-dependent activities in Pparß/δ-null mice compared with wild-type mice, 7,12-dimethylbenz[a]anthracene (DMBA)-induced skin tumorigenesis was inhibited in Pparß/δ-null mice compared with wild-type. Results from these studies demonstrate that PPARß/δ is required to mediate complete carcinogenesis by DMBA. The mechanisms underlying this PPARß/δ-dependent reduction of AHR signaling by PAH are not due to alterations in the expression of AHR auxiliary proteins, ligand binding or AHR nuclear translocation between genotypes, but are likely influenced by PPARß/δ-dependent demethylation of AHR target gene promoters including Cyp1a1 that reduces AHR accessibility as shown by reduced promoter occupancy. This PPARß/δ/AHR crosstalk is unique to keratinocytes and conserved between mice and humans.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Queratinocitos/metabolismo , PPAR delta/fisiología , PPAR-beta/fisiología , Receptores de Hidrocarburo de Aril/fisiología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Western Blotting , Carcinógenos/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Inmunoprecipitación de Cromatina , Dermis/citología , Dermis/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Queratinocitos/citología , Ratones , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
16.
PLoS Pathog ; 10(3): e1004032, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24651854

RESUMEN

The mechanisms of chronic HBV infection and immunopathogenesis are poorly understood due to a lack of a robust small animal model. Here we report the development of a humanized mouse model with both human immune system and human liver cells by reconstituting the immunodeficient A2/NSG (NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ mice with human HLA-A2 transgene) with human hematopoietic stem cells and liver progenitor cells (A2/NSG-hu HSC/Hep mice). The A2/NSG-hu HSC/Hep mouse supported HBV infection and approximately 75% of HBV infected mice established persistent infection for at least 4 months. We detected human immune responses, albeit impaired in the liver, chronic liver inflammation and liver fibrosis in infected animals. An HBV neutralizing antibody efficiently inhibited HBV infection and associated liver diseases in humanized mice. In addition, we found that the HBV mediated liver disease was associated with high level of infiltrated human macrophages with M2-like activation phenotype. Importantly, similar M2-like macrophage accumulation was confirmed in chronic hepatitis B patients with liver diseases. Furthermore, gene expression analysis showed that induction of M2-like macrophage in the liver is associated with accelerated liver fibrosis and necrosis in patients with acute HBV-induced liver failure. Lastly, we demonstrate that HBV promotes M2-like activation in both M1 and M2 macrophages in cell culture studies. Our study demonstrates that the A2/NSG-hu HSC/Hep mouse model is valuable in studying HBV infection, human immune responses and associated liver diseases. Furthermore, results from this study suggest a critical role for macrophage polarization in hepatitis B virus-induced immune impairment and liver pathology.


Asunto(s)
Modelos Animales de Enfermedad , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/patología , Cirrosis Hepática/patología , Macrófagos/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Células Madre Hematopoyéticas/citología , Virus de la Hepatitis B , Hepatocitos/citología , Hepatocitos/trasplante , Humanos , Inmunohistoquímica , Cirrosis Hepática/etiología , Ratones , Ratones Endogámicos NOD , Células Madre/citología , Trasplante Heterólogo
17.
J Gastroenterol Hepatol ; 28 Suppl 1: 120-4, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23855307

RESUMEN

Hepatitis B virus (HBV) and hepatitis C virus (HCV) infect and replicate primarily in human hepatocytes. Few reliable and easy accessible animal models are available for studying the immune system's contribution to the liver disease progression during hepatitis virus infection. Humanized mouse models reconstituted with human hematopoietic stem cells (HSCs) have been developed to study human immunology, human immunodeficiency virus 1 infection, and immunopathogenesis. However, a humanized mouse model engrafted with both human immune and human liver cells is needed to study infection and immunopathogenesis of HBV/HCV infection in vivo. We have recently developed the humanized mouse model with both human immune and human liver cells (AFC8-hu HSC/Hep) to study immunopathogenesis and therapy of HCV infection in vivo. In this review, we summarize the current models of HBV/HCV infection and their limitations in immunopathogenesis. We will then present our recent findings of HCV infection and immunopathogenesis in the AFC8-hu HSC/Hep mouse, which supports HCV infection, human T-cell response and associated liver pathogenesis. Inoculation of humanized mice with primary HCV isolates resulted in long-term HCV infection. HCV infection induced elevated infiltration of human immune cells in the livers of HCV-infected humanized mice. HCV infection also induced HCV-specific T-cell immune response in lymphoid tissues of humanized mice. Additionally, HCV infection induced liver fibrosis in humanized mice. Anti-human alpha smooth muscle actin (αSMA) staining showed elevated human hepatic stellate cell activation in HCV-infected humanized mice. We discuss the limitation and future improvements of the AFC8-hu HSC/Hep mouse model and its application in evaluating novel therapeutics, as well as studying both HCV and HBV infection, human immune responses, and associated human liver fibrosis and cancer.


Asunto(s)
Modelos Animales de Enfermedad , Hepatitis B/inmunología , Hepatitis B/terapia , Hepatitis C/inmunología , Hepatitis C/terapia , Hígado/inmunología , Animales , Fibrosis , Hepacivirus/inmunología , Células Estrelladas Hepáticas/inmunología , Virus de la Hepatitis B/inmunología , Humanos , Hígado/patología , Ratones , Ratones SCID , Ratones Transgénicos , Linfocitos T/inmunología
18.
Nat Protoc ; 7(9): 1608-17, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22899330

RESUMEN

Establishing a small animal model that accurately recapitulates hepatotropic pathogens, including hepatitis C virus (HCV) infection and immunopathogenesis, is essential for the study of hepatitis virus-induced liver disease and for therapeutics development. This protocol describes our recently developed humanized mouse model for studying HCV and other hepatotropic infections, human immune response and hepatitis and liver fibrosis. The first 5-h stage is the isolation of human liver progenitor and hematopoietic stem cells from fetal liver. Next, AFC8 immunodeficient mice are transplanted with the isolated progenitor/stem cells. This generally takes 2 h. The transplanted mice are then treated for a month with the mouse liver apoptosis-inducing AFC8 dimerizer and left for an additional 2-month period to permit human liver and immune cell growth as well as system reconstitution and development before inoculation with HCV clinical isolates. HCV infection, human immune response and liver disease are observed with high incidence from approximately 2 months after inoculation.


Asunto(s)
Quimera/inmunología , Células Madre Hematopoyéticas/citología , Hepatitis C/inmunología , Hígado/fisiopatología , Modelos Animales , Animales , Proteínas de Unión al ADN/genética , Dimerización , Trasplante de Células Madre Hematopoyéticas , Humanos , Hígado/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tacrolimus/análogos & derivados
19.
Mol Cell Biol ; 32(11): 2065-82, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473992

RESUMEN

The role of peroxisome proliferator-activated receptor ß/δ (PPARß/δ) in Harvey sarcoma ras (Hras)-expressing cells was examined. Ligand activation of PPARß/δ caused a negative selection with respect to cells expressing higher levels of the Hras oncogene by inducing a mitotic block. Mitosis-related genes that are predominantly regulated by E2F were induced to a higher level in HRAS-expressing Pparß/δ-null keratinocytes compared to HRAS-expressing wild-type keratinocytes. Ligand-activated PPARß/δ repressed expression of these genes by direct binding with p130/p107, facilitating nuclear translocation and increasing promoter recruitment of p130/p107. These results demonstrate a novel mechanism of PPARß/δ cross talk with E2F signaling. Since cotreatment with a PPARß/δ ligand and various mitosis inhibitors increases the efficacy of increasing G2/M arrest, targeting PPARß/δ in conjunction with mitosis inhibitors could become a suitable option for development of new multitarget strategies for inhibiting RAS-dependent tumorigenesis.


Asunto(s)
Factor de Transcripción E2F4/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Mitosis , PPAR delta/metabolismo , PPAR-beta/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Receptor Cross-Talk , Animales , Células Cultivadas , Queratinocitos/metabolismo , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
20.
Gastroenterology ; 140(4): 1334-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21237170

RESUMEN

BACKGROUND & AIMS: Studies of hepatitis C virus (HCV) infection, immunopathogenesis, and resulting liver diseases have been hampered by the lack of a small animal model. We developed humanized mice with human immune system and liver tissues to improve the studies of hepatitis C virus pathogenesis and treatment. METHODS: To promote engraftment of human hepatocytes, we expressed a fusion protein of the FK506 binding protein (FKBP) and caspase 8 under control of the albumin promoter (AFC8), which induces liver cell death, in Balb/C Rag2(-/-) γC-null mice. Cotransplantation of human CD34(+) human hematopoietic stem cells (HSC) and hepatocyte progenitors into the transgenic mice led to efficient engraftment of human leukocytes and hepatocytes. We then infected these humanized mice (AFC8-hu HSC/Hep) with primary HCV isolates and studied HCV-induced immune responses and liver diseases. RESULTS: AFC8-hu HSC/Hep mice supported HCV infection in the liver and generated a human immune T-cell response against HCV. HCV infection induced liver inflammation, hepatitis, and fibrosis, which correlated with activation of stellate cells and expression of human fibrogenic genes. CONCLUSIONS: AFC8-hu HSC/Hep mice are a useful model of HCV infection, the immune response, and liver disease because they contain human immune system and liver cells. These mice become infected with HCV, generate a specific immune response against the virus, and develop liver diseases that include hepatitis and fibrosis. This model might also be used to develop therapeutics for HCV infection.


Asunto(s)
Modelos Animales de Enfermedad , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Hepatocitos/trasplante , Ratones Transgénicos , Animales , Caspasa 8/genética , Caspasa 8/inmunología , Proteínas de Unión al ADN/genética , Femenino , Hepatitis C Crónica/genética , Hepatitis C Crónica/patología , Hepatocitos/inmunología , Hepatocitos/patología , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos BALB C , Células Madre/inmunología , Células Madre/patología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/inmunología , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...