Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G25-G31, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30160980

RESUMEN

There is a need for noninvasive biomarkers that can identify patients with progressive liver fibrosis and monitor response to antifibrotic therapy. An equally important need is identification of patients with spontaneous fibrosis regression, since they may not need treatment nor be included in clinical studies with fibrosis as end point. Circulating biomarkers, originating from defined fragments of the scar tissue itself, may serve as valuable tools for this aspect of precision medicine. We investigated a panel of serological collagen formation and degradation markers to identify patients likely to regress or progress in absence of a therapeutic intervention. Plasma samples from patients with moderate-stage hepatitis C receiving placebo treatment in a phase II trial of the peroxisome proliferator-activated receptor agonist farglitazar were included. The patients had matched liver biopsies at baseline and 52 wk of follow-up. Serological biomarkers of collagen formation (PRO-C3, PRO-C4, PRO-C5) and collagen degradation (C3M, C4M, and C6M) were analyzed. Logistic regression analysis including PRO-C3 and C6M identified subjects with progressive liver fibrosis with an AUROC of 0.91 ( P < 0.0001) and positive and negative predictive values (PPV/NPV) of 75.0%/88.6%. Low levels of PRO-C5 predicted a spontaneous regression phenotype, with an odds ratio of 33.8 times higher compared with patients with high levels ( P < 0.0025) with an AUROC of 0.78 ( P < 0.0001) and PPV/NPV of 60.0%/95.7%. Two collagen fragments (PRO-C3 and C6M) identified liver fibrosis progressors, and one collagen fragment (PRO-C5) identified liver fibrosis regressors. These biomarkers may improve patient stratification and monitor treatment efficacy in studies with fibrosis as clinical end point. NEW & NOTEWORTHY In this study we report two biomarkers of collagen fragments (PRO-C3 and C6M) that are able to identify liver fibrosis progressors while one biomarker (PRO-C5) identified liver fibrosis regressors. In particular, we present three noninvasive biomarkers that can be used to identify patients with progressive liver fibrosis, monitor response to antifibrotic therapy, and also identify the spontaneous liver fibrosis regression phenotype.


Asunto(s)
Colágeno/metabolismo , Fibrosis/metabolismo , Cirrosis Hepática/metabolismo , Adulto , Biomarcadores/metabolismo , Biopsia , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
2.
Gastroenterology ; 147(6): 1378-92, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25173753

RESUMEN

BACKGROUND & AIMS: Platelet-derived growth factor-ß (PDGFB) is a mitogen for hepatic stellate cells (HSCs). We studied the cellular sources of PDGFB and the effects of a high-affinity monoclonal antibody against PDGFB (MOR8457) in mouse models of biliary fibrosis. METHODS: Cellular sources of PDGFB were identified using quantitative reverse-transcription polymerase chain reaction, biochemical, and immunohistologic methods. Mice with advanced biliary fibrosis, MDR2(Abcb4)-null mice, and C57Bl/6 (control) mice were placed on 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-supplemented diets and were given weekly intraperitoneal injections of MOR8457. Platelets were depleted from MDR2-null mice by injection of an antibody against CD41, or inhibited with diets containing low-dose aspirin. Liver tissues were collected and analyzed by quantitative reverse-transcription PCR and histologic and biochemical analyses. RESULTS: Levels of PDGFB protein, but not messenger RNA, were increased in fibrotic livers of MDR2-null mice, compared with control mice. Platelet clusters were detected in the hepatic endothelium, in close proximity to HSCs, and were identified as a source of PDGFB protein in MDR2-null mice. Levels of the PDGFB were increased in serum samples from patients with early stages of liver fibrosis of various etiologies (F1-2, n = 16; P < .05), compared with nonfibrotic liver tissue (F0, n = 12). Depletion of platelets from MDR2-null mice normalized hepatic levels of PDGFB within 48 hours, reducing levels of a marker of HSC activation (α-smooth muscle actin) and expression of genes that promote fibrosis. Diets supplemented with low-dose aspirin reduced circulating serum and hepatic levels of PDGFB and significantly reduced progression of fibrosis in MDR2-null mice over 1 year. MOR8457 produced a dose-dependent decrease in liver fibrosis in MDR2-null mice, reducing collagen deposition by 45% and expression of fibrosis-associated genes by 50%, compared with mice given a control antibody. In vitro, platelets activated freshly isolated HSCs (induction of α-smooth muscle actin and fibrosis-associated genes) via a PDGFB-dependent mechanism. MOR8457 also reduced liver fibrosis in mice placed on DDC-supplemented diets. CONCLUSIONS: Platelets produce PDGFB to activate HSC and promote fibrosis in MDR2-null mice and mice on DDC-supplemented diets. Antiplatelet therapy or selective inhibition of PDGFB might reduce biliary fibrosis in patients with liver disease.


Asunto(s)
Conductos Biliares Extrahepáticos/metabolismo , Plaquetas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Aspirina/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/farmacología , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/inmunología , ARN Mensajero/metabolismo , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
3.
Immunotherapy ; 15(11): 853-865, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37291871

RESUMEN

Cancer immunotherapy is a field that garners significant interest, fueled by the clinical success of immune checkpoint inhibitors. In contrast to conventional cancer therapies, immunotherapies leverage the host's immune system by enhancing innate and adaptive immunity to control cancer progression. Despite these exciting advances, only a subset of patients respond to these drugs, and immunotherapies frequently result in immune-related toxicity. One approach to overcome these challenges is intratumoral administration of treatment to minimize systemic toxicities and maximize therapeutic effects. Intratumoral cancer therapies have shown similar or superior antitumor efficacy in both treated and distant untreated tumors, with a widely improved benefit-risk ratio over conventional therapeutic approaches. Herein, we review the current landscape of intratumoral cancer gene immunotherapy.


Immunotherapies are drugs designed to activate a patient's own immune system to fight cancer. Research in this field has soared following the US FDA's approval of the first class of these drugs. They work by blocking cancer cells' ability to hide from the body's immune system. Unfortunately, only some patients respond and many experience side effects when the medicine is delivered to the whole body. One approach to overcome these problems is to deliver these drugs directly into a patient's tumor to limit side effects while maintaining the positive effects. In this review we describe the benefits of giving these types of drugs directly into tumors over whole-body administration. We summarize the current clinical data and explain the mechanisms behind each drug.


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Inmunoterapia , Terapia Combinada , Terapia Genética
4.
Lancet Infect Dis ; 18(8): 874-883, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29909069

RESUMEN

BACKGROUND: DSM265 is a novel, long-duration inhibitor of plasmodium dihydroorotate dehydrogenase (DHODH) with excellent selectivity over human DHODH and activity against blood and liver stages of Plasmodium falciparum. This study aimed to assess the efficacy of DSM265 in patients with P falciparum or Plasmodium vivax malaria infection. METHODS: This proof-of-concept, open-label, phase 2a study was conducted at the Asociación Civil Selva Amazónica in Iquitos, Peru. Patients aged 18-70 years, weighing 45-90 kg, who had clinical malaria (P falciparum or P vivax monoinfection) and fever within the previous 24 h were eligible. Exclusion criteria were clinical or laboratory signs of severe malaria, inability to take oral medicine, and use of other antimalarial treatment in the preceding 14 days. Patients were divided into cohorts of those with P falciparum (cohort a) or P vivax (cohort b) infection. Two initial cohorts received single oral doses of 400 mg DSM265. Patients were followed up for efficacy for 28 days and safety for 35 days. Further cohorts received escalated or de-escalated doses of DSM265, after safety and efficacy assessment of the initial dose. The primary endpoints were the proportion of patients achieving PCR-adjusted adequate clinical and parasitological response (ACPR) by day 14 for patients infected with P falciparum and the proportion of patients achieving a crude cure by day 14 for those infected with P vivax. Cohort success, the criteria for dose escalation, was defined as ACPR (P falciparum) or crude cure (P vivax) in at least 80% of patients in the cohort. The primary analysis was done in the intention-to-treat population (ITT) and the per-protocol population, and safety analyses were done in all patients who received the study drug. This study is registered at ClinicalTrials.gov (NCT02123290). FINDINGS: Between Jan 12, 2015, and Dec 2, 2015, 45 Peruvian patients (24 with P falciparum [cohort a] and 21 with P vivax [cohort b] infection) were sequentially enrolled. For patients with P falciparum malaria in the per-protocol population, all 11 (100%) in the 400 mg group and eight (80%) of ten in the 250 mg group achieved ACPR on day 14. In the ITT analysis, 11 (85%) of 13 in the 400 mg group and eight (73%) of 11 in the 250 mg group achieved ACPR at day 14. For the patients with P vivax malaria, the primary endpoint was not met. In the per-protocol analysis, none of four patients who had 400 mg, three (50%) of six who had 600 mg, and one (25%) of four who had 800 mg DSM265 achieved crude cure at day 14. In the ITT analysis, none of five in the 400 mg group, three (33%) of nine in the 600 mg group, and one (14%) of seven in the 800 mg group achieved crude cure at day 14. During the 28-day extended observation of P falciparum patients, a resistance-associated mutation in the gene encoding the DSM265 target DHODH was observed in two of four recurring patients. DSM265 was well tolerated. The most common adverse events were pyrexia (20 [44%] of 45) and headache (18 [40%] of 45), which are both common symptoms of malaria, and no patients had any treatment-related serious adverse events or adverse events leading to study discontinuation. INTERPRETATION: After a single dose of DSM265, P falciparum parasitaemia was rapidly cleared, whereas against P vivax, DSM265 showed less effective clearance kinetics. Its long duration of action provides the potential to prevent recurrence of P falciparum after treatment with a single dose, which should be further assessed in future combination studies. FUNDING: The Global Health Innovative Technology Fund, the Bill & Melinda Gates Foundation, the National Institutes of Health (R01 AI103058), the Wellcome Trust, and the UK Department of International Development.


Asunto(s)
Antimaláricos/administración & dosificación , Malaria Falciparum/tratamiento farmacológico , Malaria Vivax/tratamiento farmacológico , Plasmodium falciparum/inmunología , Pirimidinas/administración & dosificación , Triazoles/administración & dosificación , Adulto , Estudios de Cohortes , Dihidroorotato Deshidrogenasa , Femenino , Humanos , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Malaria Vivax/inmunología , Malaria Vivax/parasitología , Masculino , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Perú
5.
Front Biosci ; 11: 1448-65, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16368528

RESUMEN

Mammals and other higher vertebrates have developed an adaptive immune system to defy effectively countless pathogens and cancerous cells encountered during the lifetime of an individual. B and T lymphocytes, which are essential in orchestrating adaptive immune responses, express surface receptors specific for foreign and abnormal self-antigens. Genesis of this antigen receptor repertoire poses significant risks for autoimmunity caused by self-reactive lymphocytes. Therefore, organisms with adaptive immune systems have evolved central and peripheral tolerance mechanisms. In peripheral tissues, regulatory T (Treg) cells function in a dominant, cell-extrinsic manner to limit inflammatory responses and autoimmune disorders. To tap the potential clinical utility of these specialized lymphocytes, advances have been made in understanding how Treg cell-mediated suppression of immune effector cells is achieved and regulated. Importantly, signaling induced by a recently identified member of the tumor necrosis factor receptor (TNFR) family, termed glucocorticoid-induced TNFR family-related gene (GITR), abrogates the suppressive effects of Treg cells. GITR plays a pivotal role in controlling T cell-mediated responses in experimental models of organ-specific autoimmunity, chronic infection, and anti-tumor immunity. These findings highlight the importance of elucidating the molecular underpinnings of GITR-induced signaling. We propose that GITR employs adapter proteins, including TNFR-associated factors (TRAFs), to regulate diverse signaling pathways and transcriptional programs that control the interplay between Treg cells and immune effector cells.


Asunto(s)
Transducción de Señal , Linfocitos T Reguladores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antígenos CD , Antígenos de Diferenciación/metabolismo , Enfermedades Autoinmunes/inmunología , Linfocitos B/metabolismo , Antígeno CTLA-4 , Factores de Transcripción Forkhead/metabolismo , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Glucocorticoides/metabolismo , Humanos , Sistema Inmunológico , Inflamación , Interleucina-2/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/metabolismo , Transcripción Genética
6.
Immunol Res ; 32(1-3): 15-29, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16106056

RESUMEN

Effector cells play a crucial role in the immune system of higher vertebrates in eliminating invading pathogens and transformed cells that could cause disease or death of the individual. To be effective and specific, immune responses have to distinguish between self and nonself. Mechanisms of central and peripheral tolerance have evolved to control effector cells that could respond to autoantigens. Regulatory T-cells (Treg cells) are critical modulators of effector cells in the periphery that suppress autoreactive T-cells but are also involved in modulating immune responses against invading pathogens. Identification of surface markers of Treg cells and the development of in vitro systems to study the suppressive function of Treg cells have revealed distinct phenotypic and functional subsets of Treg cells. Several tumor necrosis factor receptor (TNFR) family members have been shown to play a role in the development, homeostasis, and suppressor function of Treg cells. Recent findings suggest that TNFRs and other cell-surface molecules of Treg cells can be explored for therapeutic strategies targeting autoimmune disorders, cancer, and immune responses against pathogens.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad , Humanos , Ratones , Modelos Inmunológicos , Receptores Inmunológicos/metabolismo , Autotolerancia
7.
J Leukoc Biol ; 75(2): 350-7, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14634058

RESUMEN

Members of the TNF receptor (TNFR) superfamily are cell-surface proteins that can be found on most cell types including lymphocytes. Although some TNFR-related molecules are constitutively expressed, others, such as CD30 and Ox40, are induced upon activation of lymphocytes. CD30 and Ox40 are predominantly expressed on activated T helper (T(h))2 cells. Both receptors can activate c-Jun N-terminal kinase (JNK) and nuclear factor-kappaB (NF-kappaB) and have been suggested to play costimulatory roles in lymphocyte activation. To gain further insight into events triggered by both TNFR-related molecules, a detailed analysis of their expression patterns has been performed. We found that CD30 and Ox40 were coexpressed on T(h)2 cells. However, in contrast to CD30, Ox40 was also expressed on T(h)1 cells. Although expression of both receptors is augmented by interleukin-4, only CD30 expression is dependent on signal transducer and activator of transcription (STAT)-6-mediated signaling. Differences in the regulatory pathways controlling expression of CD30 and Ox40 suggest distinct, functional effects triggered by the two TNFR-related molecules during lymphocyte activation.


Asunto(s)
Regulación de la Expresión Génica , Antígeno Ki-1/biosíntesis , Receptores del Factor de Necrosis Tumoral/biosíntesis , Subgrupos de Linfocitos T/metabolismo , Animales , Diferenciación Celular , Interleucina-4/farmacología , Antígeno Ki-1/análisis , Activación de Linfocitos , Ratones , Ratones Endogámicos , Receptores OX40 , Receptores del Factor de Necrosis Tumoral/análisis , Factor de Transcripción STAT6 , Transducción de Señal , Transactivadores/fisiología
8.
Diabetes ; 62(8): 2870-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23835340

RESUMEN

The dysregulation of receptor tyrosine kinases (RTKs) in multiple cell types during chronic inflammation is indicative of their pathogenic role in autoimmune diseases. Among the many RTKs, vascular endothelial growth factor receptor (VEGFR) stands out for its multiple effects on immunity, vascularization, and cell migration. Herein, we examined whether VEGFR participated in the pathogenesis of type 1 diabetes (T1D) in nonobese diabetic (NOD) mice. We found that RTK inhibitors (RTKIs) and VEGF or VEGFR-2 antibodies reversed diabetes when administered at the onset of hyperglycemia. Increased VEGF expression promoted islet vascular remodeling in NOD mice, and inhibition of VEGFR activity with RTKIs abrogated the increase in islet vascularity, impairing T-cell migration into the islet and improving glucose control. Metabolic studies confirmed that RTKIs worked by preserving islet function, as treated mice had improved glucose tolerance without affecting insulin sensitivity. Finally, examination of human pancreata from patients with T1D revealed that VEGFR-2 was confined to the islet vascularity, which was increased in inflamed islets. Collectively, this work reveals a previously unappreciated role for VEGFR-2 signaling in the pathogenesis of T1D by controlling T-cell accessibility to the pancreatic islets and highlights a novel application of VEGFR-2 antagonists for the therapeutic treatment of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Indoles/uso terapéutico , Islotes Pancreáticos/efectos de los fármacos , Páncreas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirroles/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Femenino , Humanos , Hiperglucemia/patología , Hiperglucemia/fisiopatología , Indoles/farmacología , Inflamación/tratamiento farmacológico , Inflamación/patología , Inflamación/fisiopatología , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Páncreas/irrigación sanguínea , Páncreas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Sunitinib
9.
Fibrogenesis Tissue Repair ; 5(1): 21, 2012 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-23270325

RESUMEN

BACKGROUND: The possibility of extracting RNA and measuring RNA expression from paraffin sections can allow extensive investigations on stored paraffin samples obtained from diseased livers and could help with studies of the natural history of liver fibrosis and inflammation, and in particular, correlate basic mechanisms to clinical outcomes. RESULTS: To address this issue, a pilot study of multiplex gene expression using branched-chain DNA technology was conducted to directly measure mRNA expression in formalin-fixed paraffin-embedded needle biopsy samples of human liver. Twenty-five genes were selected for evaluation based on evidence obtained from human fibrotic liver, a rat BDL model and in vitro cultures of immortalized human hepatic stellate cells. The expression levels of these 25 genes were then correlated with liver fibrosis and inflammation activity scores. Statistical analysis revealed that three genes (COL3A1, KRT18, and TUBB) could separate fibrotic from non-fibrotic samples and that the expression of ten genes (ANXA2, TIMP1, CTGF, COL4A1, KRT18, COL1A1, COL3A1, ACTA2, TGFB1, LOXL2) were positively correlated with the level of liver inflammation activity. CONCLUSION: This is the first report describing this multiplex technique for liver fibrosis and has provided the proof of concept of the suitability of RNA extracted from paraffin sections for investigating the modulation of a panel of proinflammatory and profibrogenic genes. This pilot study suggests that this technique will allow extensive investigations on paraffin samples from diseased livers and possibly from any other tissue. Using identical or other genes, this multiplex expression technique could be applied to samples obtained from extensive patient cohorts with stored paraffin samples in order to correlate gene expression with valuable clinically relevant information. This method could be used to provide a better understanding of the mechanisms of liver fibrosis and inflammation, its progression, and help development of new therapeutic approaches for this indication.

10.
Hepatol Res ; 40(11): 1128-41, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20880061

RESUMEN

AIM: To evaluate the usefulness of a platelet-derived growth factor (PDGF)-B specific monoclonal antibody (mAb) as a therapeutic agent to treat chronic liver fibrosis. METHODS: Liver fibrosis was induced in ICR mice by bile duct ligation (BDL) or BALB/c mice by weekly injection of concanavalin A (ConA) for 4 or 8 weeks. A mAb specific for mouse and human PDGF-B chain, AbyD3263, was generated, tested in vitro and administered twice a week throughout the experimental period. RESULTS: AbyD3263 showed neutralizing activity against mouse and human PDGF-B chain in cell-based assays, as measured in vitro by inhibition of phosphorylation of PDGF receptor ß and proliferation of hepatic stellate cells induced by PDGF-BB. The half life of AbyD3263 in mice exceeded 7 days and dosing of animals twice a week resulted in constant plasma levels of the mAb. Induction of liver fibrosis by BDL and ConA resulted in elevated levels of alanine aminotransferase (ALT) in plasma and hydroxyproline in the liver. Treatment with AbyD3263 did not modify ALT levels, but significantly reduced hydroxyproline content in the liver with a maximum reduction of 39% and 54% in the BDL and ConA models, respectively, compared to controls. Conclusios: Consistent with the notion that PDGF-BB plays an important role in the progression of liver fibrosis, AbyD3263 exhibits efficacy in pre-clinical disease models suggesting that pharmacological inhibition of PDGF-B chain may be a therapeutic approach to treat liver fibrosis.

11.
J Biol Chem ; 281(13): 8559-64, 2006 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16452475

RESUMEN

Tumor necrosis factor receptor (TNFR) family members such as glucocorticoid-induced TNFR (GITR) control T cell activation, differentiation, and effector functions. Importantly, GITR functions as a pivotal regulator of physiologic and pathologic immune responses by abrogating the suppressive effects of T regulatory cells and costimulating T effector cells. However, the molecular mechanisms underlying GITR-triggered signal transduction pathways remain unclear. Interestingly, GITR-induced stimulation of TNFR-associated factor (TRAF) 5-deficient T cells resulted in decreased activation of nuclear factor kappaB as well as the mitogen-activated protein kinases p38 and extracellular signal-regulated protein kinase, whereas activation of c-Jun N-terminal kinase was less affected. Consistent with impaired signaling, costimulatory effects of GITR were diminished in TRAF5-/- T cells. In sum, our studies indicate that TRAF5 plays a crucial role in GITR-induced signaling pathways that augment T cell activation.


Asunto(s)
Glucocorticoides/farmacología , Transducción de Señal , Linfocitos T/efectos de los fármacos , Factor 5 Asociado a Receptor de TNF/deficiencia , Factor 5 Asociado a Receptor de TNF/metabolismo , Animales , Línea Celular , Genes Reporteros , Humanos , Immunoblotting , Luciferasas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/análisis , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Factor 5 Asociado a Receptor de TNF/genética , Transfección
12.
J Immunol ; 174(12): 7869-74, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15944292

RESUMEN

Glucocorticoid-induced TNFR (GITR) is a member of the TNFR family that can inhibit the suppressive function of regulatory T cells and promote the survival and activation of T cells. However, little is known about the molecular mechanisms regulating T cell survival and activation downstream of GITR. To gain further insight into the cellular events and signaling pathways triggered by GITR, survival, proliferation, and cytokine production as well as activation of MAPKs and NF-kappaB were monitored after cross-linking of the receptor on naive and activated T cells. GITR cross-linking provided costimulation of naive and activated T cells and resulted in activation of MAPKs and NF-kappaB. Although GITR-induced signaling pathways augmented the survival of naive T cells, they were not sufficient to inhibit activation-induced cell death triggered by CD3 cross-linking of activated T cells. Differences in the contributions of GITR to cell survival between naive and activated T cells suggest that the receptor triggers specific pathways depending on the activation state of the T cell.


Asunto(s)
Activación de Linfocitos/inmunología , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Proteínas Portadoras/metabolismo , Muerte Celular/inmunología , Proliferación Celular , Supervivencia Celular/inmunología , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Interleucina-2/biosíntesis , Ligandos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , FN-kappa B/metabolismo , FN-kappa B/fisiología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Fase de Descanso del Ciclo Celular/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología , Factores de Necrosis Tumoral
13.
J Immunol ; 174(12): 7875-82, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15944293

RESUMEN

Glucocorticoid-induced TNFR (GITR) has been implicated as an essential regulator of immune responses to self tissues and pathogens. We have recently shown that GITR-induced cellular events promote survival of naive T cells, but are insufficient to protect against activation-induced cell death. However, the molecular mechanisms of GITR-induced signal transduction that influence physiologic and pathologic immune responses are not well understood. TNFR-associated factors (TRAFs) are pivotal adapter proteins involved in signal transduction pathways of TNFR-related proteins. Yeast two-hybrid assays and studies in HEK293 cells and primary lymphocytes indicated interactions between TRAF2 and GITR mediated by acidic residues in the cytoplasmic domain of the receptor. GITR-induced activation of NF-kappaB is blocked by A20, an NF-kappaB-inducible protein that interacts with TRAFs and functions in a negative feedback mechanism downstream of other TNFRs. Interestingly, in contrast with its effects on signaling triggered by other TNFRs, our functional studies revealed that TRAF2 plays a novel inhibitory role in GITR-triggered NF-kappaB activation.


Asunto(s)
Regulación hacia Abajo/inmunología , Activación de Linfocitos/inmunología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Fase de Descanso del Ciclo Celular/inmunología , Factor 2 Asociado a Receptor de TNF/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/fisiología , Línea Celular , Secuencia Conservada , Cisteína Endopeptidasas , Citoplasma/química , Citoplasma/metabolismo , Proteínas de Unión al ADN , Regulación hacia Abajo/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Humanos , Concentración de Iones de Hidrógeno , Péptidos y Proteínas de Señalización Intracelular , Activación de Linfocitos/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Nucleares , Estructura Terciaria de Proteína/genética , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Proteínas/farmacología , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/metabolismo , Fase de Descanso del Ciclo Celular/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Transfección , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factores de Necrosis Tumoral
14.
J Immunol ; 174(3): 1621-9, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15661925

RESUMEN

Tissue injury triggers inflammatory responses that may result in release of degradation products or exposure of cryptic domains of extracellular matrix components. Previously, we have shown that a cryptic peptide (AQARSAASKVKVSMKF) in the alpha-chain of laminin-10 (alpha5beta1gamma1), a prominent basement membrane component, is chemotactic for both neutrophils (PMNs) and macrophages (Mphis) and induces matrix metalloproteinase-9 (MMP-9) production. To determine whether AQARSAASKVKVSMKF has additional effects on inflammatory cells, we performed microarray analysis of RNA from RAW264.7 Mphis stimulated with AQARSAASKVKVSMKF. Several cytokines and cytokine receptors were increased >3-fold in response to the laminin alpha5 peptide. Among these were TNF-alpha and one of its receptors, the p75 TNFR (TNFR-II), increasing 3.5- and 5.7-fold, respectively. However, the peptide had no effect on p55 TNFR (TNFR-I) expression. Corroborating the microarray data, the protein levels of TNF-alpha and TNFR-II were increased following stimulation of RAW264.7 cells with AQARSAASKVKVSMKF. In addition, we determined that the production of TNF-alpha and TNFR-II in response to AQARSAASKVKVSMKF preceded the production of MMP-9. Furthermore, using primary Mphis from mice deficient in TNFR-I, TNFR-II, or both TNF-alpha receptors (TNFRs), we determined that AQARSAASKVKVSMKF induces MMP-9 expression by Mphis through a pathway triggered by TNFR-II. However, TNF-alpha signaling is not required for AQARSAASKVKVSMKF-induced PMN release of MMP-9 or PMN emigration. These data suggest that interactions of inflammatory cells with basement membrane components may orchestrate immune responses by inducing expression of cytokines, recruitment of inflammatory cells, and release of proteinases.


Asunto(s)
Factores Quimiotácticos/fisiología , Mediadores de Inflamación/fisiología , Laminina/fisiología , Fragmentos de Péptidos/fisiología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Regulación de la Expresión Génica/inmunología , Humanos , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Monocitos/inmunología , Monocitos/metabolismo , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA