Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
bioRxiv ; 2023 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-37461447

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-35669780

RESUMO

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.


Assuntos
Infecções por HIV , Linfócitos Intraepiteliais , Animais , Medula Óssea , Modelos Animais de Doenças , Humanos , Fígado , Camundongos , Receptores de Antígenos de Linfócitos T gama-delta
3.
Curr HIV Res ; 18(1): 19-28, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31870268

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Infecções por HIV/patologia , HIV-1/imunologia , Vírus da Hepatite B/imunologia , Hepatite B/patologia , Mycobacterium tuberculosis/imunologia , Tuberculose/patologia , Animais , Coinfecção/microbiologia , Cobaias , Infecções por HIV/imunologia , Hepatite B/imunologia , Humanos , Macaca mulatta , Marmota , Camundongos , National Institute of Allergy and Infectious Diseases (U.S.) , Coelhos , Tuberculose/imunologia , Estados Unidos
4.
Mol Carcinog ; 58(9): 1612-1622, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31062422

RESUMO

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.


Assuntos
PPAR delta/metabolismo , PPAR beta/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Queratinócitos/metabolismo , Melanoma/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia
5.
Sci Rep ; 6: 39520, 2016 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-28000758

RESUMO

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.


Assuntos
Hepatite C Crônica/patologia , Cirrose Hepática/patologia , Ativação de Macrófagos , Macrófagos/citologia , Animais , Antivirais/farmacologia , Doença Crônica , Modelos Animais de Doenças , Hepacivirus , Células Estreladas do Fígado/citologia , Hepatite C Crônica/complicações , Humanos , Sistema Imunitário , Inflamação , Fígado/fisiopatologia , Fígado/virologia , Cirrose Hepática/complicações , Camundongos
6.
Antiviral Res ; 121: 1-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26099683

RESUMO

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."


Assuntos
Modelos Animais de Doenças , Hepatite B/tratamento farmacológico , Hepatite B/patologia , Animais , Hepatite B/virologia , Humanos , Camundongos SCID , Camundongos Transgênicos
7.
Korean J Parasitol ; 52(6): 695-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25548425

RESUMO

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.


Assuntos
Ferro/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/parasitologia , Macrófagos/imunologia , Opistorquíase/complicações , Opistorquíase/patologia , Opisthorchis/isolamento & purificação , Animais , Cricetinae , Perfilação da Expressão Gênica , Histocitoquímica , Humanos , Imuno-Histoquímica , Macrófagos/metabolismo , Mesocricetus
8.
Methods Mol Biol ; 1213: 379-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25173399

RESUMO

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.


Assuntos
Hepacivirus/imunologia , Hepatite C/imunologia , Hepatite C/virologia , Animais , Modelos Animais de Doenças , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fígado/citologia , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Regeneração Hepática , Camundongos , Camundongos Transgênicos , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo
9.
Carcinogenesis ; 35(7): 1602-12, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24639079

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Queratinócitos/metabolismo , PPAR delta/fisiologia , PPAR beta/fisiologia , Receptores de Hidrocarboneto Arílico/fisiologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Western Blotting , Carcinógenos/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Imunoprecipitação da Cromatina , Derme/citologia , Derme/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Técnicas Imunoenzimáticas , Queratinócitos/citologia , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
10.
PLoS Pathog ; 10(3): e1004032, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24651854

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Hepatite B Crônica/imunologia , Hepatite B Crônica/patologia , Cirrose Hepática/patologia , Macrófagos/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Células-Tronco Hematopoéticas/citologia , Vírus da Hepatite B , Hepatócitos/citologia , Hepatócitos/transplante , Humanos , Imuno-Histoquímica , Cirrose Hepática/etiologia , Camundongos , Camundongos Endogâmicos NOD , Células-Tronco/citologia , Transplante Heterólogo
11.
J Gastroenterol Hepatol ; 28 Suppl 1: 120-4, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23855307

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Hepatite B/imunologia , Hepatite B/terapia , Hepatite C/imunologia , Hepatite C/terapia , Fígado/imunologia , Animais , Fibrose , Hepacivirus/imunologia , Células Estreladas do Fígado/imunologia , Vírus da Hepatite B/imunologia , Humanos , Fígado/patologia , Camundongos , Camundongos SCID , Camundongos Transgênicos , Linfócitos T/imunologia
12.
Nat Protoc ; 7(9): 1608-17, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22899330

RESUMO

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.


Assuntos
Quimera/imunologia , Células-Tronco Hematopoéticas/citologia , Hepatite C/imunologia , Fígado/fisiopatologia , Modelos Animais , Animais , Proteínas de Ligação a DNA/genética , Dimerização , Transplante de Células-Tronco Hematopoéticas , Humanos , Fígado/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Tacrolimo/análogos & derivados
13.
Mol Cell Biol ; 32(11): 2065-82, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22473992

RESUMO

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.


Assuntos
Fator de Transcrição E2F4/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Mitose , PPAR delta/metabolismo , PPAR beta/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/biossíntese , Receptor Cross-Talk , Animais , Células Cultivadas , Queratinócitos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais
14.
Gastroenterology ; 140(4): 1334-44, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21237170

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Hepacivirus/imunologia , Hepatite C Crônica/imunologia , Hepatócitos/transplante , Camundongos Transgênicos , Animais , Caspase 8/genética , Caspase 8/imunologia , Proteínas de Ligação a DNA/genética , Feminino , Hepatite C Crônica/genética , Hepatite C Crônica/patologia , Hepatócitos/imunologia , Hepatócitos/patologia , Humanos , Cirrose Hepática/genética , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco/imunologia , Células-Tronco/patologia , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/imunologia , Transplante Heterólogo
15.
Toxicol Sci ; 113(1): 27-36, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19748995

RESUMO

Ligand activation of peroxisome proliferator-activated receptor (PPAR)-beta/delta and inhibition of cyclooxygenase-2 (COX-2) activity by nonsteroidal anti-inflammatory drugs can attenuate skin tumorigenesis. There is also evidence that attenuation of skin tumorigenesis by inhibition of COX-2 activity occurs through PPARbeta/delta-independent mechanisms. The present study examined the hypothesis that combining ligand activation of PPARbeta/delta with inhibition of COX-2 activity will cooperatively inhibit chemically induced skin tumor progression using both in vivo and ex vivo models. A two-stage chemical carcinogenesis bioassay was performed in wild-type and Pparbeta/delta-null mice. After 22 weeks, cohorts of both mouse lines were divided into four experimental groups: (1) control, (2) topical application of the PPARbeta/delta ligand GW0742, (3) dietary administration of the COX-2 inhibitor nimesulide, or (4) both GW0742 and nimesulide. Ligand activation of PPARbeta/delta did not influence skin tumor progression, while a modest decrease in skin tumor multiplicity was observed with dietary nimesulide. Interestingly, the combined treatment of GW0742 and nimesulide increased the efficacy of the decrease in papilloma multiplicity for 6 weeks in wild-type mice, but this effect was not found at later time points and was not found in similarly treated Pparbeta/delta-null mice. Neoplastic keratinocyte lines cultured with GW0742 and nimesulide also exhibited enhanced inhibition of cell proliferation coincident with increased expression of Keratin messenger RNAs. Results from these studies support the hypothesis that combining ligand activation of PPARbeta/delta with inhibition of COX-2 activity can inhibit chemically induced skin tumor progression by modulating differentiation.


Assuntos
Anticarcinógenos/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , PPAR delta/agonistas , PPAR beta/agonistas , Neoplasias Cutâneas/prevenção & controle , Sulfonamidas/farmacologia , Tiazóis/farmacologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dinoprostona/metabolismo , Modelos Animais de Doenças , Feminino , Queratinócitos/efeitos dos fármacos , Queratinócitos/enzimologia , Queratinócitos/patologia , Queratinas/genética , Ceratoacantoma/enzimologia , Ceratoacantoma/prevenção & controle , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR delta/deficiência , PPAR delta/genética , PPAR delta/metabolismo , PPAR beta/deficiência , PPAR beta/genética , PPAR beta/metabolismo , Papiloma/enzimologia , Papiloma/prevenção & controle , RNA Mensageiro/metabolismo , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Fatores de Tempo
16.
Carcinogenesis ; 29(12): 2406-14, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18799709

RESUMO

Peroxisome proliferator-activated receptor (PPAR)beta/delta-null mice exhibit enhanced tumorigenesis in a two-stage chemical carcinogenesis model as compared with wild-type mice. Previous work showed that ligand activation of PPARbeta/delta induces terminal differentiation and inhibits proliferation of primary keratinocytes, and this effect does not occur in the absence of PPARbeta/delta expression. In the present studies, the effect of ligand activation of PPARbeta/delta on skin tumorigenesis was examined using both in vivo and ex vivo skin carcinogenesis models. Inhibition of chemically induced skin tumorigenesis was observed in wild-type mice administered GW0742, and this effect was likely the result of ligand-induced terminal differentiation and inhibition of replicative DNA synthesis. These effects were not found in similarly treated PPARbeta/delta-null mice. Ligand activation of PPARbeta/delta also inhibited cell proliferation and induced terminal differentiation in initiated/neoplastic keratinocyte cell lines representing different stages of skin carcinogenesis. These studies suggest that topical administration of PPARbeta/delta ligands may be useful as both a chemopreventive and/or a chemotherapeutic approach to inhibit skin cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , PPAR delta/metabolismo , PPAR beta/metabolismo , Neoplasias Cutâneas/metabolismo , Acetona/toxicidade , Animais , Carcinógenos/toxicidade , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/fisiologia , Feminino , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Ligantes , Camundongos , Camundongos Mutantes , Neoplasias Cutâneas/induzido quimicamente , Acetato de Tetradecanoilforbol/toxicidade , Tiazóis/toxicidade
17.
Hepatology ; 47(1): 225-35, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18038451

RESUMO

UNLABELLED: Potential functional roles for the peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) in skeletal muscle fatty acid catabolism and epithelial carcinogenesis have recently been described. Whereas PPARbeta/delta is expressed in liver, its function in this tissue is less clear. To determine the role of PPARbeta/delta in chemically induced liver toxicity, wild-type and PPARbeta/delta-null mice were treated with azoxymethane (AOM) and markers of liver toxicity examined. Bile duct hyperplasia, regenerative hyperplasia, and increased serum alanine aminotransferase (ALT) were found in AOM-treated PPARbeta/delta-null mice, and these effects were not observed in similarly treated wild-type mice. Exacerbated carbon tetrachloride (CCl(4)) hepatoxicity was also observed in PPARbeta/delta-null as compared with wild-type mice. No differences in messenger RNAs (mRNAs) encoding cytochrome2E1 required for the metabolic activation of AOM and CCl(4) were observed between wild-type or PPARbeta/delta-null mice in response to CCl(4). Significant differences in the expression of genes reflecting enhanced nuclear factor kappa B (NF-kappaB) activity were noted in PPARbeta/delta-null mice. CONCLUSION: Results from these studies show that PPARbeta/delta is protective against liver toxicity induced by AOM and CCl(4), suggesting that this receptor is hepatoprotective against environmental chemicals that are metabolized in this tissue.


Assuntos
Intoxicação por Tetracloreto de Carbono/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , PPAR delta/deficiência , PPAR beta/deficiência , Animais , Azoximetano/intoxicação , Carcinógenos/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo
18.
Cell Signal ; 19(6): 1163-71, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17254750

RESUMO

The functional role of peroxisome proliferator-activated receptor-beta(PPARbeta; also referred to as PPARdelta) in epidermal cell growth remains controversial. Recent evidence suggests that ligand activation of PPARbeta/delta increases cell growth and inhibits apoptosis in epidermal cells. In contrast, other reports suggest that ligand activation of PPARbeta/delta leads to the induction of terminal differentiation and inhibition of cell growth. In the present study, the effect of the highly specific PPARbeta/delta ligand GW0742 on cell growth was examined using a human keratinocyte cell line (N/TERT-1) and mouse primary keratinocytes. Ligand activation of PPARbeta/delta with GW0742 prevented cell cycle progression from G1 to S phase and attenuated cell proliferation in N/TERT-1 cells. Despite specifically activating PPARbeta/delta as revealed by target gene induction, no changes in PTEN, PDK and ILK expression or downstream phosphorylation of Akt were found in either N/TERT-1 cells or primary keratinocytes. Further, altered cell growth resulting from serum withdrawal and the induction of caspase-3 activity by ultraviolet radiation were unchanged in the absence of PPARbeta/delta expression and/or the presence of GW0742. While no changes in the expression of mRNAs encoding cell cycle control proteins were found in response to GW0742, a significant decrease in the level of ERK phosphorylation was observed. Results from these studies demonstrate that ligand activation of PPARbeta/delta does not lead to an anti-apoptotic effect in either human or mouse keratinocytes, but rather, leads to inhibition of cell growth likely through the induction of terminal differentiation.


Assuntos
Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , PPAR delta/metabolismo , PPAR beta/metabolismo , Telomerase/metabolismo , Tiazóis/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Queratinócitos/enzimologia , Queratinócitos/efeitos da radiação , Ligantes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ativação Transcricional , Raios Ultravioleta
19.
Toxicol Sci ; 82(1): 170-82, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15310864

RESUMO

Administration of phthalates is known to cause toxicity and liver cancer in rodents through the activation of peroxisome proliferator-activated receptors (PPARs), and the monoesters appear to be the active metabolites that function as ligands of PPARs. There is evidence that PPARs exhibit significant species differences in response to ligand activation. In this study, the activation of mouse and human PPARalpha, PPARbeta, and PPARgamma by a broad class of phthalate monoesters was investigated using a trans-activation assay, functional analysis of PPARalpha target gene expression, and a PPARgamma-mediated differentiation assay. These studies demonstrated a range in the ability of various phthalate monoesters to activate PPARalpha, with the mouse PPARalpha generally being activated at lower concentrations and exhibiting a greater response than human PPARalpha. Similarly, a range in the trans-activation of mouse PPARbeta by phthalate monoesters was also observed, but this effect was not found with human PPARbeta. A number of phthalate monoesters activated both mouse and human PPARgamma, with similar sensitivity being exhibited by both receptors. These studies show that the potency and efficacy of phthalate monoesters for the activation of PPARalpha and PPARgamma increase with increasing side-chain length. These studies also show that mouse PPARalpha and PPARbeta are generally activated at lower concentrations of phthalate monoesters than human PPARalpha and PPARbeta, and that both mouse and human PPARgamma exhibit similar sensitivity to phthalate monoesters. Lastly, there is a good relationship between the relative ability of phthalate monoesters to trans-activate PPARalpha and PPARgamma, and the relative induction of PPARalpha target gene mRNA and PPARgamma-mediated adipocyte differentiation, respectively.


Assuntos
Poluentes Ambientais/toxicidade , Receptores Ativados por Proliferador de Peroxissomo/biossíntese , Ácidos Ftálicos/toxicidade , Células 3T3 , Animais , Carcinoma Hepatocelular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Camundongos , Receptores Ativados por Proliferador de Peroxissomo/classificação , Receptores Ativados por Proliferador de Peroxissomo/genética , RNA Mensageiro/metabolismo , Ratos , Especificidade da Espécie , Relação Estrutura-Atividade , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA