Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Cell Death Dis ; 7(6): e2287, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27362805

RESUMO

We previously reported that renal clear cell carcinoma cells (RCC) express both tumor necrosis factor receptor (TNFR)-1 and -2, but that, in organ culture, a TNF mutein that only engages TNFR1, but not TNFR2, causes extensive cell death. Some RCC died by apoptosis based on detection of cleaved caspase 3 in a minority TUNEL-positive cells but the mechanism of death in the remaining cells was unexplained. Here, we underpin the mechanism of TNFR1-induced cell death in the majority of TUNEL-positive RCC cells, and show that they die by necroptosis. Malignant cells in high-grade tumors displayed threefold to four fold higher expression of both receptor-interacting protein kinase (RIPK)1 and RIPK3 compared with non-tumor kidney tubular epithelium and low-grade tumors, but expression of both enzymes was induced in lower grade tumors in organ culture in response to TNFR1 stimulation. Furthermore, TNFR1 activation induced significant MLKL(Ser358) and Drp1(Ser616) phosphorylation, physical interactions in RCC between RIPK1-RIPK3 and RIPK3-phospho-MLKL(Ser358), and coincidence of phospho-MLKL(ser358) and phospho-Drp1(Ser616) at mitochondria in TUNEL-positive RCC. A caspase inhibitor only partially reduced the extent of cell death following TNFR1 engagement in RCC cells, whereas three inhibitors, each targeting a different step in the necroptotic pathway, were much more protective. Combined inhibition of caspases and necroptosis provided additive protection, implying that different subsets of cells respond differently to TNF-α, the majority dying by necroptosis. We conclude that most high-grade RCC cells express increased amounts of RIPK1 and RIPK3 and are poised to undergo necroptosis in response to TNFR1 signaling.


Assuntos
Apoptose , Carcinoma de Células Renais/patologia , Células Epiteliais/metabolismo , Neoplasias Renais/patologia , Túbulos Renais/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Acrilamidas/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Marcação In Situ das Extremidades Cortadas , Neoplasias Renais/genética , Necrose , Técnicas de Cultura de Órgãos , Quinazolinonas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos
2.
J Vasc Res ; 50(4): 324-31, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23860328

RESUMO

We describe a method for coculture of macro- or microvascular human endothelial cells (ECs) and pericytes (PCs) within a 3-dimensional (3-D) protein matrix resulting in lumenized EC cords invested by PCs. To prevent apoptotic cell death of ECs in 3-D culture, human umbilical vein or dermal microvascular ECs were transduced to express the antiapoptotic protein Bcl-2. To prevent PC-mediated gel contraction, the collagen-fibronectin gel was polymerized within a polyglycolic acid nonwoven matrix. Over the first 24-48 h, EC-only gels spontaneously formed cords that developed lumens via vacuolization; such vascular networks were maintained for up to 7 days. In EC-PC cocultures, PCs were recruited to the EC networks. PC investment of EC cords both limited the lumen diameter and increased the degree of vascular network arborization. Peg and socket junctions formed between ECs and PCs in this system, but dye transfer, indicative of gap junction formation, was not observed. This simple system can be used to analyze bidirectional signals between ECs and PCs in a 3-D geometry.


Assuntos
Comunicação Celular , Células Endoteliais/fisiologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Microvasos/fisiologia , Neovascularização Fisiológica , Pericitos/fisiologia , Apoptose , Movimento Celular , Células Cultivadas , Técnicas de Cocultura , Colágeno/metabolismo , Células Endoteliais/metabolismo , Fibronectinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Junções Intercelulares/fisiologia , Microvasos/metabolismo , Pericitos/metabolismo , Ácido Poliglicólico/química , Polimerização , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fatores de Tempo , Transdução Genética , Transfecção
3.
Am J Transplant ; 9(12): 2679-96, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19788501

RESUMO

Tumor necrosis factor (TNF) utilizes two receptors, TNFR1 and 2, to initiate target cell responses. We assessed expression of TNF, TNFRs and downstream kinases in cardiac allografts, and compared TNF responses in heart organ cultures from wild-type ((WT)C57BL/6), TNFR1-knockout ((KO)), TNFR2(KO), TNFR1/2(KO) mice. In nonrejecting human heart TNFR1 was strongly expressed coincidentally with inactive apoptosis signal-regulating kinase-1 (ASK1) in cardiomyocytes (CM) and vascular endothelial cells (VEC). TNFR2 was expressed only in VEC. Low levels of TNF localized to microvessels. Rejecting cardiac allografts showed increased TNF in microvessels, diminished TNFR1, activation of ASK1, upregulated TNFR2 co-expressed with activated endothelial/epithelial tyrosine kinase (Etk), increased apoptosis and cell cycle entry in CM. Neither TNFR was expressed significantly by cardiac fibroblasts. In (WT)C57BL/6 myocardium, TNF activated both ASK1 and Etk, and increased both apoptosis and cell cycle entry. TNF-treated TNFR1(KO) myocardium showed little ASK1 activation and apoptosis but increased Etk activation and cell cycle entry, while TNFR2(KO) myocardium showed little Etk activation and cell cycle entry but increased ASK1 activation and apoptosis. These observations demonstrate independent regulation and differential functions of TNFRs in myocardium, consistent with TNFR1-mediated cell death and TNFR2-mediated repair.


Assuntos
MAP Quinase Quinase Quinase 5/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Morte Celular , Endotélio Vascular/metabolismo , Ativação Enzimática , Rejeição de Enxerto/metabolismo , Transplante de Coração , Humanos , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Técnicas de Cultura de Órgãos , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/fisiologia
4.
Handb Exp Pharmacol ; (176 Pt 2): 135-56, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16999227

RESUMO

Vascular endothelial cells (ECs) perform a number of functions required to maintain homeostasis. Inflammation can cause EC injury and death which disrupt these processes and result in endothelial dysfunction. Three common mediators of EC injury in inflammation are macrophage-derived cytokines, such as tumour necrosis factor (TNF); neutrophil-generated reactive oxygen species (ROS) and cytolytic T lymphocytes (CTL). Here we describe the distinct but overlapping biochemical pathways of injury elicited by these different agents.


Assuntos
Apoptose , Endotélio Vascular/metabolismo , Inflamação/metabolismo , Animais , Endotélio Vascular/patologia , Granzimas/metabolismo , Humanos , Inflamação/patologia , Macrófagos/metabolismo , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
5.
Bone Marrow Transplant ; 32(11): 1077-84, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14625579

RESUMO

Cytotoxic T lymphocytes (CTLs) are important effector cells of graft-versus-host disease (GVHD) and vascular endothelial cells are target cells of allospecific CTL. A combined assessment of T-cell activation and endothelial injury should result in a specific and sensitive test for GVHD. We examined circulating T lymphocytes for effector molecules involved in CTL-mediated endothelial injury. We analyzed CD4 and CD8 T lymphocytes of 24 long-term survivors of allogeneic stem cell transplantation with or without GVHD, and nine healthy, age-matched controls for signs of CTL activation and endothelial injury. IFN-gamma transcript levels in CD8 T cells were significantly elevated in SCT recipients with GVHD compared to patients without GVHD (767 CD3epsilon units/T cell (376-2050) vs 211 CD3epsilon units/T cell (159-274), P=0.01). Fas ligand transcript levels in CD4 T cells were significantly elevated in SCT recipients without GVHD compared to patients with GVHD (20 CD3epsilon units/T cell (0-78) vs 0 CD3epsilon units/T cell (0-0), P=0.01). Von Willebrand factor plasma levels were high in patients with GVHD, but normal in patients without GVHD (209 (186-254) vs 120 (100-141), P=0.0005). This assessment of T-cell activation and endothelial injury results in a sensitive and specific test to identify patients with active chronic GVHD.


Assuntos
Endotélio Vascular/patologia , Doença Enxerto-Hospedeiro/diagnóstico , Técnicas de Diagnóstico Molecular , RNA Mensageiro/análise , Linfócitos T Citotóxicos/metabolismo , Biomarcadores/análise , Estudos de Casos e Controles , Proteína Ligante Fas , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Interferon gama/genética , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Sensibilidade e Especificidade , Sobreviventes , Transplante Homólogo , Fator de von Willebrand/análise
6.
Lab Invest ; 81(11): 1503-15, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11706058

RESUMO

Activation of the TNF signal transduction cascade is initiated by the interaction of TNF with either of two cell surface receptors, TNFR-1 and TNFR-2. The levels and regulation of expression of these two receptors has been extensively analyzed in cultured cells, but little is known of TNFR expression in situ. We analyzed the expression of TNFR-1 and -2 in normal human renal kidney and in renal transplants undergoing acute cellular rejection. Immunohistochemistry and immunogold electron microscopy indicated a strong expression of TNFR-1 on the endothelium of glomeruli of normal kidney. Immunogold colocalization for TNFR-1 and a marker of the trans-Golgi network (TGN-46) demonstrated TNFR-1 within the Golgi complex in endothelial cells in normal kidney, confirming our previous studies with cultured cells. TNFR-1 expression was lost in glomeruli from acutely rejecting kidney, but TNFR-1 was detected in abundance on infiltrating leukocytes in the interstitium of allografts with acute rejection. In contrast, TNFR-2 was demonstrated predominantly in epithelial cells of distal convoluted tubule (DCT) in acute rejection kidney near TNF-expressing leukocytes. TNF was absent in normal kidney, but present in rejecting allograft. TNF was found in infiltrating leukocytes and in adjacent tubular epithelial cells. In situ hybridization showed TNFR-1 mRNA within the endothelium of the glomeruli and of a few arterioles in normal kidney, whereas TNFR-2 mRNA was seen in tubular epithelial cells of the DCT in acute transplant rejection. These data reveal that there is both differential expression and regulation of the two TNF receptors in human kidney.


Assuntos
Antígenos CD/genética , Rejeição de Enxerto/fisiopatologia , Transplante de Rim , Rim/fisiologia , Receptores do Fator de Necrose Tumoral/genética , Doença Aguda , Antígenos CD/análise , Corantes , Amarelo de Eosina-(YS) , Corantes Fluorescentes , Expressão Gênica , Rejeição de Enxerto/patologia , Hematoxilina , Humanos , Rim/química , Rim/ultraestrutura , Falência Renal Crônica/patologia , Falência Renal Crônica/fisiopatologia , Falência Renal Crônica/cirurgia , Microscopia Imunoeletrônica , Receptores do Fator de Necrose Tumoral/análise , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/análise
7.
Ann N Y Acad Sci ; 941: 12-25, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11594565

RESUMO

Dermal microvascular endothelial cells (ECs) form a continuous lining that normally bars blood-borne T lymphocytes from entering the skin, but as part of the response to foreign antigen, dermal ECs undergo alterations in their surface proteins so as to provide signals to circulating T cells that lead to their activation and recruitment. Several observations suggest that human dermal microvascular ECs may help initiate cutaneous immune reactions by presentation of cognate antigens to circulating T memory cells: (1) antigen-specific inflammatory responses in the skin, as in other organs, involve accumulation of memory and effector T cell populations that are enriched in cells specific for the eliciting antigen; (2) recall responses to intradermal protein antigens in the skin start very rapidly within two hours of challenge; (3) dermal microvascular ECs in humans and other large mammals basally display high levels of class I and class II MHC molecules, the only known purpose of which is to present antigenic peptides to lymphocytes; (4) the lumen of dermal capillaries are narrower than the diameter of circulating T cells, ensuring surface contact; and (5) cultured human ECs effectively present antigens to resting memory T cells isolated from the circulation. Upon contact with activated T cells or their secreted products (cytokines), dermal ECs themselves become activated, increasing their capacity to recruit memory and effector T cell populations in an antigen-independent manner. Specifically, activated ECs express inducible leukocyte adhesion molecules such as E-selectin, ICAM-1, and VCAM-1; and several lines of evidence, including neutralizing antibody experiments and gene knockouts, have supported a role of these molecules in T cell recruitment. Dermal ECs have unique expression patterns of adhesion molecules that can determine the subsets of memory T cells that are recruited into the skin. For example, slow internalization of E-selectin allows more persistent expression of this protein on the surface of dermal ECs, favoring interactions with CLA-1+ T cells. VCAM-1 expression, normally confined to venular EC may extend to capillaries within the dermal papillae and contribute to epidermal inflammation, recruiting alpha4beta7 integrin-expressing T cells that also express the cadherin-binding integrin alphaEbeta7. New models involving transplantation of normal and genetically modified human dermal ECs into immunodeficient mice may be used to further explore these properties.


Assuntos
Apresentação de Antígeno , Movimento Celular , Endotélio Vascular/imunologia , Pele/imunologia , Linfócitos T/imunologia , Animais , Moléculas de Adesão Celular/metabolismo , Humanos , Memória Imunológica , Linfoma Cutâneo de Células T/imunologia , Camundongos , Modelos Animais , Pele/irrigação sanguínea , Neoplasias Cutâneas/imunologia
8.
Oncogene ; 20(44): 6482-91, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11607847

RESUMO

Tumor necrosis factor receptor-associated factors (TRAFS) were initially discovered as adaptor proteins that couple the tumor necrosis factor receptor family to signaling pathways. More recently they have also been shown to be signal transducers of Toll/interleukin-1 family members. Six members of the TRAF family have been identified. All TRAF proteins share a C-terminal homology region termed the TRAF domain that is capable of binding to the cytoplasmic domain of receptors, and to other TRAF proteins. In addition, TRAFs 2-6 have RING and zinc finger motifs that are important for signaling downstream events. TRAF proteins are thought to be important regulators of cell death and cellular responses to stress, and TRAF2, TRAF5 and TRAF6 have been demonstrated to mediate activation of NF-kappaB and JNK. TRAF proteins are expressed in normal and diseased tissue in a regulated fashion, suggesting that they play an important role in physiological and pathological processes.


Assuntos
Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais , Motivos de Aminoácidos , Animais , Humanos , Interleucina-1/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/metabolismo , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF , Fator 3 Associado a Receptor de TNF , Fator 4 Associado a Receptor de TNF , Fator 5 Associado a Receptor de TNF , Fator 6 Associado a Receptor de TNF , Fator de Transcrição AP-1/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral
9.
Transplantation ; 71(11): 1622-30, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11435975

RESUMO

BACKGROUND: We have previously demonstrated that human artery grafts transplanted to immunodeficient mice are infiltrated and injured by unsensitized allogeneic human T cells. We extended our investigations to human anti-porcine xenoresponses in this model. METHODS: Pig coronary artery segments were interposed into the infrarenal aorta of severe combined immunodeficiency/beige mice. After 7 days, certain recipients were reconstituted with human leukocytes and/or treated with proinflammatory cytokines. The grafts were harvested after 1-70 days and examined by histology, immunohistochemistry, and morphometry. RESULTS: Pig artery grafts from untreated mice had no evidence of injury, leukocytic infiltrate, or endothelial cell activation up to 70 days postoperatively, despite deposition of murine complement. Host reconstitution with human peripheral blood mononuclear cells resulted in a discrete population of circulating T cells that did not infiltrate or injure the grafts up to 28 days after adoptive transfer. Administration of porcine interferon-gamma for up to 28 days sustained the expression of graft vascular cell adhesion molecule-1 and major histocompatibility complex antigens, but did not initiate recruitment of human leukocytes. In contrast, treatment with human tumor necrosis factor for 7 days induced the de novo expression of porcine E-selectin by graft endothelial cells and elicited human T cell infiltration and human peripheral blood mononuclear cell-dependent vascular injury. CONCLUSIONS: The human peripheral blood mononuclear cell-severe combined immunodeficiency/beige mouse model identifies a significant difference between human T cell allogeneic and xenogeneic responses in vivo. Xenografts with quiescent endothelium are not infiltrated or injured by T cells under the same conditions in which allografts are rejected. Activation of pig coronary artery endothelial cells by human tumor necrosis factor, but not porcine interferon-gamma, elicits cellular xenoresponses.


Assuntos
Vasos Coronários/transplante , Endotélio Vascular/fisiologia , Endotélio Vascular/transplante , Imunodeficiência Combinada Severa/cirurgia , Animais , Artérias/efeitos dos fármacos , Artérias/patologia , Artérias/transplante , Células Sanguíneas/patologia , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Rejeição de Enxerto/induzido quimicamente , Humanos , Tolerância Imunológica , Interferon gama/farmacologia , Camundongos , Camundongos SCID , Imunodeficiência Combinada Severa/sangue , Suínos , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Imunologia de Transplantes , Fator de Necrose Tumoral alfa/farmacologia
10.
Exp Mol Pathol ; 70(3): 317-25, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11418010

RESUMO

Vascular endothelium is a major target of actions of the proinflammatory cytokine tumor necrosis factor (TNF). Increasingly, the intracellular pathways that are activated in response to TNF have been elucidated. Many of these pathways have proven to be cell type-specific, requiring that observations made in other cell types be confirmed or ruled out in endothelial cells (EC). In this review the authors will summarize the state of the field, emphasizing studies in cultured human EC.


Assuntos
Endotélio Vascular/fisiologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/fisiologia , Animais , Apoptose , Células Cultivadas , Endotélio Vascular/citologia , Ativação Enzimática , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Esfingomielina Fosfodiesterase/metabolismo
11.
Am J Pathol ; 158(5): 1757-65, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11337373

RESUMO

The protective genes that mediate endothelial cell (EC) survival during angiogenesis have not been completely characterized. Here, we show that an antisense oligonucleotide to the apoptosis inhibitor survivin suppressed de novo expression of survivin in ECs by vascular endothelial cell growth factor (VEGF). In contrast, the survivin antisense oligonucleotide did not affect anti-apoptotic bcl-2 levels in endothelium. When assessed in cell death assays, antisense targeting of survivin abolished the anti-apoptotic function of VEGF against tumor necrosis factor-alpha- or ceramide-induced cell death, enhanced caspase-3 activity, promoted the generation of a approximately 17-kd active caspase-3 subunit, and increased cleavage of the caspase substrate, polyADP ribose polymerase. In contrast, the survivin antisense oligonucleotide had no effect on EC viability in the absence of VEGF. Antisense oligonucleotides to platelet-endothelial cell adhesion molecule-1 (PECAM-1, CD31), lymphocyte function-associated molecule-3 (LFA-3, CD58), or intercellular adhesion molecule-1 (ICAM-1, CD54) did not reduce the anti-apoptotic function of VEGF in endothelium. When tested on other angiogenic activities mediated by VEGF, survivin antisense treatment induced rapid regression of three-dimensional vascular capillary networks, but did not affect EC migration/chemotaxis. These data suggest that the anti-apoptotic properties of VEGF during angiogenesis are primarily mediated by the induced expression of survivin in ECS: Manipulation of this pathway may increase EC viability in compensatory angiogenesis or facilitate EC apoptosis and promote vascular regression during tumor angiogenesis.


Assuntos
DNA Antissenso/farmacologia , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/efeitos dos fármacos , Linfocinas/farmacologia , Proteínas Associadas aos Microtúbulos , Proteínas/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , DNA/efeitos dos fármacos , DNA/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Apoptose , Proteínas de Neoplasias , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Survivina , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
12.
Lab Invest ; 81(3): 327-34, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11310826

RESUMO

Interleukin-11 (IL-11) reduces injury both in vivo and in vitro, but the mechanisms are unknown. Stimulation of serum- and growth factor-deprived HUVEC with IL-11 increased survivin mRNA and protein expression levels in a dose-dependent manner, with maximal induction at 50 to 100 ng/ml of IL-11. Survivin mRNA expression peaked after 3 to 6 hours of IL-11 treatment and decreased by 24 hours. Survivin protein expression was maximal at 6 hours of treatment and remained elevated through 24 hours. Survivin induction may be mediated by activation of protein kinase B/Akt, but IL-11 failed to activate this pathway in HUVEC. IL-11 did activate signal transducer and activator of transcription (STAT)-3 and IL-11 failed to induce survivin expression in HUVEC transduced with a dominant-negative STAT3 mutant, whereas control-transduced HUVEC responded normally. An IL-11 transgene caused increased survivin mRNA expression in mice compared with control littermates. Intradermal injection of IL-11 (500 ng) into human skin xenografts on immunodeficient mice up-regulated survivin protein in microvascular endothelium and epithelial keratinocytes. We conclude that IL-11 induces expression of survivin, an antiapoptotic protein, in vitro and in vivo, and identify STAT3 as a critical mediator of this response.


Assuntos
Endotélio Vascular/metabolismo , Interleucina-11/metabolismo , Proteínas Associadas aos Microtúbulos , Proteínas/genética , Transdução de Sinais/imunologia , Transcrição Gênica/imunologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Expressão Gênica/imunologia , Humanos , Proteínas Inibidoras de Apoptose , Interleucina-11/imunologia , Proteínas de Neoplasias , Fosforilação , Proteínas/imunologia , Proteínas/metabolismo , RNA Mensageiro/análise , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Serina/metabolismo , Survivina , Transativadores/genética , Transativadores/metabolismo , Transgenes , Veias Umbilicais/citologia
14.
J Immunol ; 166(6): 3846-54, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11238628

RESUMO

We have previously shown that human endothelial cells (EC) are less efficient than professional APC, e.g., B lymphoblastoid cells (BLC), at stimulating allogeneic CD8(+) T cells to develop into CTL. In this study we describe FACS-based limiting dilution analyses using the dilution of the intracellular dye CFSE as an indicator of CD8(+) T cell alloactivation and expansion with significantly increased sensitivity compared with conventional, cytotoxicity-based assays. In addition, this assay permits the relative size of clonal CTL populations that are generated in individual CD8(+) T cell cultures to be determined (clonal burst size). We have applied this method to quantitatively compare the generation of CTL at the clonal level following stimulation of allogeneic CD8(+) T cells by either BLC or HUVEC derived from the same donor. CD8(+) T cells expanded by allostimulation were identified as CD8(+), CFSE(low) cells and were categorized as CTL by the expression of intracellular perforin and IFN-gamma. Precursor frequencies for EC-stimulated CTL were 5- to 40-fold (mean, 7.5-fold) lower compared with BLC-stimulated CTL (p < 0.01). Concomitantly, the average clonal burst sizes in EC-stimulated CTL cultures were significantly smaller than those in conventional CTL cultures, primarily due to the occurrence of some very large clone sizes exclusively with BLC stimulation. Although EC-stimulated CTL were generated only from the memory subset of CD8(+) T cells, BLC-stimulated very large burst sizes of CTL were observed from both naive and memory CD8(+) T cell precursors. These data establish that both a lower frequency of reactive precursors and more limited clonal expansion, but not regulatory T cells, contribute to the reduced capacity of EC to promote alloreactive CTL differentiation compared with that of professional APC.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Testes Imunológicos de Citotoxicidade/métodos , Endotélio Vascular/imunologia , Fluoresceínas/metabolismo , Ativação Linfocitária , Células-Tronco/imunologia , Succinimidas/metabolismo , Subpopulações de Linfócitos T/imunologia , Antígenos CD/biossíntese , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Divisão Celular/imunologia , Linhagem Celular Transformada , Células Cultivadas , Células Clonais , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias/métodos , Endotélio Vascular/citologia , Citometria de Fluxo/métodos , Corantes Fluorescentes/metabolismo , Humanos , Memória Imunológica , Interfase/imunologia , Isoantígenos/imunologia , Contagem de Linfócitos , Células-Tronco/citologia , Células-Tronco/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
15.
Infect Immun ; 69(3): 1889-94, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179369

RESUMO

Hemolytic uremic syndrome (HUS) is associated with intestinal infection by enterohemorrhagic Escherichia coli strains that produce Shiga toxins. Globotriaosylceramide (Gb3) is the functional receptor for Shiga toxin, and tumor necrosis factor alpha (TNF-alpha) upregulates Gb3 in both human macrovascular umbilical vein endothelial cells and human microvascular brain endothelial cells. TNF-alpha treatment enhanced Shiga toxin binding and sensitivity to toxin. This upregulation was specific for Gb3 species containing normal fatty acids (NFA). Central nervous system (CNS) pathology in HUS could involve cytokine-stimulated elevation of endothelial NFA-Gb3 levels. Differential expression of Gb3 species may be a critical determinant of Shiga toxin toxicity and of CNS involvement in HUS.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Toxina Shiga/farmacologia , Triexosilceramidas/biossíntese , Fator de Necrose Tumoral alfa/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos
16.
J Biol Chem ; 276(11): 8341-9, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11112773

RESUMO

Tumor necrosis factor (TNF) receptor-associated factor (TRAF) 2 is an intracellular adapter protein, which, upon TNF stimulation, is directly recruited to the intracellular region of TNF receptor 2 (TNFR2) or indirectly, via TRADD, to the intracellular region of TNF receptor 1 (TNFR1). In cultured human umbilical vein endothelial cells, endogenous TRAF2 colocalizes with the membrane-organizing protein caveolin-1 at regions of enrichment subjacent to the plasma membrane as detected by confocal fluorescence microscopy. Both endogenous and transfected TRAF2 protein coimmunoprecipitate with caveolin-1 in the absence of ligand. Upon TNF treatment, the TRAF2-caveolin-1 complex transiently associates with TRADD, and upon overexpression of TNFR2, the TRAF2-caveolin-1 complex stably associates with and causes redistribution of this receptor as detected by confocal fluorescence microscopy. In human embryonic kidney 293 cells, which have minimal endogenous expression of caveolin-1, cotransfection of TRAF2 and caveolin-1 results in spontaneous association of these proteins which can further associate with and redistribute transfected TNFR2 molecules. The association of caveolin-1 with TNFR2 depends upon TRAF2. Cotransfection of caveolin-1 protein increases TRAF2 protein expression levels in HEK 293 cells, which correlates with enhancement of TNF and TRAF2 signaling, measured as transcription of a NF-kappaB promoter-reporter gene, although the caveolin-enhanced response to TNF is attenuated at higher caveolin levels. These findings suggest that intracellular distribution of activated TNF receptors may be regulated by caveolin-1 via its interaction with TRAF2.


Assuntos
Antígenos CD/metabolismo , Caveolinas/metabolismo , Proteínas/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Caveolina 1 , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , NF-kappa B/metabolismo , Proteínas/genética , Receptores Tipo II do Fator de Necrose Tumoral , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF , Transfecção
17.
Science ; 289(5484): 1550-4, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10968790

RESUMO

Activation of the transcription factor nuclear factor (NF)-kappaB by proinflammatory stimuli leads to increased expression of genes involved in inflammation. Activation of NF-kappaB requires the activity of an inhibitor of kappaB (IkappaB)-kinase (IKK) complex containing two kinases (IKKalpha and IKKbeta) and the regulatory protein NEMO (NF-kappaB essential modifier). An amino-terminal alpha-helical region of NEMO associated with a carboxyl-terminal segment of IKKalpha and IKKbeta that we term the NEMO-binding domain (NBD). A cell-permeable NBD peptide blocked association of NEMO with the IKK complex and inhibited cytokine-induced NF-kappaB activation and NF-kappaB-dependent gene expression. The peptide also ameliorated inflammatory responses in two experimental mouse models of acute inflammation. The NBD provides a target for the development of drugs that would block proinflammatory activation of the IKK complex without inhibiting basal NF-kappaB activity.


Assuntos
NF-kappa B/metabolismo , Peptídeos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Células COS , Células Cultivadas , Selectina E/biossíntese , Selectina E/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Quinase I-kappa B , Inflamação/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Peptídeos/química , Mutação Puntual , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo
18.
Lab Invest ; 80(8): 1185-94, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10950109

RESUMO

TNF signals are mediated through two different receptors, TNFR1 and TNFR2. In endothelial cells, TNFR1 is predominantly localized in the Golgi apparatus and TNFR2 on the plasma membrane. To investigate structural features responsible for the disparate localization, endothelial cells were transfected with epitope-tagged or green fluorescent protein-fused wild type and mutant receptor molecules. Wild type receptors recapitulated the distribution of endogenous receptors. Deletions of the entire TNFR1 intracellular domain or of the C-terminal death domain (TNFR1(-DD)) allowed expression of the receptor on the plasma membrane. However, addition of the death domain to the C-terminus of TNFR2 (TNFR2(+DD)) did not lead to Golgi-retention of this chimeric receptor. Overexpressed TNFR1, TNFR2, and TNFR2(+DD) increased basal expression of a cotransfected NF-kappaB-dependent promotor-reporter gene. Overexpressed TNFR1(-DD) did not activate NF-kappaB but acted as a ligand-specific dominant negative inhibitor of TNF actions. Unexpectedly, TNF responses were also inhibited by overexpressed TNFR1 and TNFR2(+DD), but not TNFR2. We conclude that the death domain of TNFR1 is required for retention of TNFR1 in the Golgi apparatus but is not sufficient to direct Golgi retention of a TNFR2(+DD) chimera, and that overexpressed receptors that contain the death domain (TNFR1 and TNFR2(+DD)) spontaneously activate NF-kappaB while inhibiting TNF responses.


Assuntos
Antígenos CD/metabolismo , Complexo de Golgi/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Antígenos CD/química , Antígenos CD/genética , Células Cultivadas , Humanos , Camundongos , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Transdução de Sinais , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
19.
J Immunol ; 164(12): 6601-9, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10843720

RESUMO

TNF activates endothelial cells to express cell surface molecules that are necessary to recruit a local infiltrate of leukocytes. Because the actions of this proinflammatory cytokine are not species restricted, we investigated whether human TNF can up-regulate porcine endothelial adhesion molecules to elicit human T cell infiltration and damage of pig skin xenografts in a chimeric immunodeficient mouse model. We have previously demonstrated the vigorous rejection of human skin allografts and the absence of injury to porcine skin xenografts in human PBMC-SCID/beige mice. Intradermal administration of human TNF at high doses (600 or 2000 ng) caused nonspecific inflammatory damage of pig skin grafts, whereas low concentrations of TNF (60 or 200 ng) resulted in human PBMC-dependent injury of porcine endothelial cells. There was a strong correlation among pig skin xenograft damage, human T cell infiltration, and the TNF-induced up-regulation of swine MHC class I and class II molecules, VCAM-1, and, in particular, the de novo expression of porcine E-selectin. The microvascular damage and leukocytic infiltration elicited by TNF were enhanced by porcine IFN-gamma, suggesting that xenografts may be less prone to cytokine-mediated injury due to the species-restricted effects of recipient IFN-gamma. Our results indicate that maintenance of a quiescent endothelium, which does not express E-selectin or other activation-dependent adhesion molecules, is important in preventing human anti-porcine T cell xenoresponses in vivo and that TNF signaling molecules and TNF-responsive gene products are appropriate therapeutic targets to protect against human T cell-mediated rejection of pig xenografts.


Assuntos
Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Transplante de Pele/imunologia , Transplante de Pele/patologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/toxicidade , Transferência Adotiva , Adulto , Animais , Moléculas de Adesão Celular/biossíntese , Relação Dose-Resposta Imunológica , Sinergismo Farmacológico , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/fisiopatologia , Antígenos de Histocompatibilidade/biossíntese , Humanos , Interferon gama/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microcirculação/imunologia , Microcirculação/patologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/fisiopatologia , Transplante de Pele/efeitos adversos , Suínos , Linfócitos T/transplante , Transplante Heterólogo/efeitos adversos , Regulação para Cima/imunologia
20.
J Immunol ; 164(9): 4665-71, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10779771

RESUMO

Graft endothelial cells are primary targets of host CTL-mediated injury in acute allograft rejection. As an in vitro trial of gene therapy to reduce CTL-mediated endothelial injury, we stably transduced early passage HUVEC with a caspase-resistant mutant form (D34A) of the anti-apoptotic gene Bcl-2. Bcl-2 transductants were compared with HUVEC transduced in parallel with an enhanced green fluorescent protein (EGFP) gene. Both transduced HUVEC have equivalent growth rates in complete medium and both show contact inhibition of growth. However, compared with EGFP-transduced HUVEC, the Bcl-2-transduced cells are resistant to the apoptotic effects of serum and growth factor withdrawal and are also resistant to the induction of apoptosis by staurosporine or by ceramide, with or without TNF. Transduced Bcl-2 did not reduce TNF-mediated NF-kappaB activation or constitutive expression of class I MHC molecules. HUVEC expressing D34A Bcl-2 were significantly more resistant to lysis by either class I-restricted alloreactive or PHA-redirected CTL than were HUVEC expressing EGFP. We conclude that transduction of graft endothelial cells with D34A Bcl-2 is a possible approach for reducing allograft rejection.


Assuntos
Apoptose/imunologia , Caspases/fisiologia , Citotoxicidade Imunológica , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Linfócitos T Citotóxicos/imunologia , Apoptose/efeitos dos fármacos , Divisão Celular/imunologia , Linhagem Celular Transformada , Células Cultivadas , Meios de Cultivo Condicionados , Fatores de Crescimento Endotelial/farmacologia , Vetores Genéticos/imunologia , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Retroviridae/genética , Transdução Genética/imunologia , Transfecção , Veias Umbilicais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA