Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(9)2020 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397343

RESUMO

Regulatory T cells (Tregs) are essential for the maintenance of tolerance to self and non-self through cell-intrinsic and cell-extrinsic mechanisms. Peripheral Tregs survival and clonal expansion largely depend on IL-2 and access to co-stimulatory signals such as CD28. Engagement of tumor necrosis factor receptor (TNFR) superfamily members, in particular TNFR2 and DR3, contribute to promote peripheral Tregs expansion and sustain their survival. This property can be leveraged to enhance tolerance to allogeneic transplants by tipping the balance of Tregs over conventional T cells during the course of immune reconstitution. This is of particular interest in peri-transplant tolerance induction protocols in which T cell depletion is applied to reduce the frequency of alloreactive T cells or in conditioning regimens that allow allogeneic bone marrow transplantation. These conditioning regimens are being implemented to limit long-term side effects of continuous immunosuppression and facilitate the establishment of a state of donor-specific tolerance. Lymphopenia-induced homeostatic proliferation in response to cytoreductive conditioning is a window of opportunity to enhance preferential expansion of Tregs during homeostatic proliferation that can be potentiated by agonist stimulation of TNFR.


Assuntos
Transplante de Medula Óssea , Depleção Linfocítica , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Receptores Tipo II do Fator de Necrose Tumoral/fisiologia , Linfócitos T Reguladores/imunologia , Abatacepte/farmacologia , Transferência Adotiva , Aloenxertos , Animais , Diferenciação Celular , Divisão Celular , Rejeição de Enxerto/prevenção & controle , Transplante de Coração , Homeostase , Humanos , Tolerância Imunológica , Transfusão de Linfócitos , Linfopenia/etiologia , Linfopenia/imunologia , Camundongos , Modelos Imunológicos , Linfócitos T Reguladores/efeitos dos fármacos , Condicionamento Pré-Transplante , Imunologia de Transplantes , Fator de Necrose Tumoral alfa/fisiologia
2.
Inflamm Bowel Dis ; 25(1): 14-26, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30295722

RESUMO

Background: TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain-receptor-3 (DR3), are multifunctional mediators of effector and regulatory immunity. We aimed to evaluate the functional role and therapeutic potential of TL1A/DR3 signaling in Crohn's disease-like ileitis. Methods: Ileitis-prone SAMP1/YitFc (SAMP) and TNFΔARE/+ mice were rendered deficient for DR3 or TL1A by microsatellite marker-assisted backcrossing. Pathological and immunological characteristics were compared between control and knockout mice, and mucosal immunophenotype was analyzed by Nanostring microarray assay. The therapeutic effect of pharmacological TL1A neutralization was also investigated. Results: DR3 deficiency was associated with restoration of a homeostatic mucosal immunostat in SAMP mice through the regulation of several pro- and anti-inflammatory genes. This led to suppression of effector immunity, amelioration of ileitis severity, and compromised ability of either unfractionated CD4+ or CD4+CD45RBhi mucosal lymphocytes to transfer ileitis to severe combined immunodeficient mice recipients. TNF-driven ileitis was also prevented in TNFΔARE/+xDR3-/- mice, in association with decreased expression of the pro-inflammatory cytokines TNF and IFN-γ. In contrast to DR3, TL1A was dispensable for the development of ileitis although it affected the kinetics of inflammation, as TNFΔARE/+xTL1A-/- demonstrated delayed onset of inflammation, whereas administration of a neutralizing, anti-TL1A antibody ameliorated early but not late TNFΔARE/+ ileitis. Conclusion: We found a prominent pro-inflammatory role of DR3 in chronic ileitis, which is only partially mediated via interaction with TL1A, raising the possibility for additional DR3 ligands. Death-domain-receptor-3 appears to be a master regulator of mucosal homeostasis and inflammation and may represent a candidate therapeutic target for chronic inflammatory conditions of the bowel.


Assuntos
Doença de Crohn/complicações , Regulação da Expressão Gênica , Ileíte/prevenção & controle , Inflamação/prevenção & controle , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Ileíte/etiologia , Inflamação/etiologia , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
4.
BMC Nephrol ; 15: 178, 2014 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-25399326

RESUMO

BACKGROUND: Death receptors (DRs) play an important role in renal pathology. We have shown that DR3 is inducibly expressed on renal tubular epithelial cells in the setting of inflammatory injuries. In this study we investigate the expression of DR3 in renal endothelial cells and their response to TL1A, the only known ligand of DR3. METHODS: We did RT-PCR, flow cytometry and subcellular immunoblotting to examine the expression and function of DR3 in cells in vitro. We did organ culture of human and mouse tissue to examine expression and signal of DR3 in vivo. RESULTS: DR3 is expressed in some interstitial vascular endothelial cells (EC) in human kidney in situ; these EC also respond to its ligand TL1A by activating NF-κB. Very low levels of DR3 can be detected on the cell surface of cultured human umbilical vein (HUV) EC, which do not respond to TL1A. HUVEC transfected to overexpress DR3 become responsive to TL1A, assessed by IκBα degradation and E-selectin induction, indicating that the signaling components needed for DR3 responsiveness are expressed. TL1A induces NF-κB activation in EC in renal and cardiac tissue from wild type but not DR3 knock-out mice. CONCLUSION: TL1A and DR3 activate NF-κB in vascular endothelial cells, and can be an important regulator of renal interstitial vascular injury.


Assuntos
Células Endoteliais/metabolismo , Rim/citologia , NF-kappa B/metabolismo , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas I-kappa B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/química , Inibidor de NF-kappaB alfa , Técnicas de Cultura de Órgãos , Membro 25 de Receptores de Fatores de Necrose Tumoral/biossíntese , Membro 25 de Receptores de Fatores de Necrose Tumoral/deficiência , Proteínas Recombinantes/farmacologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia
5.
Methods Mol Biol ; 1155: 69-79, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24788174

RESUMO

Death receptor 3 (DR3; also designated as Wsl-1, Apo3, LARD, TRAMP, TNFRSF25, and TR3) is a member of the tumor necrosis factor (TNF) receptor superfamily that has emerged as a major regulator of inflammation and autoimmune diseases. DR3 contains a homologous intracellular region called the death domain (DD) that can bind adaptor proteins, which also contain a DD, initiating cellular responses such as caspase activation and apoptotic cell death. However, in other circumstances DR3 can initiate induction of transcription genes and gene products that can prevent cell death from occurring. Our laboratory has reported an inducible expression of DR3 in human and mouse tubular epithelial cells in renal injury, but its function in these setting still remains unclear. To directly manipulate and evaluate the role of DR3 in vivo, I have used an in vitro organ culture (OC) model, which I have developed in our laboratory. In this chapter, I will describe in detail the OC model used to study the role of DR3 in renal injury and discuss its advantages and limitations. In my hands, the OC model has proven to be an efficient tool for studying human cell heterogeneity, basal and regulated receptor expression, signalling pathways, and various biological responses not readily achievable in traditional cell culture models. Various assays can be carried out on organ cultures including histology, biochemistry, cell biology, and molecular biology, which will not be described in detail in this chapter.


Assuntos
Injúria Renal Aguda/prevenção & controle , Modelos Biológicos , Técnicas de Cultura de Órgãos/métodos , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout
6.
Mediators Inflamm ; 2013: 258164, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24453414

RESUMO

TNF-like ligand 1A (TL1A), which binds its cognate receptor DR3 and the decoy receptor DcR3, is an identified member of the TNF superfamily. TL1A exerts pleiotropic effects on cell proliferation, activation, and differentiation of immune cells, including helper T cells and regulatory T cells. TL1A and its two receptors expression is increased in both serum and inflamed tissues in autoimmune diseases such as inflammatory bowel disease (IBD), rheumatoid arthritis (RA), and ankylosing spondylitis (AS). Polymorphisms of the TNFSF15 gene that encodes TL1A are associated with the pathogenesis of irritable bowel syndrome, leprosy, and autoimmune diseases, including IBD, AS, and primary biliary cirrhosis (PBC). In mice, blocking of TL1A-DR3 interaction by either antagonistic antibodies or deletion of the DR3 gene attenuates the severity of multiple autoimmune diseases, whereas sustained TL1A expression on T cells or dendritic cells induces IL-13-dependent small intestinal inflammation. This suggests that modulation of TL1A-DR3 interaction may be a potential therapeutic target in several autoimmune diseases, including IBD, RA, AS, and PBC.


Assuntos
Doenças Autoimunes/etiologia , Inflamação/etiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Artrite Reumatoide/etiologia , Humanos , Doenças Inflamatórias Intestinais/etiologia , Psoríase/etiologia , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
7.
J Immunol ; 189(7): 3311-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22956587

RESUMO

TNFR superfamily (TNFRSF)4 (OX40, CD134) and TNFRSF25 are costimulatory receptors that influence CD4(+) and CD8(+) T cell responses to cognate Ag. Independently, these receptors have been described to stimulate overlapping functions, including enhanced proliferation and activation for both regulatory T cells (CD4(+)Foxp3(+); Tregs) and conventional T cells (CD4(+)Foxp3(-) or CD8(+)Foxp3(-); Tconvs). To determine the relative functionality of TNFRSF4 and TNFRSF25 in T cell immunity, the activity of TNFRSF4 and TNFRS25 agonistic Abs was compared in the context of both traditional protein/adjuvant (OVA/aluminum hydroxide) and CD8(+)-specific heat shock protein-based (gp96-Ig) vaccine approaches. These studies demonstrate that both TNFRSF4 and TNFRSF25 independently and additively costimulate vaccine-induced CD8(+) T cell proliferation following both primary and secondary Ag challenge. In contrast, the activities of TNFRSF4 and TNFRSF25 were observed to be divergent in the costimulation of CD4(+) T cell immunity. TNFRSF4 agonists were potent costimulators of OVA/aluminum hydroxide-induced CD4(+) Tconv proliferation, but they only weakly costimulated Treg proliferation and IgG2a production, whereas TNFRSF25 agonists were strong costimulators of Treg proliferation, producers of IgG1, IgG2a, and IgG2b, and weak costimulators of CD4(+) Tconv proliferation. Interestingly, Ag-specific cellular and humoral responses were uncoupled upon secondary immunization, which was dramatically affected by the presence of TNFRSF4 or TNFRSF25 costimulation. These studies highlight the overlapping but nonredundant activities of TNFRSF4 and TNFRSF25 in T cell immunity, which may guide the application of receptor agonistic agents as vaccine adjuvants for infectious disease and tumor immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunização Secundária , Ativação Linfocitária/imunologia , Receptores OX40/administração & dosagem , Membro 25 de Receptores de Fatores de Necrose Tumoral/administração & dosagem , Vacinas/administração & dosagem , Vacinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/fisiologia , Compostos de Alúmen/administração & dosagem , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Proliferação de Células , Células Cultivadas , Apresentação Cruzada/imunologia , Imunização Secundária/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Receptores OX40/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia
8.
FASEB J ; 26(8): 3575-86, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593543

RESUMO

Death receptor 3 (DR3, TNFRSF25), the closest family relative to tumor necrosis factor receptor 1, promotes CD4(+) T-cell-driven inflammatory disease. We investigated the in vivo role of DR3 and its ligand TL1A in viral infection, by challenging DR3-deficient (DR3(KO)) mice and their DR3(WT) littermates with the ß-herpesvirus murine cytomegalovirus or the poxvirus vaccinia virus. The phenotype and function of splenic T-cells were analyzed using flow cytometry and molecular biological techniques. We report surface expression of DR3 by naive CD8(+) T cells, with TCR activation increasing its levels 4-fold and altering the ratio of DR3 splice variants. T-cell responses were reduced up to 90% in DR3(KO) mice during acute infection. Adoptive transfer experiments indicated this was dependent on T-cell-restricted expression of DR3. DR3-dependent CD8(+) T-cell expansion was NK and CD4 independent and due to proliferation, not decreased cell death. Notably, impaired immunity in DR3(KO) hosts on a C57BL/6 background was associated with 4- to 7-fold increases in viral loads during the acute phase of infection, and in mice with suboptimal NK responses was essential for survival (37.5%). This is the first description of DR3 regulating virus-specific T-cell function in vivo and uncovers a critical role for DR3 in mediating antiviral immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Herpesviridae/imunologia , Muromegalovirus/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Transferência Adotiva , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Camundongos , Camundongos Knockout , Carga Viral
9.
Am J Pathol ; 180(4): 1454-64, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22330679

RESUMO

The expression of death receptor 3 (DR3), a member of the tumor necrosis factor (TNF) receptor superfamily, is up-regulated in human tubular epithelial cells (TECs) during renal injury, but its function in this setting remains unknown. We used cisplatin to induce renal injury in wild-type (DR3(+/+)) or congenitally deficient DR3(-/-) mice to examine the in vivo role of DR3. Cisplatin induced the expression of DR3, its ligand, TNF-like ligand 1A (TL1A), and TNF in TECs, as observed in human renal injury. Cisplatin increased apoptotic death of DR3(-/-) TECs by twofold compared with DR3(+/+) TECs, whereas it reduced the number of tubules expressing phospho-NF-κBp65(Ser276) by 50% at 72 hours. Similar degrees of induction of DR3, TL1A, and TNF, and changes in apoptosis and phospho-NF-κBp65(Ser276), were obtained in mouse kidney organ cultures treated with cisplatin for 3 hours, suggesting a direct effect on TECs. TNF was implicated in mediating cisplatin-induced tubular damage given that the in vivo co-administration of GM6001, an inhibitor of TNF maturation and release, significantly reduced TNF production and tubular damage. Moreover, TNF exacerbated, whereas TL1A reduced, cisplatin-induced apoptosis in the DR3(+/+) mouse proximal tubule cell line, TKPTS. Our data demonstrate that cisplatin-induced nephrotoxicity is mitigated by DR3 signaling, suggesting that this occurs by antagonizing pro-apoptotic signals induced by TNF. Therefore, activating DR3 may be beneficial in reducing acute kidney injury.


Assuntos
Injúria Renal Aguda/patologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Dipeptídeos/farmacologia , Interações Medicamentosas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Ligantes , Camundongos , Camundongos Mutantes , NF-kappa B/metabolismo , Técnicas de Cultura de Órgãos , Fosforilação/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/biossíntese , Membro 25 de Receptores de Fatores de Necrose Tumoral/deficiência , Transdução de Sinais/fisiologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima/efeitos dos fármacos
10.
J Clin Invest ; 120(10): 3629-40, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20890040

RESUMO

TNF receptor superfamily member 25 (TNFRSF25; also known as DR3, and referred to herein as TNFR25) is constitutively and highly expressed by CD4(+)FoxP3(+) Tregs. However, its function on these cells has not been determined. Here we used a TNFR25-specific agonistic monoclonal antibody, 4C12, to study the effects of TNFR25 signaling on Tregs in vivo in mice. Signaling through TNFR25 induced rapid and selective expansion of preexisting Tregs in vivo such that they became 30%-35% of all CD4(+) T cells in the peripheral blood within 4 days. TNFR25-induced Treg proliferation was dependent upon TCR engagement with MHC class II, IL-2 receptor, and Akt signaling, but not upon costimulation by CD80 or CD86; it was unaffected by rapamycin. TNFR25-expanded Tregs remained highly suppressive ex vivo, and Tregs expanded by TNFR25 in vivo were protective against allergic lung inflammation, a mouse model for asthma, by reversing the ratio of effector T cells to Tregs in the lung, suppressing IL-13 and Th2 cytokine production, and blocking eosinophil exudation into bronchoalveolar fluid. Our studies define what we believe to be a novel mechanism for Treg control and important functions for TNFR25 in regulating autoaggression that balance its known role in enhancing autoimmunity.


Assuntos
Asma/prevenção & controle , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Asma/imunologia , Modelos Animais de Doenças , Antígenos de Histocompatibilidade Classe II/fisiologia , Interleucina-2/fisiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/agonistas
11.
J Immunol ; 184(10): 5827-34, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20410491

RESUMO

TNF-like protein 1A (TL1A), a TNF superfamily cytokine that binds to death receptor 3 (DR3), is highly expressed in macrophage foam cell-rich regions of atherosclerotic plaques, although its role in foam cell formation has yet to be elucidated. We investigated whether TL1A can directly stimulate macrophage foam cell formation in both THP-1 and primary human monocyte-derived macrophages with the underlying mechanisms involved. We demonstrated that TL1A promotes foam cell formation in human macrophages in vitro by increasing both acetylated and oxidized low-density lipoprotein uptake, by enhancing intracellular total and esterified cholesterol levels and reducing cholesterol efflux. This imbalance in cholesterol homeostasis is orchestrated by TL1A-mediated changes in the mRNA and protein expression of several genes implicated in the uptake and efflux of cholesterol, such as scavenger receptor A and ATP-binding cassette transporter A1. Furthermore, through the use of virally delivered DR3 short-hairpin RNA and bone marrow-derived macrophages from DR3 knockout mice, we demonstrate that DR3 can regulate foam cell formation and contributes significantly to the action of TL1A in this process in vitro. We show, for the first time, a novel proatherogenic role for both TL1A and DR3 that implicates this pathway as a target for the therapeutic intervention of atherosclerosis.


Assuntos
Diferenciação Celular/imunologia , Células Espumosas/citologia , Células Espumosas/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Transdução de Sinais/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Transporte Biológico/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Feminino , Células Espumosas/patologia , Humanos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Knockout , Membro 25 de Receptores de Fatores de Necrose Tumoral/deficiência , Regulação para Cima/imunologia
12.
J Neurosci ; 30(10): 3782-92, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20220013

RESUMO

Death receptor 3 is a proinflammatory member of the immunomodulatory tumor necrosis factor receptor superfamily, which has been implicated in several inflammatory diseases such as arthritis and inflammatory bowel disease. Intriguingly however, constitutive DR3 expression has been detected in the brains of mice, rats, and humans, although its neurological function remains unknown. By mapping the normal brain expression pattern of DR3, we found that DR3 is expressed specifically by cells of the neuron lineage in a developmentally regulated and region-specific pattern. Behavioral studies on DR3-deficient (DR3(ko)) mice showed that constitutive neuronal DR3 expression was required for stable motor control function in the aging adult. DR3(ko) mice progressively developed behavioral defects characterized by altered gait, dyskinesia, and hyperactivity, which were associated with elevated dopamine and lower serotonin levels in the striatum. Importantly, retrograde tracing showed that absence of DR3 expression led to the loss of corticostriatal innervation without significant neuronal loss in aged DR3(ko) mice. These studies indicate that DR3 plays a key nonredundant role in the retention of normal motor control function during aging in mice and implicate DR3 in progressive neurological disease.


Assuntos
Envelhecimento/fisiologia , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Destreza Motora/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Envelhecimento/genética , Animais , Comunicação Celular/genética , Comunicação Celular/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Corpo Estriado/crescimento & desenvolvimento , Corpo Estriado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurotransmissores/deficiência , Neurotransmissores/genética , Neurotransmissores/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/deficiência , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética
13.
PLoS One ; 5(1): e8579, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20062804

RESUMO

Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) is a transcription factor which regulates the expression of many cytoprotective genes. In the present study, we found that the expression of Nrf2 was suppressed in prostate tumor of the Transgenic Adenocarcinoma of Mouse Prostate (TRAMP) mice. Similarly, the expression of Nrf2 and the induction of NQO1 were also substantially suppressed in tumorigenic TRAMP C1 cells but not in non-tumorigenic TRAMP C3 cells. Examination of the promoter region of the mouse Nrf2 gene identified a CpG island, which was methylated at specific CpG sites in prostate TRAMP tumor and in TRAMP C1 cells but not in normal prostate or TRAMP C3 cells, as shown by bisulfite genomic sequencing. Reporter assays indicated that methylation of these CpG sites dramatically inhibited the transcriptional activity of the Nrf2 promoter. Chromatin immunopreceipitation (ChIP) assays revealed increased binding of the methyl-CpG-binding protein 2 (MBD2) and trimethyl-histone H3 (Lys9) proteins to these CpG sites in the TRAMP C1 cells as compared to TRAMP C3 cells. In contrast, the binding of RNA Pol II and acetylated histone H3 to the Nrf2 promoter was decreased. Furthermore, treatment of TRAMP C1 cells with DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-aza) and histone deacetylase (HDAC) inhibitor trichostatin A (TSA) restored the expression of Nrf2 as well as the induction of NQO1 in TRAMP C1 cells. Taken together, these results indicate that the expression of Nrf2 is suppressed epigenetically by promoter methylation associated with MBD2 and histone modifications in the prostate tumor of TRAMP mice. Our present findings reveal a novel mechanism by which Nrf2 expression is suppressed in TRAMP prostate tumor, shed new light on the role of Nrf2 in carcinogenesis and provide potential new directions for the detection and prevention of prostate cancer.


Assuntos
Epigênese Genética , Fator 2 Relacionado a NF-E2/genética , Neoplasias da Próstata/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Animais , Azacitidina/farmacologia , Ilhas de CpG , Metilação de DNA , Masculino , Camundongos , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Transcrição Gênica , Tricosantina/farmacologia
14.
Blood ; 113(21): 5352-60, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-19329781

RESUMO

Endothelial progenitor cells (EPCs) play a critical role in postnatal and tumor vasculogenesis. Vascular endothelial growth inhibitor (VEGI; TNFSF15) has been shown to inhibit endothelial cell proliferation by inducing apoptosis. We report here that VEGI inhibits the differentiation of EPCs from mouse bone marrow-derived Sca1(+) mononuclear cells. Analysis of EPC markers indicates a significant decline of the expression of endothelial cell markers, but not stem cell markers, on VEGI-treated cells. Consistently, the VEGI-treated cells exhibit a decreased capability to adhere, migrate, and form capillary-like structures on Matrigel. In addition, VEGI induces apoptosis of differentiated EPCs but not early-stage EPCs. When treated with VEGI, an increase of phospho-Erk and a decrease of phospho-Akt are detected in early-stage EPCs, whereas activation of nuclear factor-kappaB, jun N-terminal kinase, and caspase-3 is seen in differentiated EPCs. Furthermore, VEGI-induced apoptosis of differentiated EPC is, at least partly, mediated by death receptor-3 (DR3), which is detected on differentiated EPC only. VEGI-induced apoptosis signals can be inhibited by neutralizing antibodies against DR3 or recombinant extracellular domain of DR3. These findings indicate that VEGI may participate in the modulation of postnatal vasculogenesis by inhibiting EPC differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Endoteliais/citologia , Endotélio Vascular/citologia , Células-Tronco/citologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia
15.
J Exp Med ; 205(5): 1037-48, 2008 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-18411341

RESUMO

We identify the tumor necrosis factor receptor superfamily 25 (TNFRSF25)/TNFSF15 pair as critical trigger for allergic lung inflammation, which is a cardinal feature of asthma. TNFRSF25 (TNFR25) signals are required to exert T helper cell 2 (Th2) effector function in Th2-polarized CD4 cells and co-stimulate interleukin (IL)-13 production by glycosphingolipid-activated NKT cells. In vivo, antibody blockade of TNFSF15 (TL1A), which is the ligand for TNFR25, inhibits lung inflammation and production of Th2 cytokines such as IL-13, even when administered days after airway antigen exposure. Similarly, blockade of TNFR25 by a dominant-negative (DN) transgene, DN TNFR25, confers resistance to lung inflammation in mice. Allergic lung inflammation-resistant, NKT-deficient mice become susceptible upon adoptive transfer of wild-type NKT cells, but not after transfer of DN TNFR25 transgenic NKT cells. The TNFR25/TL1A pair appears to provide an early signal for Th2 cytokine production in the lung, and therefore may be a drug target in attempts to attenuate lung inflammation in asthmatics.


Assuntos
Hipersensibilidade/fisiopatologia , Inflamação/fisiopatologia , Pulmão/fisiopatologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Cricetinae/imunologia , Humanos , Imunoglobulina E/imunologia , Pulmão/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Ovalbumina/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Baço/imunologia , Células Th2/imunologia
16.
Carcinogenesis ; 28(2): 497-505, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17052998

RESUMO

The aryl hydrocarbon receptor (AhR) is a transcription factor that mediates the inhibitory effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on prostate growth and also modulates normal prostate development. This is evidenced by AhR null mice (Ahr-/-) having smaller dorsolateral and anterior prostates, even though all prostate lobes remain histologically normal. To test the hypothesis that loss of the AhR increases the rate of prostate carcinogenesis, the incidence of macroscopic prostate tumors was determined in Ahr+/+, Ahr+/- and Ahr-/- C57BL/6J transgenic adenocarcinoma of the mouse prostate (TRAMP) mice at 35, 70, 105, 140, 175 and 210 days of age. From 140 days, prostate tumor incidence was greater in Ahr-/- (60%) and Ahr+/- (43%) mice than in Ahr+/+ mice (16%). Allele quantification did not indicate a loss of the wild-type Ahr allele in heterozygous TRAMP tumors, suggesting that tumor formation in these mice was not due to a loss of Ahr heterozygosity. Prostatic SV40 large T antigen mRNA expression and protein localization were comparable in TRAMP mice of each Ahr genotype. Prostates from all mice of each Ahr genotype were histologically indistinguishable, exhibiting diffuse epithelial hyperplasia by 105 days of age. mRNA expression and protein localization for molecular markers of neuroendocrine differentiation, including chromogranin A and neuropilin-1, were elevated in prostate tumors compared to tumor-free ventral prostates, regardless of Ahr genotype or age. Taken together, these results demonstrate that the Ahr inhibits prostate carcinogenesis in C57BL/6J TRAMP mice by interfering with neuroendocrine differentiation.


Assuntos
Transformação Celular Neoplásica , Neoplasias da Próstata/prevenção & controle , Receptores de Hidrocarboneto Arílico/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Sequência de Bases , Peso Corporal , Diferenciação Celular , Cromogranina A/genética , Primers do DNA , Dosagem de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropilina-1/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , Receptores de Hidrocarboneto Arílico/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...