Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nat Immunol ; 12(8): 770-7, 2011 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-21685908

RESUMO

How the pore-forming protein perforin delivers apoptosis-inducing granzymes to the cytosol of target cells is uncertain. Perforin induces a transient Ca2+ flux in the target cell, which triggers a process to repair the damaged cell membrane. As a consequence, both perforin and granzymes are endocytosed into enlarged endosomes called 'gigantosomes'. Here we show that perforin formed pores in the gigantosome membrane, allowing endosomal cargo, including granzymes, to be gradually released. After about 15 min, gigantosomes ruptured, releasing their remaining content. Thus, perforin delivers granzymes by a two-step process that involves first transient pores in the cell membrane that trigger the endocytosis of granzyme and perforin and then pore formation in endosomes to trigger cytosolic release.


Assuntos
Endocitose/imunologia , Endossomos/imunologia , Granzimas/imunologia , Proteínas Citotóxicas Formadoras de Poros/imunologia , Cloreto de Amônio/farmacologia , Animais , Apoptose/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Citosol/imunologia , Citosol/metabolismo , Endossomos/metabolismo , Citometria de Fluxo , Granzimas/metabolismo , Células HeLa , Humanos , Células Matadoras Naturais , Macrolídeos/farmacologia , Microscopia Confocal , Microscopia de Vídeo , Proteínas Citotóxicas Formadoras de Poros/antagonistas & inibidores , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Ratos
2.
Am J Pathol ; 186(1): 87-100, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26610869

RESUMO

Cardiac fibrosis is observed across diverse etiologies of heart failure. Granzyme B (GzmB) is a serine protease involved in cell-mediated cytotoxicity in conjunction with the pore-forming protein, perforin. Recent evidence suggests that GzmB also contributes to matrix remodeling and fibrosis through an extracellular, perforin-independent process. However, the role of GzmB in the onset and progression of cardiac fibrosis remains elusive. The present study investigated the role of GzmB in the pathogenesis of cardiac fibrosis. GzmB was elevated in fibrotic human hearts and in angiotensin II-induced murine cardiac fibrosis. Genetic deficiency of GzmB reduced angiotensin II-induced cardiac hypertrophy and fibrosis, independently of perforin. GzmB deficiency also reduced microhemorrhage, inflammation, and fibroblast accumulation in vivo. In vitro, GzmB cleaved the endothelial junction protein, vascular endothelial (VE)-cadherin, resulting in the disruption of endothelial barrier function. Together, these results suggest a perforin-independent, extracellular role for GzmB in the pathogenesis of cardiac fibrosis.


Assuntos
Granzimas/metabolismo , Cardiopatias/enzimologia , Cardiopatias/patologia , Adulto , Idoso , Animais , Modelos Animais de Doenças , Feminino , Fibrose/enzimologia , Fibrose/patologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Adulto Jovem
3.
Proc Natl Acad Sci U S A ; 110(43): 17450-5, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24101526

RESUMO

Recent studies demonstrated that autophagy is an important regulator of innate immune response. However, the mechanism by which autophagy regulates natural killer (NK) cell-mediated antitumor immune responses remains elusive. Here, we demonstrate that hypoxia impairs breast cancer cell susceptibility to NK-mediated lysis in vitro via the activation of autophagy. This impairment was not related to a defect in target cell recognition by NK cells but to the degradation of NK-derived granzyme B in autophagosomes of hypoxic cells. Inhibition of autophagy by targeting beclin1 (BECN1) restored granzyme B levels in hypoxic cells in vitro and induced tumor regression in vivo by facilitating NK-mediated tumor cell killing. Together, our data highlight autophagy as a mechanism underlying the resistance of hypoxic tumor cells to NK-mediated lysis. The work presented here provides a cutting-edge advance in our understanding of the mechanism by which hypoxia-induced autophagy impairs NK-mediated lysis in vitro and paves the way for the formulation of more effective NK cell-based antitumor therapies.


Assuntos
Autofagia/imunologia , Citotoxicidade Imunológica/imunologia , Granzimas/imunologia , Células Matadoras Naturais/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Hipóxia Celular/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Citometria de Fluxo , Granzimas/metabolismo , Humanos , Immunoblotting , Células Matadoras Naturais/metabolismo , Células MCF-7 , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Confocal , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fagossomos/imunologia , Fagossomos/metabolismo , Imagem com Lapso de Tempo/métodos , Transplante Heterólogo , Carga Tumoral/imunologia
4.
J Neuroinflammation ; 12: 157, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26337722

RESUMO

BACKGROUND: Multiple sclerosis (MS) is an autoimmune inflammatory and neurodegenerative disease of the central nervous system (CNS). It is widely accepted that inflammatory cells play major roles in the pathogenesis of MS, possibly through the use of serine protease granzyme B (GrB) secreted from the granules of cytotoxic T cells. We have previously identified GrB as a mediator of axonal injury and neuronal death. In this study, our goal was to evaluate the effect of GrB inhibition in the human system in vitro, and in vivo in EAE using the newly isolated GrB-inhibitor serpina3n. METHODS: We used a well-established in vitro model of neuroinflammation characterized by a co-culture system between human fetal neurons and lymphocytes. In vivo, we induced EAE in 10- to 12-week-old female C57/BL6 mice and treated them intravenously with serpina3n. RESULTS: In the in vitro co-culture system, pre-treatment of lymphocytes with serpina3n prevented neuronal killing and cleavage of the cytoskeletal protein alpha-tubulin, a known substrate for GrB. Moreover, in EAE, 50 µg serpina3n substantially reduced the severity of the disease. This dose was administered intravenously twice at days 7 and 20 post EAE induction. serpina3n treatment reduced axonal and neuronal injury compared to the vehicle-treated control group and maintained the integrity of myelin. Interestingly, serpina3n treatment did not seem to reduce the infiltration of immune cells (CD4(+) and CD8(+) T cells) into the CNS. CONCLUSION: Our data suggest further studies on serpina3n as a potentially novel therapeutic strategy for the treatment of inflammatory-mediated neurodegenerative diseases such as MS.


Assuntos
Proteínas de Fase Aguda/uso terapêutico , Encefalomielite Autoimune Experimental/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Serpinas/uso terapêutico , Animais , Antígenos CD/metabolismo , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/induzido quimicamente , Feminino , Feto , Adjuvante de Freund/toxicidade , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Glicoproteína Mielina-Oligodendrócito/toxicidade , Proteínas de Neurofilamentos/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Linfócitos T/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
5.
Nat Rev Immunol ; 2(6): 401-9, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12093006

RESUMO

Cytotoxic T lymphocytes (CTLs) provide potent defences against virus infection and intracellular pathogens. However, CTLs have a dark side--their lytic machinery can be directed against self-tissues in autoimmune disorders, transplanted cells during graft rejection and host tissues to cause graft-versus-host disease, which is one of the most serious diseases related to CTL function. Although this duplicitous behaviour might seem contradictory, both beneficial and detrimental effects are the result of the same effector proteins. So, an understanding of the mechanisms that are used by CTLs to destroy targets and a knowledge of pathogen immune-evasion strategies will provide vital information for the design of new therapies.


Assuntos
Linfócitos T Citotóxicos/imunologia , Animais , Autoimunidade , Morte Celular/imunologia , Citotoxicidade Imunológica , Rejeição de Enxerto , Doença Enxerto-Hospedeiro/imunologia , Granzimas , Humanos , Células Matadoras Naturais/imunologia , Camundongos , Modelos Imunológicos , Ratos , Serina Endopeptidases/imunologia , Linfócitos T Citotóxicos/enzimologia , Vírus/imunologia
6.
J Virol ; 86(4): 2229-38, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22130540

RESUMO

During the 2009 H1N1 influenza virus pandemic (pdmH1N1) outbreak, it was found that most individuals lacked antibodies against the new pdmH1N1 virus, and only the elderly showed anti-hemagglutinin (anti-HA) antibodies that were cross-reactive with the new strains. Different studies have demonstrated that prior contact with the virus can confer protection against strains with some degree of dissimilarity; however, this has not been sufficiently explored within the context of a pdmH1N1 virus infection. In this study, we have found that a first infection with the A/Brisbane/59/2007 virus strain confers heterologous protection in ferrets and mice against a subsequent pdmH1N1 (A/Mexico/4108/2009) virus infection through a cross-reactive but non-neutralizing antibody mechanism. Heterologous immunity is abrogated in B cell-deficient mice but maintained in CD8(-/-) and perforin-1(-/-) mice. We identified cross-reactive antibodies from A/Brisbane/59/2007 sera that recognize non-HA epitopes in pdmH1N1 virus. Passive serum transfer showed that cross-reactive sH1N1-induced antibodies conferred protection in naive recipient mice during pdmH1N1 virus challenge. The presence or absence of anti-HA antibodies, therefore, is not the sole indicator of the effectiveness of protective cross-reactive antibody immunity. Measurement of additional antibody repertoires targeting the non-HA antigens of influenza virus should be taken into consideration in assessing protection and immunization strategies. We propose that preexisting cross-protective non-HA antibody immunity may have had an overall protective effect during the 2009 pdmH1N1 outbreak, thereby reducing disease severity in human infections.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Antígenos CD8/imunologia , Proteção Cruzada , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Embrião de Galinha , Feminino , Furões , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/epidemiologia , Influenza Humana/virologia , Masculino , México/epidemiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pandemias
7.
PLoS Pathog ; 7(12): e1002447, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22194691

RESUMO

Cytotoxic T lymphocytes (CTLs) are the major killer of virus-infected cells. Granzyme B (GrB) from CTLs induces apoptosis in target cells by cleavage and activation of substrates like caspase-3 and Bid. However, while undergoing apoptosis, cells are still capable of producing infectious viruses unless a mechanism exists to specifically inhibit viral production. Using proteomic approaches, we identified a novel GrB target that plays a major role in protein synthesis: eukaryotic initiation factor 4 gamma 3 (eIF4G3). We hypothesized a novel role for GrB in translation of viral proteins by targeting eIF4G3, and showed that GrB cleaves eIF4G3 specifically at the IESD(1408)S sequence. Both GrB and human CTL treatment resulted in degradation of eIF4G3 and reduced rates of translation. When Jurkat cells infected with vaccinia virus were treated with GrB, there was a halt in viral protein synthesis and a decrease in production of infectious new virions. The GrB-induced inhibition of viral translation was independent of the activation of caspases, as inhibition of protein synthesis still occurred with addition of the pan-caspase inhibitor zVAD-fmk. This demonstrated for the first time that GrB prevents the production of infectious vaccinia virus by targeting the host translational machinery.


Assuntos
Fator de Iniciação Eucariótico 4G/metabolismo , Granzimas/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Linfócitos T Citotóxicos/metabolismo , Vaccinia virus/metabolismo , Replicação Viral , Apoptose/fisiologia , Caspase 3/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Células Jurkat , Proteólise , Proteômica , Vaccinia virus/fisiologia , Replicação Viral/fisiologia
8.
J Immunol ; 187(9): 4861-72, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21964027

RESUMO

Multiple sclerosis (MS) is considered an autoimmune disease of the CNS and is characterized by inflammatory cells infiltrating the CNS and inducing demyelination, axonal loss, and neuronal death. Recent evidence strongly suggests that axonal and neuronal degeneration underlie the progression of permanent disability in MS. In this study, we report that human neurons are selectively susceptible to the serine-protease granzyme B (GrB) isolated from cytotoxic T cell granules. In vitro, purified human GrB induced neuronal death to the same extent as the whole activated T cell population. On the contrary, activated T cells isolated from GrB knockout mice failed to induce neuronal injury. We found that following internalization through various parts of neurons, GrB accumulated in the neuronal soma. Within the cell body, GrB diffused out of endosomes possibly through a perforin-independent mechanism and induced subsequent activation of caspases and cleavage of α-tubulin. Inhibition of caspase-3, a well-known substrate for GrB, significantly reduced GrB-mediated neurotoxicity. We demonstrated that treatment of neurons with mannose-6-phosphate prevented GrB entry and inhibited GrB-mediated neuronal death, suggesting mannose-6-phosphate receptor-dependent endocytosis. Together, our data unveil a novel mechanism by which GrB induces selective neuronal injury and suggest potential new targets for the treatment of inflammatory-mediated neurodegeneration in diseases such as MS.


Assuntos
Grânulos Citoplasmáticos/enzimologia , Grânulos Citoplasmáticos/imunologia , Granzimas/fisiologia , Ativação Linfocitária/imunologia , Neurônios/enzimologia , Neurônios/imunologia , Linfócitos T Citotóxicos/imunologia , Adulto , Animais , Encéfalo/embriologia , Encéfalo/enzimologia , Encéfalo/imunologia , Morte Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Grânulos Citoplasmáticos/metabolismo , Testes Imunológicos de Citotoxicidade/métodos , Granzimas/metabolismo , Granzimas/toxicidade , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Neurônios/citologia , Linfócitos T Citotóxicos/enzimologia , Linfócitos T Citotóxicos/metabolismo
9.
Exp Cell Res ; 317(4): 531-8, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21134367

RESUMO

LL-37 is a human cationic host defense peptide (antimicrobial peptide) belonging to the cathelicidin family of peptides. In this study, LL-37 was shown to kill stimulated CD8(+) T cells (Cytotoxic T lymphocytes; CTLs) via apoptosis, while having no cytotoxic effect on non-stimulated CD8(+) or CD4(+) T cells or stimulated CD4(+) T cells. Of interest, the CD8(+) cells were much more sensitive to LL-37 than many other cell types. LL-37 exposure resulted in DNA fragmentation, chromatin condensation, and the release of both granzyme A and granzyme B from intracellular granules. The importance of granzyme family members in the apoptosis of CTLs following LL-37 treatment was analyzed by using C57BL/6 lymphocytes obtained from mice that were homozygous for null mutations in the granzyme B gene, the granzyme A gene, or both granzymes A and B. Granzymes A and B were both shown to play an important role in LL-37-induced apoptosis of CTLs. Further analysis revealed that apoptosis occurred primarily through granzyme A-mediated caspase-independent apoptosis. However, caspase-dependent cell death was also observed. This suggests that LL-37 induces apoptosis in CTLs via multiple different mechanisms, initiated by the LL-37-induced leakage of granzymes from cytolytic granules. Our results imply the existence of a novel mechanism of crosstalk between the inflammatory and adaptive immune systems. Cells such as neutrophils, at the site of a tumor for example, could influence the effector, activity of CTL through the secretion of LL-37.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Apoptose/efeitos dos fármacos , Granzimas/fisiologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Imunidade Adaptativa , Animais , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Receptor Cross-Talk , Catelicidinas
10.
J Exp Med ; 196(9): 1127-39, 2002 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-12417624

RESUMO

M11L, an antiapoptotic protein essential for the virulence of the myxoma poxvirus, is targeted to mitochondria and prevents the loss of mitochondrial membrane potential that accompanies cell death. In this study we show, using a cross-linking approach, that M11L physically associates with the mitochondrial peripheral benzodiazepine receptor (PBR) component of the permeability transition (PT) pore. Close association of M11L and the PBR is also indicated by fluorescence resonance energy transfer (FRET) analysis. Stable expression of M11L prevents the release of mitochondrial cytochrome c induced by staurosporine or protoporphyrin IX (PPIX), a ligand of the PBR. Transiently expressed M11L also prevents mitochondrial membrane potential loss induced by PPIX, or induced by staurosporine in combination with PK11195, another ligand of the PBR. Myxoma virus infection and the associated expression of early proteins, including M11L, protects cells from staurosporine- and Fas-mediated mitochondrial membrane potential loss and this effect is augmented by the presence of PBR. We conclude that M11L regulates the mitochondrial permeability transition pore complex, most likely by direct modulation of the PBR.


Assuntos
Apoptose , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Myxoma virus/metabolismo , Receptores de GABA-A/metabolismo , Proteínas Virais/metabolismo , Animais , Células COS , Chlorocebus aethiops , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Humanos , Células Jurkat , Potenciais da Membrana , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Myxoma virus/genética , Myxoma virus/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Protoporfirinas/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/genética
11.
Mol Cancer Res ; 7(5): 689-702, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19435812

RESUMO

LL-37 is a human cationic host defense peptide (antimicrobial peptide) belonging to the cathelicidin family of peptides. In this study, LL-37 was shown to kill Jurkat T leukemia cells via apoptosis. A loss of mitochondrial membrane potential, DNA fragmentation, and phosphatidylserine externalization were detected following LL-37 exposure, whereas apoptosis was independent of caspase family members. The specific apoptotic pathway induced by LL-37 was defined through the utilization of Jurkat cells modified to express antiapoptotic proteins, as well as cells deficient in various proteins associated with apoptosis. Of interest, both Bcl-2-overexpressing cells and cells deficient in Bax and Bak proteins displayed a significant reduction in LL-37-induced apoptosis. In addition, Jurkat cells modified in the Fas receptor-associated pathway showed no reduction in apoptosis when exposed to LL-37. Analysis of the involvement of apoptosis-inducing factor (AIF) in LL-37-mediated apoptosis revealed that AIF transferred from the mitochondria to the nucleus of cells exposed to LL-37, where it may lead to large-scale DNA fragmentation and chromatin condensation. AIF knockdown analysis resulted in LL-37-resistant cells. This suggests that AIF is mandatory in LL-37-mediated killing. Lastly, chelation or inhibition of Ca(2+) or calpains inhibited LL-37-mediated killing. Further analysis revealed that calpains were required for LL-37-mediated Bax translocation to mitochondria. Together, these data show that LL-37-induced apoptosis is mediated via the mitochondria-associated pathway in a caspase-independent and calpain- and AIF-dependent manner that involves Bax activation and translocation to mitochondria.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Fator de Indução de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Calpaína/metabolismo , Proteína X Associada a bcl-2/metabolismo , Sequência de Aminoácidos , Peptídeos Catiônicos Antimicrobianos/síntese química , Fator de Indução de Apoptose/genética , Cálcio/metabolismo , Calpaína/antagonistas & inibidores , Caspase 9/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Células Jurkat , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Oligopeptídeos/farmacologia , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Catelicidinas
12.
Eur J Immunol ; 39(2): 552-60, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19130477

RESUMO

The NK immunological synapse (NKIS) is a dynamic structure dependent on the assembly of membrane, cytoskeletal and signaling components. These serve to focus and generate stimuli for adhesion and orientation of the cytoskeleton for targeted cytolytic granule release. Previous studies have demonstrated the importance of the cytoskeleton in these processes. We previously identified PPP1R9B (neurabin 2, spinophilin) as a cytoskeletal component of the NK-like cell line YTS. We demonstrate that (i) PPP1R9B gradually accumulates at the NKIS in a maturation stage-dependent manner; (ii) it mimics the early kinetics of actin recruitment to the NKIS but it precedes actin departure from the site; (iii) it is recruited by CD18 stimulation but not by CD28 ligation; (iv) it is required for the maintenance of the cortical F-actin organization in the YTS cells and knocking down PPP1R9B reduces the frequency of YTS-target cell conjugation, possibly due to the collapsed F-actin cytoskeleton in these cells. These results indicate that PPP1R9B is required for synapse formation in the NK cells and suggest that it may be involved in the maintenance of cellular architecture by regulation of actin assembly, possibly acting to stabilize the NKIS until granule release is eminent.


Assuntos
Actinas/imunologia , Sinapses Imunológicas/imunologia , Células Matadoras Naturais/imunologia , Proteínas dos Microfilamentos/imunologia , Proteínas do Tecido Nervoso/imunologia , Actinas/metabolismo , Antígenos CD18/imunologia , Antígenos CD18/metabolismo , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Linhagem Celular Tumoral , Citoesqueleto/imunologia , Citoesqueleto/metabolismo , Humanos , Sinapses Imunológicas/metabolismo , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo
13.
Mol Biol Cell ; 17(2): 623-33, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16280358

RESUMO

During granule-mediated killing by cytotoxic T lymphocytes or natural killer cells, the serine protease granzyme B enters the target cell by endocytosis and induces apoptosis. Previous studies suggested a role for the mannose 6-phosphate receptor, but further experiments with purified granzyme B indicated this was not essential. Additionally, it is now clear that grB is exocytosed from killer cells in a high-molecular-weight complex with the proteoglycan serglycin. Here granzyme B was delivered as a purified monomer, or in complex with either glycosaminoglycans or serglycin, and killing was evaluated. When granzyme B was a monomer, soluble mannose 6-phosphate had a limited impact, whereas apoptosis induced by the complexed grB was effectively inhibited by mannose 6-phosphate. Most importantly, when granzyme B and perforin were delivered together from granules, inhibition by mannose 6-phosphate was also observed. In pulldown assays mediated by the cation-independent mannose 6-phosphate receptor, granzyme B bound to the receptor more intensely in the presence of immobilized heparan sulfate. We therefore propose the model that under physiological conditions serglycin-bound granzyme B is critically endocytosed by a mannose 6-phosphate receptor, and receptor binding is enhanced by cell surface heparan sulfate.


Assuntos
Heparitina Sulfato/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/fisiologia , Receptor IGF Tipo 2/fisiologia , Vesículas Secretórias/fisiologia , Serina Endopeptidases/fisiologia , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose , Linhagem Celular , Glicosaminoglicanos/metabolismo , Granzimas , Heparitina Sulfato/química , Humanos , Células Jurkat , Camundongos , Modelos Biológicos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Proteoglicanas/fisiologia , Vesículas Secretórias/enzimologia , Linfócitos T Citotóxicos/enzimologia , Proteínas de Transporte Vesicular/fisiologia
14.
Biochem Biophys Res Commun ; 368(2): 357-63, 2008 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-18222115

RESUMO

The granzyme B gene is activated upon cytotoxic T cell stimulation and the protein is a key inducer of apoptosis in target cells. Previous studies have identified important proximal regulatory regions but these proved insufficient to drive expression in vivo. We identified a DNase1 hypersensitive site (HS2) 3.9kb upstream of the transcription start site that was present in stimulated but not resting CD8+ cells. The CTL line CTLL R8 was stably transfected with GFP reporter constructs and showed consistently higher fluorescence values when HS2 was included. In transgenic mice the presence of the relevant region of DNA resulted in inducible, CTL-specific transcription of the transgene in all transgenic founder lines analyzed. Deletion of HS2 resulted in a 10-fold reduction in expression. This is the first report of a major distal regulatory element in the control of granzyme B transcription.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Regulação Enzimológica da Expressão Gênica/genética , Granzimas/genética , Camundongos Transgênicos/genética , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico/genética , Ativação Transcricional/genética , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
15.
Biochem Biophys Res Commun ; 366(3): 617-23, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18042464

RESUMO

Natural killer cells recognize and induce apoptosis in foreign, transformed or virus-infected cells through the release of perforin and granzymes from secretory lysosomes. Clinically, NK-cell mediated killing is a major limitation to successful allo- and xenotransplantation. The molecular mechanisms that regulate the fusion of granzyme B-containing secretory lysosomes to the plasma membrane in activated NK cells, prior to target cell killing, are not fully understood. Using the NK cell line YT-Indy as a model, we have investigated the expression of SNAP REceptors (SNAREs), both target (t-) and vesicular (v-) SNAREs, and their function in granzyme B-mediated target cell killing. Our data showed that YT-Indy cells express VAMP-7 and SNAP-23, but not VAMP-2. VAMP-7 was associated with granzyme B-containing lysosomal granules. Using VAMP-7 small interfering RNA (siRNA), we successfully knocked down the expression of VAMP-7 protein in YT-Indy to less than 10% of untreated cells in 24h. VAMP7-deficient YT-Indy cells activated via co-culture with Jurkat cells released <1ng/mL of granzyme B, compared to 1.5-2.5 microg/mL from controls. Using Jurkat cells as targets, we showed a 7-fold reduction in NK cell-mediated killing by VAMP-7 deficient YT-Indy cells. Our results show that VAMP-7 is a crucial component of granzyme B release and target cell killing in the NK cell line YT-Indy. Thus, targeting VAMP-7 expression specifically with siRNA, following transplantation, may be a viable strategy for preventing NK cell-mediated transplant rejection, in vivo.


Assuntos
Apoptose/fisiologia , Granzimas/metabolismo , Células Matadoras Naturais/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Linhagem Celular , Humanos , Células Jurkat
16.
Cell Transplant ; 16(10): 1029-38, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18351019

RESUMO

Sertoli cells (SC) protect islet allografts from immune destruction in diabetic rodents. In this study, we examined the difference between successful and rejected islet/SC cografts in order to further improve this procedure for optimal extension of islet allograft survival. We cotransplanted 500 BALB/c islets with 1-8 million BALB/c SC under the kidney capsule of diabetic BALB/c, C3H-HeJ, and C57BL/6 mice. Cotransplantation of islets with up to 8 million SC was not detrimental to long-term islet graft function in syngeneic mice. However, large numbers of SC were detrimental to islet graft survival in allogeneic mice with the optimal dose for cotransplantation of 4 or 1 million SC in C3H-HeJ or C57BL/6 mice, respectively. Examination of successful grafts, from euglycemic recipients, revealed the presence of SC arranged in tubule structures with islets surrounding these tubules. Cellular infiltrate in successful grafts revealed CD4 T cells and macrophages along the periphery and within the grafts, and very few CD8 T cells. Conversely, examination of unsuccessful grafts, harvested from hyperglycemic recipients at the time of rejection, revealed the presence of SC arranged randomly with islets adjacent to the Sertoli cells, when present, and massive CD4 and CD8 T cell as well as macrophage cell infiltration. Prolongation of islet allograft survival appeared to be a function of SC transplant mass and recipient genetic background. A consequence of long-term graft acceptance is the formation of SC tubule structures, which may be an additional requirement for optimal protection of islet allografts.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Sobrevivência de Enxerto , Transplante das Ilhotas Pancreáticas , Células de Sertoli/transplante , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Estreptozocina , Transplante Homólogo
17.
Sci Rep ; 7(1): 15857, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-29158532

RESUMO

Decorin (DCN) is a small-leucine rich proteoglycan that mediates collagen fibrillogenesis, organization, and tensile strength. Adventitial DCN is reduced in abdominal aortic aneurysm (AAA) resulting in vessel wall instability thereby predisposing the vessel to rupture. Recombinant DCN fusion protein CAR-DCN was engineered with an extended C-terminus comprised of CAR homing peptide that recognizes inflamed blood vessels and penetrates deep into the vessel wall. In the present study, the role of systemically-administered CAR-DCN in AAA progression and rupture was assessed in a murine model. Apolipoprotein E knockout (ApoE-KO) mice were infused with angiotensin II (AngII) for 28 days to induce AAA formation. CAR-DCN or vehicle was administrated systemically until day 15. Mortality due to AAA rupture was significantly reduced in CAR-DCN-treated mice compared to controls. Although the prevalence of AAA was similar between vehicle and CAR-DCN groups, the severity of AAA in the CAR-DCN group was significantly reduced. Histological analysis revealed that CAR-DCN treatment significantly increased DCN and collagen levels within the aortic wall as compared to vehicle controls. Taken together, these results suggest that CAR-DCN treatment attenuates the formation and rupture of Ang II-induced AAA in mice by reinforcing the aortic wall.


Assuntos
Aneurisma da Aorta Abdominal/genética , Apolipoproteínas E/genética , Decorina/genética , Proteínas Recombinantes/genética , Angiotensina II/genética , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/fisiopatologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Proteínas Recombinantes/administração & dosagem
18.
J Immunol Methods ; 308(1-2): 156-66, 2006 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-16375915

RESUMO

We have established novel ELISA- and ELISPOT-based assays specific for the detection of a potent cytotoxic mediator, granzyme B (GrB), for the assessment of antigen-specific cytotoxic T lymphocyte responses in mice. The sensitivity and specificity of our assays was demonstrated by ELISA using purified mouse GrB and supernatants and cell lysates of cytotoxic lymphocytes derived from GrB-deficient mice. No reactivity was observed by the GrB ELISA in GrB-deficient cells. The mouse GrB ELISPOT was successfully employed to detect antigen-specific effector cell responses of two CTL clones, producing GrB ELISPOT results that correlated strongly with target cell lysis, as assessed by 51Cr-release. Furthermore, we were able to demonstrate direct correlations between GrB ELISPOT and killing by LCMV gp33-specific effector and memory T cells generated in vivo. Thus, the mouse GrB ELISPOT may be used to detect cytotoxic responses, at the single-cell level, for the functional assessment of antigen-specific CD8+ T cell responses in mouse models of infection.


Assuntos
Testes Imunológicos de Citotoxicidade/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Serina Endopeptidases/análise , Linfócitos T Citotóxicos/enzimologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos Virais , Degranulação Celular , Testes Imunológicos de Citotoxicidade/estatística & dados numéricos , Ensaio de Imunoadsorção Enzimática/estatística & dados numéricos , Feminino , Granzimas , Memória Imunológica , Técnicas In Vitro , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Sensibilidade e Especificidade , Serina Endopeptidases/biossíntese
19.
Arterioscler Thromb Vasc Biol ; 24(12): 2245-50, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15472125

RESUMO

OBJECTIVE: T cell-induced cytotoxicity, of which granzyme B is a key mediator, is believed to contribute to the pathogenesis of inflammatory vascular diseases. In this report, we investigate the mechanism of granzyme B-induced smooth muscle cell (SMC) death. METHODS AND RESULTS: The addition of purified granzyme B alone to cultured SMCs caused a significant reduction in cell viability. Chromatin condensation, phosphatidylserine externalization, and membrane blebbing were observed, indicating that the mechanism of granzyme B-induced SMC death was through apoptosis. Activated splenocytes from perforin-knockout mice induced SMC death through a granzyme B-mediated pathway. Inhibition of the proteolytic activities of caspases and granzyme B prevented granzyme B-induced SMC death, whereas attenuation of granzyme B internalization with mannose-6-phosphate (M6P) did not. Further, granzyme B induced the cleavage of several SMC extracellular proteins, including fibronectin, and reduced focal adhesion kinase phosphorylation. CONCLUSIONS: These results indicate that granzyme B can induce apoptosis of SMCs in the absence of perforin by cleaving extracellular proteins, such as fibronectin.


Assuntos
Apoptose/fisiologia , Matriz Extracelular/metabolismo , Glicoproteínas de Membrana/deficiência , Miócitos de Músculo Liso/fisiologia , Serina Endopeptidases/fisiologia , Animais , Caspase 3 , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Vasos Coronários/enzimologia , Vasos Coronários/metabolismo , Matriz Extracelular/enzimologia , Proteínas da Matriz Extracelular/metabolismo , Granzimas , Células HeLa/química , Células HeLa/metabolismo , Humanos , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/enzimologia , Perforina , Proteínas Citotóxicas Formadoras de Poros , Ratos , Serina Endopeptidases/metabolismo , Baço/citologia , Baço/metabolismo
20.
Protein Eng Des Sel ; 28(1): 9-17, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25492933

RESUMO

The intracellular roles of Granzyme B (GrB) in immune-mediated cell killing have been extensively studied. Recent data also implicate GrB in extracellular pathways of inflammation, cytokine activation and autoimmunity. Targeting (GrB) provides a new pharmaceutical agent for various inflammatory disorders. Serpina3n is a mouse extracellular inhibitor of GrB. There is no apparent equivalent in humans. In this study, we used a novel applied genetics approach to engineer a new extracellular GrB serpin. A chimeric protein was generated in which the reactive center loop (RCL) of human extracellular antichymotrypsin (ACT) was replaced with that of serpina3n. This serpin contained 27 amino acid residues from the serpina3n RCL and the remaining 395 residues from human ACT. The insertion converted human ACT into a GrB-inhibitory serpin. Several critical residues were identified by scanning mutagenesis on the chimera and serpina3n. Targeted mutagenesis was conducted on wild-type human ACT by specifically substituting those critical residues, creating a novel inhibitor that contains 99.3% human ACT sequence with only three point mutations. Wild-type human ACT had a kass for GrB of 2.26 × 10(4) M(-1) s(-1), whereas the novel inhibitor binds GrB with a kass of 7.65 × 10(5) M(-1) s(-1). This new drug candidate can be developed in animal models and further tested in clinical trials to help us understand the role of GrB in numerous disorders.


Assuntos
Proteínas de Fase Aguda/metabolismo , Granzimas/antagonistas & inibidores , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Inibidores de Serina Proteinase/metabolismo , Serpinas/metabolismo , Proteínas de Fase Aguda/química , Proteínas de Fase Aguda/genética , Animais , Apoptose , Humanos , Células Jurkat , Camundongos , Mutação , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/genética , Serpinas/química , Serpinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA