Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 20: 258-275, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33473359

RESUMO

Chronic hepatitis B virus (HBV) infection is a major public health problem. New treatment approaches are needed because current treatments do not target covalently closed circular DNA (cccDNA), the template for HBV replication, and rarely clear the virus. We harnessed adeno-associated virus (AAV) vectors and CRISPR-Staphylococcus aureus (Sa)Cas9 to edit the HBV genome in liver-humanized FRG mice chronically infected with HBV and receiving entecavir. Gene editing was detected in livers of five of eight HBV-specific AAV-SaCas9-treated mice, but not control mice, and mice with detectable HBV gene editing showed higher levels of SaCas9 delivery to HBV+ human hepatocytes than those without gene editing. HBV-specific AAV-SaCas9 therapy significantly improved survival of human hepatocytes, showed a trend toward decreasing total liver HBV DNA and cccDNA, and was well tolerated. This work provides evidence for the feasibility and safety of in vivo gene editing for chronic HBV infections, and it suggests that with further optimization, this approach may offer a plausible way to treat or even cure chronic HBV infections.

2.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28250128

RESUMO

Syrian hamsters are permissive for the replication of species C human adenoviruses (HAdV-C). The virus replicates to high titers in the liver of these animals after intravenous infection, while respiratory infection results in virus replication in the lung. Here we show that two types belonging to species C, HAdV-C5 and HAdV-C6, replicate to significantly different extents and cause pathology with significantly different severities, with HAdV-C6 replicating better and inducing more severe and more widespread lesions. The virus burdens in the livers of HAdV-C6-infected hamsters are higher than the virus burdens in HAdV-C5-infected ones because more of the permissive hepatocytes get infected. Furthermore, when hamsters are infected intravenously with HAdV-C6, live, infectious virus can be isolated from the lung and the kidney, which is not seen with HAdV-C5. Similarly to mouse models, in hamsters, HAdV-C6 is sequestered by macrophages to a lesser degree than HAdV-C5. Depletion of Kupffer cells from the liver greatly increases the replication of HAdV-C5 in the liver, while it has only a modest effect on the replication of HAdV-C6. Elimination of Kupffer cells also dramatically increases the pathology induced by HAdV-C5. These findings indicate that in hamsters, pathology resulting from intravenous infection with adenoviruses is caused mostly by replication in hepatocytes and not by the abortive infection of Kupffer cells and the following cytokine storm.IMPORTANCE Immunocompromised human patients can develop severe, often lethal adenovirus infections. Respiratory adenovirus infection among military recruits is a serious problem, in some cases requiring hospitalization of the patient. Furthermore, adenovirus-based vectors are frequently used as experimental viral therapeutic agents. Thus, it is imperative that we investigate the pathogenesis of adenoviruses in a permissive animal model. Syrian hamsters are susceptible to infection with certain human adenoviruses, and the pathology accompanying these infections is similar to what is observed with adenovirus-infected human patients. We demonstrate that replication in permissive cells in a susceptible host animal is a major part of the mechanism by which systemic adenovirus infection induces pathology, as opposed to the chiefly immune-mediated pathology observed in nonsusceptible hosts. These findings support the use of compounds inhibiting adenovirus replication as a means to block adenovirus-induced pathology.


Assuntos
Infecções por Adenovirus Humanos/patologia , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/patogenicidade , Fígado/virologia , Carga Viral , Replicação Viral , Adenovírus Humanos/classificação , Adenovírus Humanos/fisiologia , Animais , Linhagem Celular , Cricetinae , Modelos Animais de Doenças , Humanos , Rim/virologia , Células de Kupffer/virologia , Fígado/patologia , Pulmão/virologia , Macrófagos/virologia , Mesocricetus
4.
PLoS Pathog ; 11(8): e1005084, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26291525

RESUMO

Human adenoviruses have been studied extensively in cell culture and have been a model for studies in molecular, cellular, and medical biology. However, much less is known about adenovirus replication and pathogenesis in vivo in a permissive host because of the lack of an adequate animal model. Presently, the most frequently used permissive immunocompetent animal model for human adenovirus infection is the Syrian hamster. Species C human adenoviruses replicate in these animals and cause pathology that is similar to that seen with humans. Here, we report findings with a new Syrian hamster strain in which the STAT2 gene was functionally knocked out by site-specific gene targeting. Adenovirus-infected STAT2 knockout hamsters demonstrated an accentuated pathology compared to the wild-type control animals, and the virus load in the organs of STAT2 knockout animals was 100- to 1000-fold higher than that in wild-type hamsters. Notably, the adaptive immune response to adenovirus is not adversely affected in STAT2 knockout hamsters, and surviving hamsters cleared the infection by 7 to 10 days post challenge. We show that the Type I interferon pathway is disrupted in these hamsters, revealing the critical role of interferon-stimulated genes in controlling adenovirus infection. This is the first study to report findings with a genetically modified Syrian hamster infected with a virus. Further, this is the first study to show that the Type I interferon pathway plays a role in inhibiting human adenovirus replication in a permissive animal model. Besides providing an insight into adenovirus infection in humans, our results are also interesting from the perspective of the animal model: STAT2 knockout Syrian hamster may also be an important animal model for studying other viral infections, including Ebola-, hanta-, and dengue viruses, where Type I interferon-mediated innate immunity prevents wild type hamsters from being effectively infected to be used as animal models.


Assuntos
Infecções por Adenoviridae/imunologia , Adenovírus Humanos/patogenicidade , Modelos Animais de Doenças , Interferon Tipo I/imunologia , Fator de Transcrição STAT2/deficiência , Infecções por Adenoviridae/patologia , Adenovírus Humanos/imunologia , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Cricetinae , Citometria de Fluxo , Técnicas de Inativação de Genes , Humanos , Mesocricetus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT2/imunologia
5.
Viruses ; 7(3): 1409-28, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25807051

RESUMO

Adenovirus infections of immunocompromised pediatric hematopoietic stem cell transplant patients can develop into serious and often deadly multi-organ disease. There are no drugs approved for adenovirus infections. Cidofovir (an analog of 2-deoxycytidine monophosphate) is used at times but it can be nephrotoxic and its efficacy has not been proven in clinical trials. Brincidofovir, a promising lipid-linked derivative of cidofovir, is in clinical trials. Ganciclovir, an analog of 2-deoxyguanosine, has been employed occasionally but with unknown efficacy in the clinic. In this study, we evaluated valganciclovir against disseminated adenovirus type 5 (Ad5) infection in our permissive immunosuppressed Syrian hamster model. We administered valganciclovir prophylactically, beginning 12 h pre-infection or therapeutically starting at Day 1, 2, 3, or 4 post-infection. Valganciclovir significantly increased survival, reduced viral replication in the liver, and mitigated the pathology associated with Ad5 infection. In cultured cells, valganciclovir inhibited Ad5 DNA replication and blocked the transition from early to late stage of infection. Valganciclovir directly inhibited Ad5 DNA polymerase in vitro, which may explain, at least in part, its mechanism of action. Ganciclovir and valganciclovir are approved to treat infections by certain herpesviruses. Our results support the use of valganciclovir to treat disseminated adenovirus infections in immunosuppressed patients.


Assuntos
Infecções por Adenovirus Humanos/tratamento farmacológico , Infecções por Adenovirus Humanos/patologia , Adenovírus Humanos/efeitos dos fármacos , Antivirais/uso terapêutico , Ganciclovir/análogos & derivados , Hospedeiro Imunocomprometido , Replicação Viral/efeitos dos fármacos , Adenovírus Humanos/fisiologia , Animais , Antivirais/farmacologia , Linhagem Celular , DNA Polimerase Dirigida por DNA/metabolismo , Modelos Animais de Doenças , Células Epiteliais/virologia , Feminino , Ganciclovir/farmacologia , Ganciclovir/uso terapêutico , Humanos , Fígado/virologia , Masculino , Mesocricetus , Análise de Sobrevida , Resultado do Tratamento , Valganciclovir , Carga Viral
6.
Cancer Res ; 73(7): 2117-26, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23378345

RESUMO

Chronic inflammation is a major risk factor for cancer, including gastric cancers and other gastrointestinal cancers. For example, chronic inflammation caused by autoimmune gastritis (AIG) is associated with an increased risk of gastric polyps, gastric carcinoid tumors, and possibly adenocarcinomas. In this study, we characterized the progression of gastric cancer in a novel mouse model of AIG. In this model, disease was caused by CD4(+) T cells expressing a transgenic T-cell receptor specific for a peptide from the H(+)/K(+) ATPase proton pump, a protein expressed by parietal cells in the stomach. AIG caused epithelial cell aberrations that mimicked most of those seen in progression of human gastric cancers, including chronic gastritis followed by oxyntic atrophy, mucous neck cell hyperplasia, spasmolytic polypeptide-expressing metaplasia, dysplasia, and ultimately gastric intraepithelial neoplasias. Our work provides the first direct evidence that AIG supports the development of gastric neoplasia and provides a useful model to study how inflammation drives gastric cancer.


Assuntos
Doenças Autoimunes/etiologia , Linfócitos T CD4-Positivos/imunologia , Gastrite/complicações , ATPase Trocadora de Hidrogênio-Potássio/imunologia , Inflamação/complicações , Receptores de Antígenos de Linfócitos T/fisiologia , Neoplasias Gástricas/etiologia , Adenocarcinoma/etiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Citometria de Fluxo , Imunofluorescência , Mucosa Gástrica/imunologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrite/imunologia , Gastrite/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/imunologia , Inflamação/patologia , Interferon gama/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Metaplasia/complicações , Metaplasia/imunologia , Metaplasia/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
7.
Genes Cancer ; 1(9): 964-71, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21779475

RESUMO

BCL-2/E1B-19 kDa-interacting protein 3 (BNIP3) is a BH3-only mitochondrial protein. Expression of BNIP3 is strongly stimulated by hypoxia. Up-regulation of BNIP3 has been detected in several human carcinomas including carcinomas of the lung and breast. The significance of BNIP3 overexpression in these cancers is not known. To determine whether BNIP3 plays a role in tumor growth, we generated A549 lung carcinoma cells that overexpressed BNIP3 and examined their ability to form tumors in the mouse xenograft model. All cell lines that overexpressed BNIP3 formed larger tumors compared to the parental or vector-transformed A549 cells. Breast carcinoma cell lines that overexpressed BNIP3 also induced tumors in athymic mice in the absence of hormone administration, while the parental cell line did not. Stable shRNA-mediated knockdown of endogenous BNIP3 severely impaired the tumorigenic activity of A549 cells. The tumor growth-enhancing activity was reduced by deletion of the BH3 domain of BNIP3. Expression of a dominant-negative mutant of BNIP3 lacking the C-terminal transmembrane domain also inhibited the tumorigenic potential of A549 cells. These results suggest that BNIP3 plays a fundamental role in the development of certain solid tumors such as the lung and breast carcinomas.

8.
Cancer Res ; 69(24): 9354-9, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19934312

RESUMO

Breast cancer is the leading cause of cancer death among women. We have shown previously an antiproliferative effect of MBP-1 on several human cancer cells. In this study, we have examined the potential of MBP-1 as a gene therapeutic candidate in regression of breast cancer growth and metastasis in an immunocompetent mouse model. For this, we have used a mouse breast cancer cell line (EO771) and syngeneic C57BL/6 mice. EO771 cells were implanted into the mammary fat pad of C57BL/6 mice. Replication-deficient recombinant adenovirus expressing MBP-1 was administered intratumorally to determine gene therapeutic potential. The results showed a significant regression of primary and distant (lung) tumor growth. Animals exhibited prolonged survival on treatment with MBP-1 compared with the control group (dl312). Subsequent studies suggested that MBP-1 inhibits matrix metalloproteinase expression in human breast cancer cells. Cells transduced with MBP-1 displayed inhibition of migration in a wound-healing assay. The conditioned medium from MBP-1-transduced cells blocked in vitro tube formation assay and inhibited expression of several angiogenic molecules. Taken together, our study shows that MBP-1 acts as a double-edged sword by inhibiting primary and metastatic tumor growth and modulating matrix metalloproteinase expression with a therapeutic potential against breast cancer progression.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Neoplasias Mamárias Experimentais/terapia , Adenoviridae/genética , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas de Ligação a DNA/genética , Células Endoteliais/patologia , Terapia Genética/métodos , Humanos , Imunocompetência , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
9.
Mol Ther ; 16(10): 1665-73, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18665155

RESUMO

We recently described an immunocompetent Syrian hamster model for oncolytic adenoviruses (Ads) that permits virus replication in tumor cells as well as some normal tissues. This model allows exploration of interactions between the virus, tumor, normal organs, and host immune system that could not be examined in the immunodeficient or nonpermissive animal models previously used in the oncolytic Ad field. Here we asked whether the immune response to oncolytic Ad enhances or limits antitumor efficacy. We first determined that cyclophosphamide (CP) is a potent immunosuppressive agent in the Syrian hamster and that CP alone had no effect on tumor growth. Importantly, we found that the antitumor efficacy of oncolytic Ads was significantly enhanced in immunosuppressed animals. In animals that received virus therapy plus immunosuppression, significant differences were observed in tumor histology, and in many cases little viable tumor remained. Notably, we also determined that immunosuppression allowed intratumoral virus levels to remain elevated for prolonged periods. Although favorable tumor responses can be achieved in immunocompetent animals, the rate of virus clearance from the tumor may lead to varied antitumor efficacy. Immunosuppression, therefore, allows sustained Ad replication and oncolysis, which leads to substantially improved suppression of tumor growth.


Assuntos
Adenoviridae/genética , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Replicação Viral/imunologia , Adenoviridae/imunologia , Adenoviridae/fisiologia , Animais , Antineoplásicos/uso terapêutico , Divisão Celular , Cricetinae , Ciclofosfamida/uso terapêutico , Imuno-Histoquímica , Mesocricetus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Testes de Neutralização
10.
Proc Natl Acad Sci U S A ; 105(20): 7293-7, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18490659

RESUMO

Adenoviruses (Ads) cause a wide array of end-organ and disseminated diseases in severely immunosuppressed patients. For example, approximately 20% of pediatric allogeneic hematopoietic stem cell transplant recipients develop disseminated Ad infection, and the disease proves fatal in as many as 50-80% of these patients. Ad infections are a serious problem for solid-organ transplant recipients and AIDS patients as well. Unfortunately, there are no antiviral drugs approved specifically to treat these infections. A suitable animal model that is permissive for Ad replication would help in the discovery process. Here we identify an animal model to study Ad pathogenesis and the efficacy of antiviral compounds. We show that human serotype 5 Ad (Ad5) causes severe systemic disease in immunosuppressed Syrian hamsters that is similar to that seen in immunocompromised patients. We also demonstrate that hexadecyloxypropyl-cidofovir (CMX001) rescues the hamsters from a lethal challenge with Ad5. The antiviral drug provided protection both prophylactically and when given up to 2 days after i.v. exposure to Ad5. CMX001 acts by reducing Ad replication in key target organs. Thus, the immunosuppressed Syrian hamster is a powerful model to evaluate anti-Ad drugs, and its use can facilitate the entry of drugs such as CMX001 into clinical trials.


Assuntos
Citosina/análogos & derivados , Imunossupressores/uso terapêutico , Organofosfonatos/farmacologia , Adenoviridae/metabolismo , Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/terapia , Animais , Antivirais/farmacologia , Cricetinae , Citosina/farmacologia , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Células-Tronco Hematopoéticas/citologia , Humanos , Fígado/metabolismo , Mesocricetus , Modelos Biológicos
11.
Comp Med ; 52(6): 568-71, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12540173

RESUMO

Epididymal cribriform hyperplasia (ECH) is a variant of normal epididymal histologic features in men, and has also been reported in rats, mice, dogs, cats, and bulls. The epididymal change has been associated with aging, testicular atrophy, cryptorchidism, and germ cell tumors. Epididymal cribriform hyperplasia was observed in p53 homozygous knockout mice on a mixed 129/Sv-FVB/N background, but not in wild-type or heterozygous mice. The aim of the study reported here was to determine the prevalence and characterize the morphologic, immunohistochemical, and ultrastructural features of ECH in these mice. Epididymal cribriform hyperplasia was present in 88% (72/82) of male mice ranging in age from seven to 65 weeks. The lesion was characterized microscopically by epithelial cells with atypical hyperchromatic nuclei, vacuolization, intratubular lumina formation, infrequent apoptosis, and rare mitotic figures. In contrast to germ cells, the cells of ECH did not express alpha-fetoprotein, carcinoembryonic antigen, or S-100. Ultrastructurally, the cells were pleomorphic with stereocilia at their apical borders and within intratubular lumina, and were supported by a basement membrane. Although 14% (10/72) of mice had concomitant testicular neoplasia, ECH did not appear to be a preneoplastic change. Investigators using these mice for modeling human disease should be aware of the background prevalence of this lesion.


Assuntos
Núcleo Celular/patologia , Epididimo/patologia , Doenças Testiculares/patologia , Proteína Supressora de Tumor p53/genética , Fatores Etários , Animais , Cruzamento , DNA/análise , Primers do DNA/química , Modelos Animais de Doenças , Epididimo/metabolismo , Células Epiteliais/ultraestrutura , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Genótipo , Homozigoto , Hiperplasia/patologia , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Doenças Testiculares/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA