Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hepatology ; 74(4): 1766-1781, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33991110

RESUMEN

BACKGROUND AND AIMS: Therapeutic strategies against HBV focus, among others, on the activation of the immune system to enable the infected host to eliminate HBV. Hypoxia-inducible factor 1 alpha (HIF1α) stabilization has been associated with impaired immune responses. HBV pathogenesis triggers chronic hepatitis-related scaring, leading inter alia to modulation of liver oxygenation and transient immune activation, both factors playing a role in HIF1α stabilization. APPROACH AND RESULTS: We addressed whether HIF1α interferes with immune-mediated induction of the cytidine deaminase, apolipoprotein B mRNA editing enzyme catalytic subunit 3B (APOBEC3B; A3B), and subsequent covalently closed circular DNA (cccDNA) decay. Liver biopsies of chronic HBV (CHB) patients were analyzed by immunohistochemistry and in situ hybridization. The effect of HIF1α induction/stabilization on differentiated HepaRG or mice ± HBV ± LTßR-agonist (BS1) was assessed in vitro and in vivo. Induction of A3B and subsequent effects were analyzed by RT-qPCR, immunoblotting, chromatin immunoprecipitation, immunocytochemistry, and mass spectrometry. Analyzing CHB highlighted that areas with high HIF1α levels and low A3B expression correlated with high HBcAg, potentially representing a reservoir for HBV survival in immune-active patients. In vitro, HIF1α stabilization strongly impaired A3B expression and anti-HBV effect. Interestingly, HIF1α knockdown was sufficient to rescue the inhibition of A3B up-regulation and -mediated antiviral effects, whereas HIF2α knockdown had no effect. HIF1α stabilization decreased the level of v-rel reticuloendotheliosis viral oncogene homolog B protein, but not its mRNA, which was confirmed in vivo. Noteworthy, this function of HIF1α was independent of its partner, aryl hydrocarbon receptor nuclear translocator. CONCLUSIONS: In conclusion, inhibiting HIF1α expression or stabilization represents an anti-HBV strategy in the context of immune-mediated A3B induction. High HIF1α, mediated by hypoxia or inflammation, offers a reservoir for HBV survival in vivo and should be considered as a restricting factor in the development of immune therapies.


Asunto(s)
Citidina Desaminasa/genética , Hepatitis B Crónica/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hígado/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Factor de Transcripción ReIB/genética , Aminoácidos Dicarboxílicos/farmacología , Animales , Línea Celular , Citidina Desaminasa/metabolismo , ADN Circular/metabolismo , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Virus de la Hepatitis B , Hepatitis B Crónica/metabolismo , Hepatitis B Crónica/virología , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Receptor beta de Linfotoxina/agonistas , Ratones , Viabilidad Microbiana , Antígenos de Histocompatibilidad Menor/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción ReIB/efectos de los fármacos , Factor de Transcripción ReIB/metabolismo
2.
Gastroenterology ; 156(4): 1190-1205.e14, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30445013

RESUMEN

BACKGROUND & AIMS: Cholangiocyte proliferation and ductular reaction contribute to the onset and progression of liver diseases. Little is known about the role of the transcription factor nuclear factor-κB (NF-κB) in this process. We investigated the activities of the RELB proto-oncogene NF-κB subunit in human cholangiocytes and in mouse models of liver disease characterized by a ductular reaction. METHODS: We obtained liver tissue samples from patients with primary sclerosing cholangitis, primary biliary cholangitis, hepatitis B or C virus infection, autoimmune hepatitis, alcoholic liver disease, or without these diseases (controls) from a tissue bank in Germany. Tissues were analyzed by immunohistochemistry for levels of RELB and lymphotoxin ß (LTB). We studied mice with liver parenchymal cell (LPC)-specific disruption of the cylindromatosis (CYLD) lysine 63 deubiquitinase gene (Cyld), with or without disruption of Relb (CyldΔLPC mice and Cyld/RelbΔLPC mice) and compared them with C57BL/6 mice (controls). Mice were fed 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or standard chow diets to induce biliary injury or were given injections of CCl4 to induce non-cholestatic liver fibrosis. Liver tissues were analyzed by histology, immunohistochemistry, immunoblots, in situ hybridization, and quantitative real-time polymerase chain reaction. Cholangiocytes were isolated from normal human liver, incubated with LTB receptor agonist, and transfected with small interfering RNAs to knock down RELB. RESULTS: In liver tissues from patients with primary sclerosing cholangitis, primary biliary cholangitis, chronic infection with hepatitis B or C virus, autoimmune hepatitis, or alcoholic liver disease, we detected increased nuclear translocation of RELB and increased levels of LTB in cholangiocytes that formed reactive bile ducts compared with control liver tissues. Human cholangiocytes, but not those with RELB knockdown, proliferated with exposure to LTB. The phenotype of CyldΔLPC mice, which included ductular reaction, oval cell activation, and biliary fibrosis, was completely lost from Cyld/RelbΔLPC mice. Compared with livers from control mice, livers from CyldΔLPC mice (but not Cyld/RelbΔLPC mice) had increased levels of mRNAs encoding cytokines (LTB; CD40; and tumor necrosis factor superfamily [TNFSF] members TNFSF11 [RANKL], TNFSF13B [BAFF], and TNFSF14 [LIGHT]) produced by reactive cholangiocytes. However, these strains of mice developed similar levels of liver fibrosis in response to CCl4 exposure. CyldΔLPC mice and Cyld/RelbΔLPC mice had improved liver function on the DDC diet compared with control mice fed the DDC diet. CONCLUSION: Reactive bile ducts in patients with chronic liver diseases have increased levels of LTB and nuclear translocation of RELB. RELB is required for the ductular reaction and development of biliary fibrosis in CyldΔLPC mice. Deletion of RELB and CYLD from LPCs protects mice from DDC-induced cholestatic liver fibrosis.


Asunto(s)
Conductos Biliares/metabolismo , Conductos Biliares/patología , Colangitis Esclerosante/metabolismo , Citocinas/genética , Hepatopatías/metabolismo , Factor de Transcripción ReIB/metabolismo , Adolescente , Adulto , Anciano , Animales , Tetracloruro de Carbono , Núcleo Celular , Proliferación Celular , Células Cultivadas , Cisteína Endopeptidasas/genética , Enzima Desubiquitinante CYLD , Dicarbetoxidihidrocolidina , Células Epiteliales/metabolismo , Femenino , Fibrosis , Técnicas de Silenciamiento del Gen , Humanos , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Receptor beta de Linfotoxina/agonistas , Linfotoxina beta/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Tejido Parenquimatoso/patología , Transporte de Proteínas , Proto-Oncogenes Mas , ARN Mensajero/metabolismo , Factor de Transcripción ReIB/genética , Adulto Joven
3.
Int Immunopharmacol ; 28(1): 220-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26096887

RESUMEN

Human lymphotoxin-ß receptor (LTßR), a member of the tumor necrosis factor receptor superfamily, plays an essential role in secondary lymphoid organ development, host defense, chemokine secretion, and apoptosis. In our study, LTßR activations by different stimulations were all found to induce RANTES secretion. Overexpression of LTßR or stimulation LTßR by ligands or agonistic antibody in human lung epithelial cells induced RANTES secretion However, the regulatory mechanism and the signaling cascade have not been fully elucidated. Therefore, the aim of this study was to elucidate the mechanism underlying LTßR-mediated RANTES production. Our study indicated that activation of JNK and ERK was important for the regulation of RANTES secretion. In addition, dominant negative mutants of ASK1, TAK1, and MEKK1 inhibited LTßR-induced RANTES expression. The dominant negative mutants of TRAF2, 3, and 5 also inhibited LTßR-mediated RANTES secretion. Chromatin immunoprecipitation analysis showed that LTßR activation induced the binding of c-Jun and NF-κB to the RANTES promoter. The results of this study show that LTßR activates ASK1, TAK1, and MEKK1 cascades via TRAF2, 3, and 5, resulting in the activation of JNK and ERK, which promotes the binding of c-Jun and NF-κB to the RANTES promoter, thereby increasing RANTES expression and secretion.


Asunto(s)
Quimiocina CCL5/biosíntesis , Receptor beta de Linfotoxina/agonistas , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Quinasas Janus/efectos de los fármacos , Receptor beta de Linfotoxina/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Regiones Promotoras Genéticas/genética , Transducción de Señal/efectos de los fármacos , Factor 2 Asociado a Receptor de TNF/genética , Regulación hacia Arriba
5.
Science ; 344(6189): 1237, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24926010

RESUMEN

Lucifora et al. (Research Articles, 14 March 2014, p. 1221) report that the hepatitis B virus (HBV) transcriptional template, a long-lived covalently closed circular DNA (cccDNA) molecule, is degraded noncytolytically by agents that up-regulate APOBEC3A and 3B. If these results can be independently confirmed, they would represent a critical first step toward development of a cure for the 400 million patients who are chronically infected by HBV.


Asunto(s)
Antivirales/farmacología , ADN Circular/metabolismo , ADN Viral/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Hepatocitos/efectos de los fármacos , Interferón-alfa/farmacología , Receptor beta de Linfotoxina/agonistas , Animales , Humanos
6.
Science ; 344(6189): 1237, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24926011

RESUMEN

Chisari et al. challenge our central conclusion that the hepatitis B virus (HBV) persistent form, the covalently closed circular DNA (cccDNA), is degraded in a noncytotoxic and specific fashion in the nucleus of infected hepatocytes. Specificity of the assays used, exclusion of cell division or death, and activity of APOBEC3 deaminases in the nucleus, however, were addressed in the paper.


Asunto(s)
Antivirales/farmacología , ADN Circular/metabolismo , ADN Viral/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Hepatocitos/efectos de los fármacos , Interferón-alfa/farmacología , Receptor beta de Linfotoxina/agonistas , Animales , Humanos
8.
Science ; 343(6176): 1221-8, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24557838

RESUMEN

Current antiviral agents can control but not eliminate hepatitis B virus (HBV), because HBV establishes a stable nuclear covalently closed circular DNA (cccDNA). Interferon-α treatment can clear HBV but is limited by systemic side effects. We describe how interferon-α can induce specific degradation of the nuclear viral DNA without hepatotoxicity and propose lymphotoxin-ß receptor activation as a therapeutic alternative. Interferon-α and lymphotoxin-ß receptor activation up-regulated APOBEC3A and APOBEC3B cytidine deaminases, respectively, in HBV-infected cells, primary hepatocytes, and human liver needle biopsies. HBV core protein mediated the interaction with nuclear cccDNA, resulting in cytidine deamination, apurinic/apyrimidinic site formation, and finally cccDNA degradation that prevented HBV reactivation. Genomic DNA was not affected. Thus, inducing nuclear deaminases-for example, by lymphotoxin-ß receptor activation-allows the development of new therapeutics that, in combination with existing antivirals, may cure hepatitis B.


Asunto(s)
Antivirales/farmacología , ADN Circular/metabolismo , ADN Viral/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Hepatocitos/efectos de los fármacos , Interferón-alfa/farmacología , Receptor beta de Linfotoxina/agonistas , Animales , Anticuerpos Monoclonales , Antivirales/uso terapéutico , Línea Celular , Núcleo Celular/virología , Citidina/metabolismo , Citidina Desaminasa/biosíntesis , Virus de la Hepatitis B/metabolismo , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Interferón-alfa/uso terapéutico , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/virología , Receptor beta de Linfotoxina/antagonistas & inhibidores , Ratones SCID , Antígenos de Histocompatibilidad Menor , Proteínas , Regulación hacia Arriba
9.
Mol Immunol ; 54(1): 40-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23220069

RESUMEN

Our earlier studies indicated that activation of the lymphotoxin beta receptor (LTßR) by T cell derived LTα(1)ß(2) regulates inflammatory cytokine expression. While characterizing the cellular and molecular mechanisms responsible for the down regulation of the inflammatory reaction after LTßR stimulation we were able to identify the specific induction of TRIM30α expression as a result of LTßR signalling in mouse macrophages. Furthermore, we could demonstrate that LTßR activation in these cells results in the down regulation of pro-inflammatory cytokine (e.g. TNF and IL-6) and mediator expression upon TLR4 and TLR9 re-stimulation, demonstrating that LTßR activation on mouse macrophages dampens pro-inflammatory cytokine and mediator expression. Thus, LTßR signalling renders macrophages hypo-responsive to subsequent stimulation with TLR ligands. The observation of an LTßR-mediated TLR-tolerance in the human monocyte cell line THP-1 suggests that similar signalling mechanisms seem to exist in human cells. Signalling pathway analysis clearly demonstrated that LTßR-induced TRIM30α expression is mediated by an IκBα-dependent signalling pathway. Furthermore, the LTßR-induced TRIM30α expression seems to be TRAF3 dependent. Our data suggest that LTßR activation on mouse macrophages is involved in the control of pro-inflammatory cytokine and mediator expression by activation of a signalling pathway that controls exacerbating inflammatory cytokine production.


Asunto(s)
Citocinas/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Receptor beta de Linfotoxina/fisiología , Factor 3 Asociado a Receptor de TNF/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptor beta de Linfotoxina/agonistas , Receptor beta de Linfotoxina/antagonistas & inhibidores , Receptor beta de Linfotoxina/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/fisiología , Ratones , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo
10.
Cytokine ; 47(1): 69-76, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19501526

RESUMEN

M cells assist mucosal immune surveillance by transcytosis of particles to underlying lymphoid tissue, but the mechanisms of M cell differentiation are poorly understood. To develop a better defined cell culture model of M cell differentiation, we treated human (Caco-2BBe) and rat (IEC-6) intestinal epithelial cell lines with lymphotoxin beta receptor (LTbetaR) and TNF receptor (TNFR) agonists. Treated cells were studied for regulation of genes associated with M cell and follicle-associated epithelium (FAE). We found that LTbetaR and TNFR agonists induce transcription of FAE-specific genes (Ccl20 and Lamb3) in Caco2-BBe cells and IEC-6 cells as well as rodent M cell specific genes such as Sgne-1/Scg5, Cldn4, and Gp2. The cytokines have distinct but complementary effects; TNFR agonists mainly induced FAE-specific genes, while the LTbetaR agonist induced M cell specific genes. The combination of cytokines showed additive induction of the FAE-associated Ccl20, Lamb3 and a surprising induction of CD137/Tnfrsf9. On the other hand TNF agonists appeared to suppress expression of some LTbetaR-induced genes. Functionally, cytokine treatment led to the reorganization of microvilli and Claudin-4 redistribution. These studies suggest complex interactions between these cytokines in the context of either inflammation or tissue differentiation.


Asunto(s)
Células Epiteliales/metabolismo , Mucosa Intestinal/citología , Receptor beta de Linfotoxina/agonistas , Receptores del Factor de Necrosis Tumoral/agonistas , Regulación hacia Arriba/efectos de los fármacos , Animales , Anexina A5/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Células CACO-2 , Moléculas de Adhesión Celular/genética , Línea Celular , Línea Celular Tumoral , Quimiocina CCL20/genética , Claudina-4 , Citocinas/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Proteínas Ligadas a GPI , Humanos , Receptor beta de Linfotoxina/inmunología , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microvellosidades/efectos de los fármacos , Proteína 7B2 Secretora Neuroendocrina/genética , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Transporte de Proteínas/efectos de los fármacos , Ratas , Staphylococcus aureus/inmunología , Factor de Transcripción ReIB/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/farmacología , Kalinina
11.
Biochim Biophys Acta ; 1789(5): 386-94, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19303953

RESUMEN

Canonical and alternative NF-kappaB pathways depend on distinct NF-kappaB members and regulate expression of different gene subset in inflammatory and steady state conditions, respectively. In intestinal epithelial cells, both pathways control the transcription of the gene coding the CCL20 chemokine. Lymphotoxin beta receptor (LTbetaR) mediates long lasting CCL20 expression whereas Toll-like receptor 5 (TLR5) signals promote inducible and transient activation. Here, we investigated whether the regulation of ccl20 expression involves different promoter sites and NF-kappaB molecules in response to TLR5 and LTbetaR stimulation. In epithelial cells, both stimulation required the same promoter regions, especially the NF-kappaB binding site but involved different NF-kappaB isoforms: p65/p50 and p52/RelB, for TLR5 and LTbetaR-dependent activation, respectively. The dynamic of activation and interaction with CCL20-specific NF-kappaB site correlated with gene transcription. Similar Ccl20 expression and NF-kappaB activation was found in the small intestine of mice stimulated with TLR5 and LTbetaR agonists. In summary, different NF-kappaB pathways modulate CCL20 transcription by operating on the same NF-kappaB binding site in the same cell type.


Asunto(s)
Quimiocina CCL20/genética , Receptor beta de Linfotoxina/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 5/metabolismo , Transporte Activo de Núcleo Celular , Animales , Secuencia de Bases , Sitios de Unión , Células CACO-2 , Línea Celular , Núcleo Celular/metabolismo , Quimiocina CCL20/biosíntesis , Quimiocina CCL20/metabolismo , Flagelina/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Receptor beta de Linfotoxina/agonistas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal
12.
J Biol Chem ; 282(22): 16105-16, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17430892

RESUMEN

Cells lacking functional NF-kappaB die after ligation of some tumor necrosis factor (TNF) receptor family members through failure to express NF-kappaB-dependent anti-apoptotic genes. NF-kappaB activation requires the IkappaB kinase (IKK) complex containing two catalytic subunits named IKKalpha and IKKbeta that regulate distinct NF-kappaB pathways. IKKbeta is critical for classical signaling that induces pro-inflammatory and anti-apoptotic gene profiles, whereas IKKalpha regulates the non-canonical pathway involved in lymphoid organogenesis and B-cell development. To determine whether IKKalpha and IKKbeta differentially function in rescuing cells from death induced by activators of the classical and non-canonical pathways, we analyzed death after ligation of the TNF and lymphotoxin-beta receptors, respectively. Using murine embryonic fibroblasts (MEFs) lacking each of the IKKs, the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, and dominant negative Fas-associated death domain protein, we found that deletion of these kinases sensitized MEFs to distinct cell death pathways. MEFs lacking IKKalpha were sensitized to death in response to both cytokines that was entirely caspase-dependent, demonstrating that IKKalpha functions in this process. Surprisingly, death of IKKbeta-/- MEFs was not blocked by caspase inhibition, demonstrating that IKKbeta negatively regulates caspase-independent cell death (CICD). CICD was strongly activated by both TNF and lymphotoxin-beta receptor ligation in IKKbeta-/- MEFs and was accompanied by loss of mitochondrial membrane potential and the generation of reactive oxygen species. CICD was inhibited by the anti-oxidant butylated hydroxyanosole and overexpression of Bcl-2, neither of which blocked caspase-dependent apoptosis. Our findings, therefore, demonstrate that both IKKalpha and IKKbeta regulate cytokine-induced apoptosis, and IKKbeta additionally represses reactive oxygen species- and mitochondrial-dependent CICD.


Asunto(s)
Apoptosis/genética , Caspasas/metabolismo , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Inhibidores de Caspasas , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/metabolismo , Receptor beta de Linfotoxina/agonistas , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Organogénesis/efectos de los fármacos , Organogénesis/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Agregación de Receptores/efectos de los fármacos , Agregación de Receptores/genética , Factor de Necrosis Tumoral alfa/farmacología , Quinasa de Factor Nuclear kappa B
13.
Cancer Res ; 66(19): 9617-24, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018619

RESUMEN

The lymphotoxin-beta receptor (LT beta R) is a tumor necrosis factor receptor family member critical for the development and maintenance of various lymphoid microenvironments. Herein, we show that agonistic anti-LT beta R monoclonal antibody (mAb) CBE11 inhibited tumor growth in xenograft models and potentiated tumor responses to chemotherapeutic agents. In a syngeneic colon carcinoma tumor model, treatment of the tumor-bearing mice with an agonistic antibody against murine LT beta R caused increased lymphocyte infiltration and necrosis of the tumor. A pattern of differential gene expression predictive of cellular and xenograft response to LT beta R activation was identified in a panel of colon carcinoma cell lines and when applied to a panel of clinical colorectal tumor samples indicated 35% likelihood a tumor response to CBE11. Consistent with this estimate, CBE11 decreased tumor size and/or improved long-term animal survival with two of six independent orthotopic xenografts prepared from surgical colorectal carcinoma samples. Targeting of LT beta R with agonistic mAbs offers a novel approach to the treatment of colorectal and potentially other types of cancers.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/uso terapéutico , Neoplasias del Colon/terapia , Receptor beta de Linfotoxina/agonistas , Neoplasias del Cuello Uterino/terapia , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Terapia Combinada , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/inmunología , Inmunoglobulina M/uso terapéutico , Irinotecán , Linfocitos Infiltrantes de Tumor/inmunología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Distribución Aleatoria , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Método Simple Ciego , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA