Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(37): e2306965120, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37669366

RESUMO

Fibrosis is regulated by interactions between immune and mesenchymal cells. However, the capacity of cell types to modulate human fibrosis pathology is poorly understood due to lack of a fully humanized model system. MISTRG6 mice were engineered by homologous mouse/human gene replacement to develop an immune system like humans when engrafted with human hematopoietic stem cells (HSCs). We utilized MISTRG6 mice to model scleroderma by transplantation of healthy or scleroderma skin from a patient with pansclerotic morphea to humanized mice engrafted with unmatched allogeneic HSC. We identified that scleroderma skin grafts contained both skin and bone marrow-derived human CD4 and CD8 T cells along with human endothelial cells and pericytes. Unlike healthy skin, fibroblasts in scleroderma skin were depleted and replaced by mouse fibroblasts. Furthermore, HSC engraftment alleviated multiple signatures of fibrosis, including expression of collagen and interferon genes, and proliferation and activation of human T cells. Fibrosis improvement correlated with reduced markers of T cell activation and expression of human IL-6 by mesenchymal cells. Mechanistic studies supported a model whereby IL-6 trans-signaling driven by CD4 T cell-derived soluble IL-6 receptor complexed with fibroblast-derived IL-6 promoted excess extracellular matrix gene expression. Thus, MISTRG6 mice transplanted with scleroderma skin demonstrated multiple fibrotic responses centered around human IL-6 signaling, which was improved by the presence of healthy bone marrow-derived immune cells. Our results highlight the importance of IL-6 trans-signaling in pathogenesis of scleroderma and the ability of healthy bone marrow-derived immune cells to mitigate disease.


Assuntos
Basidiomycota , Esclerodermia Localizada , Humanos , Animais , Camundongos , Interleucina-6 , Células Endoteliais , Pele , Modelos Animais de Doenças
2.
Nat Commun ; 10(1): 2247, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113953

RESUMO

Complement promotes vascular inflammation in transplant organ rejection and connective tissue diseases. Here we identify ZFYVE21 as a complement-induced Rab5 effector that induces non-canonical NF-κB in endothelial cells (EC). In response to membrane attack complexes (MAC), ZFYVE21 is post-translationally stabilized on MAC+Rab5+ endosomes in a Rab5- and PI(3)P-dependent manner. ZFYVE21 promotes SMURF2-mediated polyubiquitinylation and proteasome-dependent degradation of endosome-associated PTEN to induce vesicular enrichment of PI(3,4,5)P3 and sequential recruitment of activated Akt and NF-κB-inducing kinase (NIK). Pharmacologic alteration of cellular phosphoinositide content with miltefosine reduces ZFYVE21 induction, EC activation, and allograft vasculopathy in a humanized mouse model. ZFYVE21 induction distinctly occurs in response to MAC and is detected in human renal and synovial tissues. Our data identifies ZFYVE21 as a Rab5 effector, defines a Rab5-ZFYVE21-SMURF2-pAkt axis by which it mediates EC activation, and demonstrates a role for this pathway in complement-mediated conditions.


Assuntos
Proteínas de Transporte/metabolismo , Endossomos/metabolismo , Rejeição de Enxerto/patologia , NF-kappa B/metabolismo , Vasculite/patologia , Aloenxertos/patologia , Animais , Linhagem Celular , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Vasos Coronários/patologia , Vasos Coronários/transplante , Modelos Animais de Doenças , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Camundongos , Camundongos SCID , Fosfatos de Fosfatidilinositol/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo
3.
J Immunol ; 197(6): 2400-8, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27534549

RESUMO

A classical hallmark of acute inflammation is neutrophil infiltration of tissues, a multistep process that involves sequential cell-cell interactions of circulating leukocytes with IL-1- or TNF-activated microvascular endothelial cells (ECs) and pericytes (PCs) that form the wall of the postcapillary venules. The initial infiltrating cells accumulate perivascularly in close proximity to PCs. IL-17, a proinflammatory cytokine that acts on target cells via a heterodimeric receptor formed by IL-17RA and IL-17RC subunits, also promotes neutrophilic inflammation but its effects on vascular cells are less clear. We report that both cultured human ECs and PCs strongly express IL-17RC and, although neither cell type expresses much IL-17RA, PCs express significantly more than ECs. IL-17, alone or synergistically with TNF, significantly alters inflammatory gene expression in cultured human PCs but not ECs. RNA sequencing analysis identifies many IL-17-induced transcripts in PCs encoding proteins known to stimulate neutrophil-mediated immunity. Conditioned media from IL-17-activated PCs, but not ECs, induce pertussis toxin-sensitive neutrophil polarization, likely mediated by PC-secreted chemokines, and they also stimulate neutrophil production of proinflammatory molecules, including TNF, IL-1α, IL-1ß, and IL-8. Furthermore, IL-17-activated PCs, but not ECs, can prolong neutrophil survival by producing G-CSF and GM-CSF, delaying the mitochondrial outer membrane permeabilization and caspase-9 activation. Importantly, neutrophils exhibit enhanced phagocytic capacity after activation by conditioned media from IL-17-treated PCs. We conclude that PCs, not ECs, are the major target of IL-17 within the microvessel wall and that IL-17-activated PCs can modulate neutrophil functions within the perivascular tissue space.


Assuntos
Endotélio Vascular/fisiologia , Interleucina-17/imunologia , Neutrófilos/imunologia , Pericitos/fisiologia , Receptores de Interleucina-17/imunologia , Caspase 9/metabolismo , Células Cultivadas , Meios de Cultura , Citocinas/biossíntese , Citocinas/imunologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Fator Estimulador de Colônias de Granulócitos/biossíntese , Fator Estimulador de Colônias de Granulócitos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-17/genética , Interleucina-17/farmacologia , Infiltração de Neutrófilos , Neutrófilos/fisiologia , Pericitos/efeitos dos fármacos , Pericitos/imunologia , Receptores de Interleucina-17/fisiologia , Análise de Sequência de RNA , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Vênulas/citologia , Vênulas/imunologia
4.
Proc Natl Acad Sci U S A ; 112(31): 9686-91, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26195760

RESUMO

Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Endossomos/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Proteína 3 com Repetições IAP de Baculovírus , Clatrina/metabolismo , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/metabolismo , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Estabilidade Enzimática/efeitos dos fármacos , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hidrazonas/farmacologia , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos SCID , Biossíntese de Proteínas/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 3 Associado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Quinase Induzida por NF-kappaB
5.
J Exp Med ; 211(3): 395-404, 2014 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-24516119

RESUMO

Recruitment of circulating leukocytes into inflamed tissues depends on adhesion molecules expressed by endothelial cells (ECs). Here we report that rapamycin pretreatment reduced the ability of TNF-treated ECs to capture T cells under conditions of venular flow. This functional change was caused by inhibition of TNF-induced expression of vascular cell adhesion molecule-1 (VCAM-1) and could be mimicked by knockdown of mammalian target of rapamycin (mTOR) or rictor, but not raptor, implicating mTORC2 as the target of rapamycin for this effect. Mechanistically, mTORC2 acts through Akt to repress Raf1-MEK1/2-ERK1/2 signaling, and inhibition of mTORC2 consequently results in hyperactivation of ERK1/2. Increased ERK1/2 activity antagonizes VCAM-1 expression by repressing TNF induction of the transcription factor IRF-1. Preventing activation of ERK1/2 reduced the ability of rapamycin to inhibit TNF-induced VCAM-1 expression. In vivo, rapamycin inhibited mTORC2 activity and potentiated activation of ERK1/2. These changes correlated with reduced endothelial expression of TNF-induced VCAM-1, which was restored via pharmacological inhibition of ERK1/2. Functionally, rapamycin reduced infiltration of leukocytes into renal glomeruli, an effect which was partially reversed by inhibition of ERK1/2. These data demonstrate a novel mechanism by which rapamycin modulates the ability of vascular endothelium to mediate inflammation and identifies endothelial mTORC2 as a potential therapeutic target.


Assuntos
Células Endoteliais/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Molécula 1 de Adesão de Célula Vascular/metabolismo , Análise de Variância , Western Blotting , Adesão Celular/imunologia , Imunoprecipitação da Cromatina , Primers do DNA/genética , Citometria de Fluxo , Humanos , Immunoblotting , Alvo Mecanístico do Complexo 2 de Rapamicina , Microscopia de Fluorescência , Complexos Multiproteicos/metabolismo , Proteína Oncogênica v-akt/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T/imunologia , Serina-Treonina Quinases TOR/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 32(2): 353-60, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22053072

RESUMO

OBJECTIVE: Perioperative nonimmune injuries to an allograft can decrease graft survival. We have developed a model for studying this process using human materials. METHODS AND RESULTS: Human artery segments were transplanted as infrarenal aortic interposition grafts into an immunodeficient mouse host, allowed to "heal in" for 30 days, and then retransplanted into a second mouse host. To induce a reperfusion injury, the healed-in artery segments were incubated for 3 hours under hypoxic conditions ex vivo before retransplantation. To induce immunologic rejection, the animals receiving the retransplanted artery segment were adoptively transferred with human peripheral blood mononuclear cells or purified T cells from a donor allogeneic to the artery 1 week before surgery. To compare rejection of injured versus healthy tissues, these manipulations were combined. Results were analyzed ex vivo by histology, morphometry, immunohistochemistry, and mRNA quantitation or in vivo by ultrasound. Our results showed that reperfusion injury, which otherwise heals with minimal sequelae, intensifies the degree of allogeneic T cell-mediated injury to human artery segments. CONCLUSIONS: We developed a new human-mouse chimeric model demonstrating interactions of reperfusion injury and alloimmunity using human cells and tissues that may be adapted to study other forms of nonimmune injury and other types of adaptive immune responses.


Assuntos
Imunidade Adaptativa/fisiologia , Artérias/imunologia , Artérias/transplante , Quimera/imunologia , Traumatismo por Reperfusão/fisiopatologia , Linfócitos T/imunologia , Adulto , Animais , Artérias/patologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/fisiopatologia , Sobrevivência de Enxerto/imunologia , Sobrevivência de Enxerto/fisiologia , Humanos , Camundongos , Camundongos SCID , Modelos Animais , Linfócitos T/patologia , Transplante Homólogo
7.
J Biol Chem ; 284(29): 19331-9, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19473970

RESUMO

Microvascular endothelial cell (EC) expression of tumor necrosis factor receptor (TNFR) 2 is induced in situ by ischemia/reperfusion injury. To assess effects of molecular oxygen on TNFR2 expression, we subjected cultured human dermal microvascular ECs (HDMECs) to hypoxic conditions (1% O(2)) or to hypoxic conditions followed by return to normoxic conditions. TNFR2 mRNA and protein are expressed under normoxic conditions but are rapidly reduced by hypoxia; they fall even further upon reoxygenation but rebound by 6-9 h. TNFR1 expression is unaffected by hypoxia or reoxygenation in these same cells. We identified a potential FOXO3a binding site in the 5' enhancer region of the TNFR2 gene. FOXO3a from normoxic but not hypoxic HDMECs binds an oligonucleotide sequence matching this site, and the endogenous enhancer binds FOXO3a at this site in HDMECs under normoxic but not hypoxic conditions. Unphosphorylated FOXO3a is present in the nucleus of HDMECs under normoxic conditions. Hypoxia leads to FOXO3a phosphorylation at an Akt/protein kinase B target site and subsequent nuclear export; these processes are reversed by reoxygenation and blocked by LY294002, a phosphatidylinositol 3-kinase inhibitor that blocks Akt activation. LY294002 also prevents the hypoxia-mediated decrease in TNFR2 expression. Transiently transfected FOXO3a activates a TNFR2 promoter/reporter construct in HDMECs, whereas small interference RNA knockdown of FOXO3a reduces TNFR2 but not TNFR1 expression under normoxic conditions. Reduction in TNFR2 by small interference RNA sensitizes HDMECs to TNFR1-mediated apoptosis. We conclude that FOXO3a regulates oxygen-dependent changes in expression of TNFR2 in HDMECs, controlling sensitivity to TNF-mediated apoptosis.


Assuntos
Células Endoteliais/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Oxigênio/farmacologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Apoptose/efeitos dos fármacos , Sequência de Bases , Hipóxia Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Cromonas/farmacologia , Cicloeximida/farmacologia , Derme/citologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Immunoblotting , Microscopia de Fluorescência , Dados de Sequência Molecular , Morfolinas/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , RNA Interferente Pequeno/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
8.
Transplantation ; 87(2): 189-97, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19155972

RESUMO

BACKGROUND: Nearly half of all infiltrating leukocytes in rejecting human allografts are macrophages, yet, in comparison with T cells, much less is known about the contribution of this cell type to rejection. Our laboratory has previously described models of rejection of human skin or artery grafts in immunodeficient mouse hosts mediated by adoptively transferred allogeneic T cells. However, mature human monocyte/macrophages have consistently failed to engraft in these animals. Here, we describe the introduction of human CD68+ macrophages into irradiated immunodeficient mice by transplantation of enriched CD34+ hematopoietic stem-cells isolated from peripheral blood of G-colony-stimulating factor pretreated adults. METHODS: We investigated strains of immunodeficient mice bearing human tissue grafts (skin and artery) inoculated with 1 x 10(6) human CD34+ adult hematopoietic stem cells, peripheral blood monuclear cells autologous to the CD34 donor, or both for human cell engraftment. RESULTS: In the absence of T cells, CD68+ CD14+ macrophages infiltrate allogeneic human skin but produce little injury or thrombosis. Both responses are enhanced when combined with adoptive transfer of T cells autologous to the hematopoietic stem cells as exemplified by the induction of the macrophage activation marker CD163. CD68+ macrophages also infiltrate allogeneic arterial interposition grafts, producing intimal expansion and calcification in the absence of T cells. CONCLUSIONS: These new models may be used to study the role of human macrophages in transplant rejection and other pathologies in vivo.


Assuntos
Células-Tronco Adultas/imunologia , Artérias/imunologia , Artérias/transplante , Rejeição de Enxerto/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Macrófagos/imunologia , Transplante de Pele/imunologia , Pele/imunologia , Transferência Adotiva , Animais , Antígenos CD/análise , Antígenos CD34/análise , Antígenos de Diferenciação Mielomonocítica/análise , Artérias/patologia , Modelos Animais de Doenças , Rejeição de Enxerto/patologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Receptores de Lipopolissacarídeos/análise , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Receptores de Superfície Celular/análise , Pele/patologia , Linfócitos T/imunologia , Linfócitos T/transplante , Transplante Homólogo , Irradiação Corporal Total
9.
J Exp Med ; 205(13): 3145-58, 2008 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19075290

RESUMO

Interleukin (IL) 1alpha produced by human endothelial cells (ECs), in response to tumor necrosis factor (TNF) or to co-culture with allogeneic T cells in a TNF-dependent manner, can augment the release of cytokines from alloreactive memory T cells in vitro. In a human-mouse chimeric model of artery allograft rejection, ECs lining the transplanted human arteries express IL-1alpha, and blocking IL-1 reduces the extent of human T cell infiltration into the artery intima and selectively inhibits IL-17 production by infiltrating T cells. In human skin grafts implanted on immunodeficient mice, administration of IL-17 is sufficient to induce mild inflammation. In cultured cells, IL-17 acts preferentially on vascular smooth muscle cells rather than ECs to enhance production of proinflammatory mediators, including IL-6, CXCL8, and CCL20. Neutralization of IL-17 does not reduce T cell infiltration into allogeneic human artery grafts, but markedly reduces IL-6, CXCL8, and CCL20 expression and selectively inhibits CCR6(+) T cell accumulation in rejecting arteries. We conclude that graft-derived IL-1 can promote T cell intimal recruitment and IL-17 production during human artery allograft rejection, and suggest that targeting IL-1 in the perioperative transplant period may modulate host alloreactivity.


Assuntos
Artérias/transplante , Linfócitos T CD4-Positivos/imunologia , Rejeição de Enxerto/imunologia , Interleucina-17/imunologia , Interleucina-1alfa/imunologia , Transplante Homólogo/imunologia , Túnica Íntima/imunologia , Animais , Artérias/anatomia & histologia , Artérias/imunologia , Linfócitos T CD4-Positivos/citologia , Células Cultivadas , Quimiocinas/imunologia , Citocinas/imunologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Humanos , Inflamação/imunologia , Ativação Linfocitária , Camundongos , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Receptores CCR6/genética , Receptores CCR6/imunologia , Transplante de Pele , Túnica Íntima/citologia
10.
J Immunol ; 172(3): 1391-6, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734714

RESUMO

IL-11 can reduce tissue injury in animal models of inflammation but the mechanism(s) is unknown. When C.B-17 SCID/beige mice bearing human skin grafts are injected i.p. with human PBMC allogeneic to the donor skin, infiltrating T cells destroy human microvessels by day 21. Intradermal injection of human IL-11 (500 ng/day) delays the time course of graft microvessel loss without reducing the extent of T cell infiltration. Protective actions of IL-11 are most pronounced on day 15. IL-11 has no effect on T cell activation marker, effector molecule, cytokine expression, or endothelial ICAM-1 expression. IL-11 up-regulates the expression of survivin, a cytoprotective protein, in graft keratinocytes and endothelial cells. Topical application of survivin antisense oligonucleotide down-regulates survivin expression in both cell types and largely abrogates the protective effect of IL-11. We conclude that in this human transplant model, IL-11 exerts a cytoprotective rather than anti-inflammatory or immunomodulatory effect mediated through induction of survivin.


Assuntos
Citoproteção/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Interleucina-11/administração & dosagem , Proteínas Associadas aos Microtúbulos/biossíntese , Transplante de Pele/patologia , Pele/irrigação sanguínea , Pele/patologia , Adjuvantes Imunológicos/administração & dosagem , Administração Tópica , Adulto , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Citoproteção/genética , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Endotélio Vascular/imunologia , Humanos , Proteínas Inibidoras de Apoptose , Injeções Intradérmicas , Interleucina-11/antagonistas & inibidores , Interleucina-11/uso terapêutico , Leucócitos Mononucleares/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Microcirculação/imunologia , Microcirculação/metabolismo , Microcirculação/patologia , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas de Neoplasias , Pomadas , Oligonucleotídeos Antissenso/administração & dosagem , Pele/imunologia , Pele/metabolismo , Transplante de Pele/efeitos adversos , Transplante de Pele/imunologia , Survivina , Transplante Homólogo/efeitos adversos , Transplante Homólogo/imunologia , Transplante Homólogo/patologia
11.
J Immunol ; 171(3): 1526-33, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12874246

RESUMO

Human TRAIL can efficiently kill tumor cells in vitro and kill human tumor xenografts in mice with little effect on normal mouse cells or tissues. The effects of TRAIL on normal human tissues have not been described. In this study, we report that endothelial cells (EC), isolated from human umbilical veins or human dermal microvessels, express death domain-containing TRAIL-R1 and -R2. Incubation with TRAIL for 15 h causes approximately 30% of cultured EC to die, as assessed by propidium iodide uptake. Death is apoptotic, as assessed by Annexin V staining, 4',6'-diamidino-2-phenylindole staining, and DNA fragment ELISA. EC death is increased by cotreatment with cycloheximide but significantly reduced by caspase inhibitors or transduced dominant-negative Fas-associated death domain protein. In surviving cells, TRAIL activates NF-kappaB, induces expression of E-selectin, ICAM-1, and IL-8, and promotes adhesion of leukocytes. Injection of TRAIL into human skin xenografts promotes focal EC injury accompanied by limited neutrophil infiltration. These data suggest that TRAIL is an inducer of tissue injury in humans, an outcome that may influence antitumor therapy with TRAIL.


Assuntos
Apoptose/fisiologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Regulação da Expressão Gênica/fisiologia , Mediadores da Inflamação/metabolismo , Glicoproteínas de Membrana/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Linhagem Celular , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Células HL-60 , Células HeLa , Humanos , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/farmacologia , Injeções Intradérmicas , Leucócitos Mononucleares/patologia , Glicoproteínas de Membrana/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Infiltração de Neutrófilos/fisiologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/biossíntese , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Transplante de Pele/patologia , Ligante Indutor de Apoptose Relacionado a TNF , Transplante Heterólogo/patologia , Fator de Necrose Tumoral alfa/administração & dosagem
12.
J Biol Chem ; 278(27): 25014-23, 2003 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-12724316

RESUMO

Oncostatin M (OnM) signals through cell surface receptors, which utilize the gp130 subunit. In cultured human umbilical vein endothelial cells (HUVEC), OnM transiently elevates mRNA encoding for suppressor of cytokine signaling-3 (SOCS-3). By 1 h of OnM treatment, HUVEC become refractory to the restimulation by OnM, measured as failure to reinduce SOCS-3 mRNA. OnM-induced desensitization also prevents responses to other gp130-signaling cytokines (e.g. leukemia inhibitory factor and interleukin 11). OnM treatment does not affect gp130 expression levels and desensitizes signaling mediated by a transduced chimeric receptor containing extracellular domains of platelet-derived growth factor receptor-beta (PDGFRbeta) and the cytoplasmic region of gp130. Interestingly, a chimeric PDGFRbeta-gp130 mutant receptor, in which intracellular Tyr residue 759 of gp130 is replaced by a Phe residue, mediates prolonged signaling and is not cross-desensitized by OnM. Phospho-Tyr759 is the binding site for both SOCS-3 and for Src homology domain 2-containing tyrosine phosphatase 2 (SHP-2). In human aortic smooth muscle cells, neither prevention of SOCS-3 protein induction, using STAT3 or SOCS-3 antisense, nor prevention of SHP-2 expression, also with antisense, ablates desensitization. These data suggest that desensitization of vascular cells to OnM is mediated in trans and involves Tyr residue 759 in gp130 but is not mediated by either SOCS-3 or SHP-2, the only two proteins currently known to bind to gp130 at this site.


Assuntos
Antígenos CD/metabolismo , Endotélio Vascular/metabolismo , Glicoproteínas de Membrana/metabolismo , Peptídeos/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Repressoras , Fatores de Transcrição , Animais , Antígenos CD/genética , Células Cultivadas , Receptor gp130 de Citocina , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/genética , Camundongos , Oncostatina M , Proteína Fosfatase 2 , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas/metabolismo , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Tirosina , Domínios de Homologia de src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA