Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 5(2): 101421, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38340727

RESUMEN

Chimeric antigen receptor T cell (CAR T) therapy is a potent treatment for relapsed/refractory (r/r) B cell lymphomas but provides lasting remissions in only ∼40% of patients and is associated with serious adverse events. We identify an upregulation of CD80 and/or CD86 in tumor tissue of (r/r) diffuse large B cell lymphoma (DLBCL) patients treated with tisagenlecleucel. This finding leads to the development of the CAR/CCR (chimeric checkpoint receptor) design, which consists of a CD19-specific first-generation CAR co-expressed with a recombinant CTLA-4-linked receptor with a 4-1BB co-stimulatory domain. CAR/CCR T cells demonstrate superior efficacy in xenograft mouse models compared with CAR T cells, superior long-term activity, and superior selectivity in in vitro assays with non-malignant CD19+ cells. In addition, immunocompetent mice show an intact CD80-CD19+ B cell population after CAR/CCR T cell treatment. The results reveal the CAR/CCR design as a promising strategy for further translational study.


Asunto(s)
Linfoma de Células B Grandes Difuso , Linfocitos T , Humanos , Animales , Ratones , Antígeno CTLA-4 , Linfoma de Células B Grandes Difuso/terapia , Linfoma de Células B Grandes Difuso/etiología , Inmunoterapia Adoptiva/métodos , Linfocitos B , Antígenos CD19/genética
2.
Cell Mol Gastroenterol Hepatol ; 13(6): 1701-1716, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35219894

RESUMEN

BACKGROUND & AIMS: Liver fibrosis arises from long-term chronic liver injury, accompanied by an accelerated wound healing response with interstitial accumulation of extracellular matrix (ECM). Activated hepatic stellate cells (HSC) are the main source for ECM production. MicroRNA29a (miR-29a) is a crucial antifibrotic miRNA that is repressed during fibrosis, resulting in up-regulation of collagen synthesis. METHODS: Intracellular and extracellular miRNA levels of primary and immortalized myofibroblastic HSC in response to profibrogenic stimulation by transforming growth factor ß (TGFß) or platelet-derived growth factor-BB (PDGF-BB) or upon inhibition of vesicular transport and autophagy processes were determined by quantitative polymerase chain reaction. Autophagy flux was studied by electron microscopy, flow cytometry, immunoblotting, and immunocytochemistry. Hepatic and serum miR-29a levels were quantified by using both liver tissue and serum samples from a cohort of chronic hepatitis C virus patients and a murine CCl4 induced liver fibrosis model. RESULTS: In our study, we show that TGFß and PDGF-BB resulted in decrease of intracellular miR-29a and a pronounced increase of vesicular miR-29a release into the supernatant. Strikingly, miR-29a vesicular release was accompanied by enhanced autophagic activity and up-regulation of the autophagy marker protein LC3. Moreover, autophagy inhibition strongly prevented miR-29a secretion and repressed its targets' expression such as Col1A1. Consistently, hepatic miR-29a loss and increased LC3 expression in myofibroblastic HSC were associated with increased serum miR-29a levels in CCl4-treated murine liver fibrosis and specimens of hepatitis C virus patients with chronic liver disease. CONCLUSIONS: We provide evidence that activation-associated autophagy in HSC induces release of miR-29a, whereas inhibition of autophagy represses fibrogenic gene expression in part through attenuated miR-29a secretion.


Asunto(s)
Hepatitis C Crónica , MicroARNs/genética , Animales , Autofagia , Becaplermina/metabolismo , Células Estrelladas Hepáticas/patología , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/patología , Humanos , Cirrosis Hepática/patología , Ratones , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
Cell Mol Gastroenterol Hepatol ; 4(3): 339-363, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28884137

RESUMEN

BACKGROUND & AIMS: The human hepatitis B virus (HBV) is a major cause of chronic hepatitis and hepatocellular carcinoma, but molecular mechanisms driving liver disease and carcinogenesis are largely unknown. We therefore studied cellular pathways altered by HBV infection. METHODS: We performed gene expression profiling of primary human hepatocytes infected with HBV and proved the results in HBV-replicating cell lines and human liver tissue using real-time polymerase chain reaction and Western blotting. Activation of signal transducer and activator of transcription (STAT3) was examined in HBV-replicating human hepatocytes, HBV-replicating mice, and liver tissue from HBV-infected individuals using Western blotting, STAT3-luciferase reporter assay, and immunohistochemistry. The consequences of STAT3 activation on HBV infection and cell survival were studied by chemical inhibition of STAT3 phosphorylation and small interfering RNA-mediated knockdown of STAT3. RESULTS: Gene expression profiling of HBV-infected primary human hepatocytes detected no interferon response, while genes encoding for acute phase and antiapoptotic proteins were up-regulated. This gene regulation was confirmed in liver tissue samples of patients with chronic HBV infection and in HBV-related hepatocellular carcinoma. Pathway analysis revealed activation of STAT3 to be the major regulator. Interleukin-6-dependent and -independent activation of STAT3 was detected in HBV-replicating hepatocytes in cell culture and in vivo. Prevention of STAT3 activation by inhibition of Janus tyrosine kinases as well as small interfering RNA-mediated knockdown of STAT3-induced apoptosis and reduced HBV replication and gene expression. CONCLUSIONS: HBV activates STAT3 signaling in hepatocytes to foster its own replication but also to prevent apoptosis of infected cells. This very likely supports HBV-related carcinogenesis.

4.
Hepatology ; 66(1): 252-265, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28318036

RESUMEN

Use of adeno-associated viral (AAV) vectors for liver-directed gene therapy has shown considerable success, particularly in patients with severe hemophilia B. However, the high vector doses required to reach therapeutic levels of transgene expression caused liver inflammation in some patients that selectively destroyed transduced hepatocytes. We hypothesized that such detrimental immune responses can be avoided by enhancing the efficacy of AAV vectors in hepatocytes. Because autophagy is a key liver response to environmental stresses, we characterized the impact of hepatic autophagy on AAV infection. We found that AAV induced mammalian target of rapamycin (mTOR)-dependent autophagy in human hepatocytes. This cell response was critically required for efficient transduction because under conditions of impaired autophagy (pharmacological inhibition, small interfering RNA knockdown of autophagic proteins, or suppression by food intake), recombinant AAV-mediated transgene expression was markedly reduced, both in vitro and in vivo. Taking advantage of this dependence, we employed pharmacological inducers of autophagy to increase the level of autophagy. This resulted in greatly improved transduction efficiency of AAV vectors in human and mouse hepatocytes independent of the transgene, driving promoter, or AAV serotype and was subsequently confirmed in vivo. Specifically, short-term treatment with a single dose of torin 1 significantly increased vector-mediated hepatic expression of erythropoietin in C57BL/6 mice. Similarly, coadministration of rapamycin with AAV vectors resulted in markedly enhanced expression of human acid-α-glucosidase in nonhuman primates. CONCLUSION: We identified autophagy as a pivotal cell response determining the efficiency of AAVs intracellular processing in hepatocytes and thus the outcome of liver-directed gene therapy using AAV vectors and showed in a proof-of-principle study how this virus-host interaction can be employed to enhance efficacy of this vector system. (Hepatology 2017;66:252-265).


Asunto(s)
Autofagia/genética , Dependovirus/genética , Terapia Genética/métodos , Hepatocitos/citología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Transducción Genética
5.
Gastroenterology ; 150(1): 194-205, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26416327

RESUMEN

BACKGROUND & AIMS: Viral clearance involves immune cell cytolysis of infected cells. However, studies of hepatitis B virus (HBV) infection in chimpanzees have indicated that cytokines released by T cells also can promote viral clearance via noncytolytic processes. We investigated the noncytolytic mechanisms by which T cells eliminate HBV from infected hepatocytes. METHODS: We performed a cytokine enzyme-linked immunosorbent assay of serum samples from patients with acute and chronic hepatitis B. Liver biopsy specimens were analyzed by in situ hybridization. HepG2-H1.3 cells, HBV-infected HepaRG cells, and primary human hepatocytes were incubated with interferon-γ (IFNγ) or tumor necrosis factor-α (TNF-α), or co-cultured with T cells. We measured markers of HBV replication, including the covalently closed circular DNA (cccDNA). RESULTS: Levels of IFNγ and TNF-α were increased in serum samples from patients with acute vs chronic hepatitis B and controls. In human hepatocytes with stably replicating HBV, as well as in HBV-infected primary human hepatocytes or HepaRG cells, IFNγ and TNF-α each induced deamination of cccDNA and interfered with its stability; their effects were additive. HBV-specific T cells, through secretion of IFNγ and TNF-α, inhibited HBV replication and reduced cccDNA in infected cells without the direct contact required for cytolysis. Blocking IFNγ and TNF-α after T-cell stimulation prevented the loss of cccDNA. Deprivation of cccDNA required activation of nuclear APOBEC3 deaminases by the cytokines. In liver biopsy specimens from patients with acute hepatitis B, but not chronic hepatitis B or controls, hepatocytes expressed APOBEC3A and APOBEC3B. CONCLUSIONS: IFNγ and TNF-α, produced by T cells, reduce levels of HBV cccDNA in hepatocytes by inducing deamination and subsequent cccDNA decay.


Asunto(s)
Hepatitis B/metabolismo , Interferón gamma/farmacología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Células Cultivadas , Técnicas de Cocultivo , Replicación del ADN/efectos de los fármacos , ADN Viral/efectos de los fármacos , ADN Viral/inmunología , Ensayo de Inmunoadsorción Enzimática , Células Hep G2/inmunología , Células Hep G2/metabolismo , Hepacivirus/metabolismo , Hepatitis B/fisiopatología , Hepatitis B Crónica/inmunología , Humanos , Linfocitos T/inmunología , Carga Viral
6.
Hepatology ; 59(6): 2110-20, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24425003

RESUMEN

UNLABELLED: Gene therapy has become an accepted concept for the treatment of a variety of different diseases. In contrast to preclinical models, subjects enrolled in clinical trials, including gene therapy, possess a history of infection with microbes that may influence its safety and efficacy. Especially, viruses that establish chronic infections in the liver, one of the main targets for in vivo gene therapy, raise important concerns. Among them is the hepatitis B virus (HBV), which has chronically infected more than 350 million people worldwide. Here, we investigated the effect of HBV on adeno-associated viral (AAV) vectors, the most frequently applied gene transfer vehicles for in vivo gene therapy. Unexpectedly, we found that HBV greatly improved AAV transduction in cells replicating HBV and identified HBV protein x (HBx) as a key factor. Whereas HBV-positive and -negative cells were indistinguishable with respect to cell-entry efficiency, significantly higher numbers of AAV vector genomes were successfully delivered to the nucleus in the presence of HBV. The HBV-promoting effect was abolished by inhibitors of phosphatidylinositol 3-kinase (PI3K). PI3K was required for efficient trafficking of AAV to the nucleus and was enhanced in HBV-replicating cells and upon HBx expression. Enhancement of AAV transduction was confirmed in vivo using HBV transgenic mice and could successfully be applied to inhibit HBV progeny release. CONCLUSION: Our results demonstrate that acute, as well as chronic, infections with unrelated viruses change the intracellular milieu, thereby likely influencing gene therapy outcomes. In the case of HBV, HBx-mediated enhancement of AAV transduction is an advantage that could be exploited for development of novel treatments of HBV infection.


Asunto(s)
Dependovirus/genética , Predisposición Genética a la Enfermedad , Virus de la Hepatitis B/genética , Hepatitis B/virología , Animales , Activación Enzimática/genética , Femenino , Vectores Genéticos , Células HEK293 , Hepatitis B/genética , Hepatitis B/terapia , Humanos , Interferón gamma/genética , Masculino , Ratones , Ratones Transgénicos , ARN Interferente Pequeño/genética , Transactivadores/genética , Transducción Genética , Proteínas Reguladoras y Accesorias Virales , Replicación Viral/genética
7.
Hepatology ; 55(1): 287-97, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21898480

RESUMEN

UNLABELLED: Adeno-associated viral vectors (rAAV) are frequently used in gene therapy trials. Although rAAV vectors are of low immunogenicity, humoral as well as T cell responses may be induced. While the former limits vector reapplication, the expansion of cytotoxic T cells correlates with liver inflammation and loss of transduced hepatocytes. Because adaptive immune responses are a consequence of recognition by the innate immune system, we aimed to characterize cell autonomous immune responses elicited by rAAV in primary human hepatocytes and nonparenchymal liver cells. Surprisingly, Kupffer cells, but also liver sinusoidal endothelial cells, mounted responses to rAAV, whereas neither rAAV2 nor rAAV8 were recognized by hepatocytes. Viral capsids were sensed at the cell surface as pathogen-associated molecular patterns by Toll-like receptor 2. In contrast to the Toll-like receptor 9-mediated recognition observed in plasmacytoid dendritic cells, immune recognition of rAAV in primary human liver cells did not induce a type I interferon response, but up-regulated inflammatory cytokines through activation of nuclear factor κB. CONCLUSION: Using primary human liver cells, we identified a novel mechanism of rAAV recognition in the liver, demonstrating that alternative means of sensing rAAV particles have evolved. Minimizing this recognition will be key to improving rAAV-mediated gene transfer and reducing side effects in clinical trials due to immune responses against rAAV.


Asunto(s)
Dependovirus/inmunología , Terapia Genética/métodos , Vectores Genéticos/inmunología , Hepatocitos/inmunología , Inmunidad Innata/inmunología , Receptor Toll-Like 2/inmunología , Biopsia , Cápside/inmunología , Citocinas/inmunología , Dependovirus/genética , Células Endoteliales/citología , Células Endoteliales/inmunología , Células Endoteliales/virología , Células HEK293 , Hepatocitos/citología , Hepatocitos/virología , Humanos , Macrófagos del Hígado/citología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/virología , FN-kappa B/inmunología , FN-kappa B/metabolismo , Cultivo Primario de Células , Transducción de Señal/inmunología , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba/inmunología
8.
Virology ; 409(1): 77-83, 2011 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20974479

RESUMEN

The unique region of the VP1 capsid protein of adeno-associated viruses (AAV) in common with autonomously replicating parvoviruses comprises a secreted phospholipase A2 (sPLA2) homology domain. While the sPLA2 domain of Minute Virus of Mice has recently been shown to mediate endosomal escape by lipolytic pore formation, experimental evidence for a similar function in AAV infection is still lacking. Here, we explored the function of the sPLA2 domain of AAV by making use of the serotype 2 mutant (76)HD/AN. The sPLA2 defect in (76)HD/AN, which severely impairs AAV's infectivity, could be complemented in trans by co-infection with wild-type AAV2. Furthermore, co-infection with endosomolytically active, but not with inactive adenoviral variants partially rescued (76)HD/AN, providing the first evidence for a function of this domain in endosomal escape of incoming AAV particles.


Asunto(s)
Proteínas de la Cápside/metabolismo , Dependovirus/enzimología , Dependovirus/patogenicidad , Endosomas/virología , Fosfolipasas A2/metabolismo , Virión/metabolismo , Proteínas de la Cápside/genética , Dependovirus/clasificación , Dependovirus/genética , Endosomas/fisiología , Células HEK293 , Células HeLa , Humanos , Mutación , Fosfolipasas A2/química , Fosfolipasas A2/genética , Serotipificación
9.
J Virol ; 84(22): 11994-2001, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20719950

RESUMEN

Apoptosis of infected cells is critically involved in antiviral defense. Apoptosis, however, may also support the release and spread of viruses. Although the elimination of infected hepatocytes is required to combat hepatitis B virus (HBV) infection, it is still unknown which consequences hepatocyte apoptosis has for the virus and whether or not it is advantageous to the virus. To study this, we designed a cell culture model consisting of both HBV-producing cell lines and primary human hepatocytes serving as an infection model. We showed that the release of mature, enveloped virions was 80% to 90% reduced 24 h after the induction of apoptosis in HBV-replicating hepatoma cells or HBV-infected hepatocytes. Importantly, HBV particles released from apoptotic hepatocytes were immature and nonenveloped and proved not to be infectious. We found an inverse correlation between the strength of an apoptotic stimulus and the infectivity of the virus particles released: the more potent the apoptotic stimulus, the higher the ratio of nonenveloped capsids to virions and the lower their infectivity. Furthermore, we demonstrated that HBV replication and, particularly, the expression of the HBx protein transcribed from the viral genome during replication do not sensitize cells to apoptosis. Our data clearly reject the hypothesis that the apoptosis of infected hepatocytes facilitates the propagation of HBV. Rather, these data indicate that HBV needs to prevent the apoptosis of its host hepatocyte to ensure the release of infectious progeny and, thus, virus spread in the liver.


Asunto(s)
Apoptosis , Virus de la Hepatitis B/fisiología , Hepatitis B/fisiopatología , Hepatocitos/citología , Liberación del Virus , Línea Celular , Hepatitis B/virología , Virus de la Hepatitis B/genética , Hepatocitos/virología , Humanos , Replicación Viral
10.
J Biol Chem ; 285(34): 26223-32, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20538593

RESUMEN

Proteins of the nucleotide-binding domain, leucine-rich repeat (NLR)-containing family recently gained attention as important components of the innate immune system. Although over 20 of these proteins are present in humans, only a few members including the cytosolic pattern recognition receptors NOD1, NOD2, and NLRP3 have been analyzed extensively. These NLRs were shown to be pivotal for mounting innate immune response toward microbial invasion. Here we report on the characterization of human NLRC5 and provide evidence that this NLR has a function in innate immune responses. We found that NLRC5 is a cytosolic protein expressed predominantly in hematopoetic cells. NLRC5 mRNA and protein expression was inducible by the double-stranded RNA analog poly(I.C) and Sendai virus. Overexpression of NLRC5 failed to trigger inflammatory responses such as the NF-kappaB or interferon pathways in HEK293T cells. However, knockdown of endogenous NLRC5 reduced Sendai virus- and poly(I.C)-mediated type I interferon pathway-dependent responses in THP-1 cells and human primary dermal fibroblasts. Taken together, this defines a function for NLRC5 in anti-viral innate immune responses.


Asunto(s)
Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/inmunología , Virus/inmunología , Células Cultivadas , Células Madre Hematopoyéticas , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Poli I-C/inmunología , ARN Mensajero/análisis , Virus Sendai/inmunología , Activación Transcripcional/inmunología
11.
Mol Ther ; 18(7): 1302-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20424598

RESUMEN

In chronic renal disease, tubulointerstitial fibrosis is a leading cause of renal failure. Here, we made use of one of the most promising gene therapy vector platforms, the adeno-associated viral (AAV) vector system, and the COL4A3-deficient mice, a genetic mouse model of renal tubulointerstitial fibrosis, to develop a novel bidirectional treatment strategy to prevent renal fibrosis. By comparing different AAV serotypes in reporter studies, we identified AAV9 as the most suitable delivery vector to simultaneously target liver parenchyma for endocrine and renal tubular epithelium for paracrine therapeutic expression of the antifibrogenic cytokine human hepatocyte growth factor (hHGF). We used transcriptional targeting to drive hHGF expression from the newly developed CMV-enhancer-Ksp-cadherin-promoter (CMV-Ksp) in renal and hepatic tissue following tail vein injection of rAAV9-CMV-Ksp-hHGF into COL4A3-deficient mice. The therapeutic efficiency of our approach was demonstrated by a remarkable attenuation of tubulointerstitial fibrosis and repression of fibrotic markers such as collagen1alpha1 (Col1A1), platelet-derived growth factor receptor-beta (PDGFR-beta), and alpha-smooth muscle actin (SMA). Taken together, our results show the great potential of rAAV9 as an intravenously applicable vector for the combined paracrine and endocrine expression of antifibrogenic factors in the treatment of renal failure caused by tubulointerstitial fibrosis.


Asunto(s)
Dependovirus/genética , Fibrosis/terapia , Regulación de la Expresión Génica , Factor de Crecimiento de Hepatocito/metabolismo , Enfermedades Renales/terapia , Animales , Autoantígenos/genética , Colágeno Tipo IV/deficiencia , Colágeno Tipo IV/genética , Elementos de Facilitación Genéticos/genética , Elementos de Facilitación Genéticos/fisiología , Factor de Crecimiento de Hepatocito/genética , Humanos , Inmunohistoquímica , Riñón/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Transducción Genética
12.
Hepatology ; 50(6): 1773-82, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19937696

RESUMEN

UNLABELLED: With about 350 million virus carriers, hepatitis B virus (HBV) infection remains a major health problem. HBV is a noncytopathic virus causing persistent infection, but it is still unknown whether host recognition of HBV may activate an innate immune response. We describe that upon infection of primary human liver cells, HBV is recognized by nonparenchymal cells of the liver, mainly by liver macrophages (Kupffer cells), although they are not infected. Within 3 hours, this recognition leads to the activation of nuclear factor kappa B (NF-kappaB) and subsequently to the release of interleukin-6 (IL-6) and other proinflammatory cytokines (IL-8, TNF-alpha, IL-1beta), but does not induce an interferon response. The activation of proinflammatory cytokines, however, is transient, and even inhibits responsiveness toward a subsequent challenge. IL-6 released by Kupffer cells after activation of NF-kappaB controls HBV gene expression and replication in hepatocytes at the level of transcription shortly after infection. Upon binding to its receptor complex, IL-6 activates the mitogen-activated protein kinases exogenous signal-regulated kinase 1/2, and c-jun N-terminal kinase, which inhibit expression of hepatocyte nuclear factor (HNF) 1alpha and HNF 4alpha, two transcription factors essential for HBV gene expression and replication. CONCLUSION: Our results demonstrate recognition of HBV patterns by nonparenchymal liver cells, which results in IL-6-mediated control of HBV infection at the transcriptional level. Thus, IL-6 ensures early control of the virus, limiting activation of the adaptive immune response and preventing death of the HBV-infected hepatocyte. This pattern recognition may be essential for a virus, which infects a new host with only a few virions. Our data also indicate that therapeutic neutralization of IL-6 for treatment of certain diseases may represent a risk if the patient is HBV-infected.


Asunto(s)
Regulación de la Expresión Génica , Hepatitis B/inmunología , Interferones/fisiología , Interleucina-6/fisiología , Células Cultivadas , Citocinas/biosíntesis , Hepatitis B/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Factor Nuclear 1-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/antagonistas & inhibidores , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Transcripción Genética , Replicación Viral
13.
Cell Microbiol ; 10(7): 1478-90, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18346225

RESUMEN

Hepatitis B virus (HBV) is an important human pathogen, which targets the liver extremely efficient, gaining access to hepatocytes by a so far unknown receptor and replicating in a hepatocyte-specific fashion. Cell differentiation seems to determine HBV replication. We here show that the level of hepatocyte differentiation, as indicated by hepatocyte polarization and metabolic activity, is closely correlated to the transcription of the HBV RNA pregenome. Pregenome transcription determined the level of HBV replication in various cell lines of hepatocellular origin and in primary human hepatocytes. A variety of hepatocyte-enriched nuclear factors have been described to regulate transcription of the pregenome, but it remained unknown which factors link HBV replication to hepatocyte differentiation. We determined that high expression levels of HNF4alpha but not its potential cofactors or other hepatocyte-enriched transcription factors were essential for efficient HBV replication, and link it to hepatocyte differentiation. HNF1alpha contributed to the control of HBV replication because it regulated the expression of HNF4alpha. Thus, a concerted action of HNF4alpha and HNF1alpha, which also determines morphological and functional differentiation of hepatocytes, links HBV replication to hepatocyte differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Virus de la Hepatitis B/fisiología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/fisiología , Hepatocitos/virología , Replicación Viral/fisiología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/citología , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
14.
J Virol ; 82(8): 4159-63, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18256153

RESUMEN

Adenovirus type 12 (Ad12) propagation in hamster BHK21 cells is blocked prior to viral DNA replication. The amounts of Ad12 DNA in the nuclei or cytoplasm of hamster cells are about 2 orders of magnitude (2 h postinfection [p.i.]) and 4 to 5 orders of magnitude (48 h p.i.) lower than in permissive human cells. Cell line BHK21-hCAR is transgenic for and expresses the human coxsackie- and adenovirus receptor (hCAR) gene. Nuclear uptake of Ad12 DNA in BHK21-hCAR cells is markedly increased compared to that in naïve BHK21 cells. Ad12 elicits a cytopathic effect in BHK21-hCAR cells but not in BHK21 cells. Quantitative PCR or [(3)H]thymidine labeling followed by zone velocity sedimentation fails to detect Ad12 DNA replication in BHK21 or BHK21-hCAR cells. Newly assembled Ad12 virions cannot be detected. Thus, the block in Ad12 DNA replication in hamster cells is not released by enhanced nuclear import of Ad12 DNA.


Asunto(s)
Adenoviridae/fisiología , Núcleo Celular/virología , ADN Viral/metabolismo , Receptores Virales/biosíntesis , Internalización del Virus , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Núcleo Celular/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Cricetinae , Receptores Virales/genética , Replicación Viral/fisiología
15.
Gastroenterology ; 133(4): 1156-65, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17919491

RESUMEN

BACKGROUND & AIMS: Induction of heme oxygenase-1 (HO-1) has been shown to be beneficial in immune-mediated liver damage. We now investigate the effects of HO-1 induction in models of human hepatitis B virus (HBV) infection. METHODS: Adenoviral transfer of an HBV 1.3 genome into wild-type mice was used as a model for acute hepatitis B. HBV transgenic animals were used as a model for chronic HBV infection. HBV replication was assessed by HBV viremia, antigenemia, and Southern blotting, liver damage was assessed by serum alanine aminotransferase activities and histopathology of liver sections. To investigate HO-1 effects on HBV replication at a molecular level, stably HBV-transfected hepatoma cells were used. HBV gene expression, protein stability, transcription, and replication were determined. HO-1 was induced by either cobalt-protoporphyrin-IX or over expressed by adenoviral gene transfer. RESULTS: In the acute hepatitis B model, liver injury was reduced significantly after HO-1 induction. In addition, HO-1 showed a pronounced antiviral effect, which was confirmed in stably HBV-transfected hepatoma cells and in persistently HBV replicating transgenic mice. We showed that HO-1 induction repressed HBV replication directly in hepatocytes at a posttranscriptional step by reducing stability of HBV core protein and thus blocking refill of nuclear HBV covalently closed circular (ccc)DNA. Small interfering RNA directed against HO-1 proved that this effect depended on the expression level of HO-1. CONCLUSIONS: Besides its hepatoprotective effect, HO-1 showed a pronounced antiviral activity in HBV infection. Therefore, induction of HO-1 might be a novel therapeutic option for inflammatory flares of hepatitis B.


Asunto(s)
Antivirales/farmacología , Hemo-Oxigenasa 1/metabolismo , Virus de la Hepatitis B/genética , Hepatitis B/prevención & control , Hígado/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Protoporfirinas/farmacología , Adenoviridae/genética , Animales , Antivirales/uso terapéutico , Línea Celular Tumoral , ADN Viral/metabolismo , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Hemo-Oxigenasa 1/biosíntesis , Hemo-Oxigenasa 1/genética , Hepatitis B/enzimología , Hepatitis B/genética , Hepatitis B/patología , Humanos , Hígado/enzimología , Hígado/patología , Hígado/virología , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Protoporfirinas/uso terapéutico , Interferencia de ARN , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , Transfección , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Replicación Viral/efectos de los fármacos
16.
EMBO J ; 26(14): 3308-21, 2007 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-17599063

RESUMEN

The WD-repeat protein factor associated with nSMase activity (FAN) is a member of the family of TNF receptor adaptor proteins that are coupled to specific signaling cascades. However, the precise functional involvement of FAN in specific cellular TNF responses remain unclear. Here, we report the involvement of FAN in TNF-induced actin reorganization and filopodia formation mediated by activation of Cdc42. The pleckstrin-homology (PH) domain of FAN specifically binds to phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P), which targets FAN to the plasma membrane. Site-specific mutagenesis revealed that the ability of FAN to mediate filopodia formation was blunted either by the destruction of the PtdIns(4,5)P binding motif, or by the disruption of intramolecular interactions between the PH domain and the adjacent beige and Chediak-Higashi (BEACH) domain. Furthermore, FAN was shown to interact with the actin cytoskeleton in TNF-stimulated cells via direct filamentous actin (F-actin) binding. The results of this study suggest that PH-mediated plasma membrane targeting of FAN is critically involved in TNF-induced Cdc42 activation and cytoskeleton reorganization.


Asunto(s)
Actinas/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Factores de Necrosis Tumoral/farmacología , Células 3T3 , Animales , Células COS , Chlorocebus aethiops , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Ratones , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP cdc42/metabolismo
17.
Hepatology ; 43(3): 539-47, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16496321

RESUMEN

Dendritic cells (DC) of hepatitis B virus (HBV) carriers have been reported to exhibit functional impairment. Possible explanations for this phenomenon are infection of HBV by DC or alteration of DC function by HBV. We therefore analyzed whether DC support the different steps of HBV infection and replication: uptake, deposition of the HBV genome in the nucleus, antigen expression, and progeny virus release. When HBV genomes were artificially introduced into monocyte-derived DC by adenoviral vectors, low-level expression of hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) but no HBV replication was detected. When monocyte-derived DC were subjected to wild-type HBV or a recombinant HBV expressing Renilla luciferase under a non-liver-specific promoter, intracellular HBV DNA was detected in a low percentage of cells. However, neither nuclear cccDNA was formed nor luciferase activity was detected, indicating that either uncoating or nucleocytoplasmic transport were blocked. To verify our observation in the in vivo situation, myeloid and plasmacytoid DC were isolated from blood of high viremic HBV carriers, and analyzed by quantitative polymerase chain reaction (PCR) and electron microscopy. Although circulating DC had in vivo been exposed to more than 10(4) HBV virions per cell, HBV genomic DNA was hardly detected, and no nuclear cccDNA was detected at all. By using electron microscopy, subviral particles were found in endocytic vesicles, but virions were undetectable as were viral capsids in the cytoplasm. In conclusion, circulating DC may take up HBV antigens, but neither support nucleocytoplasmic transport nor replication of HBV.


Asunto(s)
Células Dendríticas/virología , Antígenos de la Hepatitis B/aislamiento & purificación , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Hepatitis B/inmunología , Adenoviridae , Portador Sano , Células Cultivadas , Células Dendríticas/inmunología , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos , Hepatitis B/genética , Humanos , Virión/genética , Virión/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
18.
J Virol ; 79(4): 2404-12, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15681441

RESUMEN

The infection of human cells by adenoviruses leads to a gradual reduction in the activity of host cell functions while viral gene expression progresses in a regulated way. We used the DNA microarray technique to determine the transcriptional activity profiles of cellular genes upon infection with adenovirus type 12 (Ad12). The microarray data were validated by quantitative real-time PCR for genes which showed significant alterations after Ad12 infection. At 12 h postinfection, there is a striking up-regulation between 10- and 30-fold in the expression of the G1P2, IFIT1, and IFIT2 cellular immune response genes compared to mock-infected cells. At later stages of infection, when the majority of regulated cellular genes has been turned down, a limited number of cellular genes exhibit increased activities by factors of 3 or less. These genes belong to the signal transduction or transcriptional regulator classes or are active in protein degradation, like ANPEP, an aminopeptidase. The SCD and CYP2S1 genes function in lipid metabolism. The eucaryotic translation initiation factor 4 is up-regulated, and one of the major histocompatibility complex genes is diminished in activity. For two of the genes, one up-regulated (CTSF gene) and one down-regulated (CYR61 gene), alterations in gene activity were confirmed at the protein level by Western blotting experiments. Increased genetic activity of cellular genes late in adenovirus infection has not been reported previously and demonstrates that Ad12 has a sustained control of host cell gene expression well into the late phase of infection.


Asunto(s)
Infecciones por Adenovirus Humanos/metabolismo , Adenovirus Humanos/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , ARN Mensajero/metabolismo , Infecciones por Adenovirus Humanos/genética , Adenovirus Humanos/genética , Células HeLa , Humanos , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
19.
Virus Res ; 98(1): 45-56, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14609629

RESUMEN

The intramuscular (i.m.) injection of human adenovirus type 12 (Ad12) into newborn Syrian hamsters caused widespread dissemination of up to 15 tumors over the entire peritoneal cavity in 70-90% of the animals within 30-50 days. Subcutaneous (s.c.) injections led to local tumor formation only. Independent of location, tumor histology revealed Homer-Wright rosette-like structures typical for primitive neuroectodermal tumors (PNET). All tumor cells showed markers indicative of neuroectodermal and mesenchymal derivations. Each Ad12-induced tumor cell carried multiple copies of integrated Ad12 genomes at one chromosomal site which was different for each tumor. For Ad12 tumor induction in hamsters, the patterns of Ad12 viral and cellular gene expression were important and were affected by changes in DNA methylation, both in the integrated Ad12 DNA and the cellular genome. By applying the bisulfite protocol, the de novo DNA methylation in the integrated Ad12 genomes was determined. These patterns were complex, characterized by regional initiation and by excluding genome segments in the E1A and E1B promoters. In all tumors, the Ad12 segments E1A, E1B, E2A, parts of E3 and E4 were similarly transcribed, as shown by the RT-PCR and DNA microarray methods. Changes in the transcription of a large number of cellular genes was assessed by using mouse gene microarrays encompassing about 1980 different mouse genes with 87-96% homology to hamster genes. Similarities and differences existed in the transcription of cellular genes of different functional classes among the different Ad12-induced tumors. These alterations in cellular gene transcription may be an important parameter in the oncogenic transformation by Ad12.


Asunto(s)
Adenovirus Humanos/patogenicidad , Neoplasias Experimentales/virología , Tumores Neuroectodérmicos/virología , Cavidad Peritoneal/patología , Proteínas/metabolismo , Proteínas Virales/metabolismo , Animales , Animales Recién Nacidos , Cricetinae , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inyecciones Intramusculares , Mesocricetus , Metilación , Ratones , Neoplasias Experimentales/patología , Tumores Neuroectodérmicos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , Análisis de Secuencia de ADN , Transcripción Genética , Proteínas Virales/genética , Integración Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...