Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(12): e0262198, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34972191

RESUMO

BACKGROUND: Colorectal cancer (CRC) is a leading cause of cancer-related deaths, with a 5% 5-year survival rate for metastatic disease, yet with limited therapeutic advancements due to insufficient understanding of and inability to accurately capture high-risk CRC patients who are most likely to recur. We aimed to improve high-risk classification by identifying biological pathways associated with outcome in adjuvant stage II/III CRC. METHODS AND FINDINGS: We included 1062 patients with stage III or high-risk stage II colon carcinoma from the prospective three-arm randomized phase 3 AVANT trial, and performed expression profiling to identify a prognostic signature. Data from validation cohort GSE39582, The Cancer Genome Atlas, and cell lines were used to further validate the prognostic biology. Our retrospective analysis of the adjuvant AVANT trial uncovered a prognostic signature capturing three biological functions-stromal, proliferative and immune-that outperformed the Consensus Molecular Subtypes (CMS) and recurrence prediction signatures like Oncotype Dx in an independent cohort. Importantly, within the immune component, high granzyme B (GZMB) expression had a significant prognostic impact while other individual T-effector genes were less or not prognostic. In addition, we found GZMB to be endogenously expressed in CMS2 tumor cells and to be prognostic in a T cell independent fashion. A limitation of our study is that these results, although robust and derived from a large dataset, still need to be clinically validated in a prospective study. CONCLUSIONS: This work furthers our understanding of the underlying biology that propagates stage II/III CRC disease progression and provides scientific rationale for future high-risk stratification and targeted treatment evaluation in biomarker defined subpopulations of resectable high-risk CRC. Our results also shed light on an alternative GZMB source with context-specific implications on the disease's unique biology.


Assuntos
Neoplasias Colorretais/metabolismo , Granzimas/fisiologia , Transcriptoma , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Análise por Conglomerados , Neoplasias Colorretais/mortalidade , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Granzimas/química , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , Estudos Prospectivos , Estudos Retrospectivos , Risco , Linfócitos T/metabolismo , Resultado do Tratamento
2.
Front Immunol ; 12: 712678, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34413857

RESUMO

Mycobacterium tuberculosis (Mtb), the pathological agent that causes tuberculosis (TB) is the number one infectious killer worldwide with one fourth of the world's population currently infected. Data indicate that γ9δ2 T cells secrete Granzyme A (GzmA) in the extracellular space triggering the infected monocyte to inhibit growth of intracellular mycobacteria. Accordingly, deletion of GZMA from γ9δ2 T cells reverses their inhibitory capacity. Through mechanistic studies, GzmA's action was investigated in monocytes from human PBMCs. The use of recombinant human GzmA expressed in a mammalian system induced inhibition of intracellular mycobacteria to the same degree as previous human native protein findings. Our data indicate that: 1) GzmA is internalized within mycobacteria-infected cells, suggesting that GzmA uptake could prevent infection and 2) that the active site is not required to inhibit intracellular replication. Global proteomic analysis demonstrated that the ER stress response and ATP producing proteins were upregulated after GzmA treatment, and these proteins abundancies were confirmed by examining their expression in an independent set of patient samples. Our data suggest that immunotherapeutic host interventions of these pathways may contribute to better control of the current TB epidemic.


Assuntos
Trifosfato de Adenosina/biossíntese , Estresse do Retículo Endoplasmático/imunologia , Granzimas/fisiologia , Monócitos/microbiologia , Mycobacterium bovis/fisiologia , Subpopulações de Linfócitos T/imunologia , Western Blotting , Divisão Celular , Granzimas/biossíntese , Granzimas/genética , Granzimas/farmacologia , Células HEK293 , Humanos , Células T de Memória/imunologia , Células T de Memória/metabolismo , Proteoma , Receptores de Antígenos de Linfócitos T gama-delta/análise , Proteínas Recombinantes/farmacologia , Subpopulações de Linfócitos T/metabolismo , Eletroforese em Gel Diferencial Bidimensional
3.
Cell Signal ; 76: 109804, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33035645

RESUMO

Chronic inflammation and impaired wound healing play important roles in the pathophysiology of cardiovascular diseases. Moreover, the aberrant secretion of proteases plays a critical role in pathological tissue remodeling in chronic inflammatory conditions. Human Granzymes (Granule secreted enzymes - Gzms) comprise a family of five (GzmA, B, H, K, M) cell-secreted serine proteases. Although each unique in function and substrate specificities, Gzms were originally thought to share redundant, intracellular roles in cytotoxic lymphocyte-induced cell death. However, an abundance of evidence has challenged this dogma. It is now recognized, that individual Gzms exhibit unique substrate repertoires and functions both intracellularly and extracellularly. In the extracellular milieu, Gzms contribute to inflammation, vascular dysfunction and permeability, reduced cell adhesion, release of matrix-sequestered growth factors, receptor activation, and extracellular matrix cleavage. Despite these recent findings, the non-cytotoxic functions of Gzms in the context of cardiovascular disease pathogenesis remain poorly understood. Minimally detected in tissues and bodily fluids of normal individuals, GzmB is elevated in patients with acute coronary syndromes, coronary artery disease, and myocardial infarction. Pre-clinical animal models have exemplified the importance of GzmB in atherosclerosis, aortic aneurysm, and cardiac fibrosis as animals deficient in GzmB exhibit reduced tissue remodeling, improved disease phenotypes and increased survival. Although a role for GzmB in cardiovascular disease is described, further work to elucidate the mechanisms that underpin the remaining human Gzms activity in cardiovascular disease is necessary. The present review provides a summary of the pre-clinical and clinical evidence, as well as emerging areas of research pertaining to Gzms in tissue remodeling and cardiovascular disease.


Assuntos
Doenças Cardiovasculares/metabolismo , Granzimas/fisiologia , Animais , Humanos
4.
JCI Insight ; 5(18)2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32809971

RESUMO

Acute graft-versus-host disease (aGVHD) can occur after hematopoietic cell transplant in patients undergoing treatment for hematological malignancies or inborn errors. Although CD4+ T helper (Th) cells play a major role in aGVHD, the mechanisms by which they contribute, particularly within the intestines, have remained elusive. We have identified a potentially novel subset of Th cells that accumulated in the intestines and produced the serine protease granzyme A (GrA). GrA+ Th cells were distinct from other Th lineages and exhibited a noncytolytic phenotype. In vitro, GrA+ Th cells differentiated in the presence of IL-4, IL-6, and IL-21 and were transcriptionally unique from cells cultured with either IL-4 or the IL-6/IL-21 combination alone. In vivo, both STAT3 and STAT6 were required for GrA+ Th cell differentiation and played roles in maintenance of the lineage identity. Importantly, GrA+ Th cells promoted aGVHD-associated morbidity and mortality and contributed to crypt destruction within intestines but were not required for the beneficial graft-versus-leukemia effect. Our data indicate that GrA+ Th cells represent a distinct Th subset and are critical mediators of aGVHD.


Assuntos
Doença Enxerto-Hospedeiro/patologia , Efeito Enxerto vs Leucemia/imunologia , Granzimas/fisiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Intestinos/patologia , Ativação Linfocitária/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Feminino , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/metabolismo , Neoplasias Hematológicas/terapia , Intestinos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT3/fisiologia , Fator de Transcrição STAT6/fisiologia
5.
Diabetes ; 66(12): 3061-3071, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28877912

RESUMO

Events defining the progression to human type 1 diabetes (T1D) have remained elusive owing to the complex interaction between genetics, the immune system, and the environment. Type 1 interferons (T1-IFN) are known to be a constituent of the autoinflammatory milieu within the pancreas of patients with T1D. However, the capacity of IFNα/ß to modulate human activated autoreactive CD8+ T-cell (cytotoxic T lymphocyte) responses within the islets of patients with T1D has not been investigated. Here, we engineer human ß-cell-specific cytotoxic T lymphocytes and demonstrate that T1-IFN augments cytotoxicity by inducing rapid phosphorylation of STAT4, resulting in direct binding at the granzyme B promoter within 2 h of exposure. The current findings provide novel insights concerning the regulation of effector function by T1-IFN in human antigen-experienced CD8+ T cells and provide a mechanism by which the presence of T1-IFN potentiates diabetogenicity within the autoimmune islet.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Granzimas/fisiologia , Interferon Tipo I/farmacologia , Fator de Transcrição STAT4/fisiologia , Adolescente , Adulto , Criança , Feminino , Granzimas/genética , Humanos , Masculino , Regiões Promotoras Genéticas , Adulto Jovem
6.
Nat Commun ; 8(1): 231, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28794449

RESUMO

Group 3 innate lymphoid cells (ILC3) promote lymphoid organogenesis and potentiate immune responses against bacterial infection. However, how ILC3 cells are developed and maintained is still unclear. Here, we show that carboxypeptidase CCP2 is highly expressed in common helper-like innate lymphoid progenitors, the progenitor of innate lymphoid cells, and CCP2 deficiency increases ILC3 numbers. Interleukin-7 receptor subunit alpha (IL-7Rα) is identified as a substrate of CCP2 for deglutamylation, and IL-7Rα polyglutamylation is catalyzed by polyglutamylases TTLL4 and TTLL13 in common helper-like innate lymphoid progenitors. IL-7Rα polyglutamylation triggers STAT5 activation to initiate transcription factor Sall3 expression in common helper-like innate lymphoid progenitors, which drives ILC3 cell differentiation. Moreover, Ttll4 -/- or Ttll13 -/- mice have reduced IL-7Rα polyglutamylation and Sall3 expression in common helper-like innate lymphoid progenitors. Importantly, mice with IL-7Rα E446A mutation have reduced Sall3 expression and ILC3 population. Thus, polyglutamylation and deglutamylation of IL-7Rα tightly controls the development and effector functions of ILC3s.Innate lymphoid cells (ILC) are important regulators of mucosal immunity, but how their development and homeostasis are modulated is still unclear. Here the authors show that the differentiation of group 3 ILCs is controlled by the glutamylation of IL-7Rα and the induction of transcription factor Sall3.


Assuntos
Ácido Glutâmico/metabolismo , Proteínas de Homeodomínio/metabolismo , Imunidade Inata , Linfócitos/imunologia , Receptores de Interleucina-7/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Granzimas/genética , Granzimas/fisiologia , Proteínas de Homeodomínio/genética , Linfócitos/citologia , Camundongos , Peptídeo Sintases/metabolismo , Fatores de Transcrição/genética
7.
Diabetes ; 66(12): 3041-3050, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28733313

RESUMO

Granzyme A is a protease implicated in the degradation of intracellular DNA. Nucleotide complexes are known triggers of systemic autoimmunity, but a role in organ-specific autoimmune disease has not been demonstrated. To investigate whether such a mechanism could be an endogenous trigger for autoimmunity, we examined the impact of granzyme A deficiency in the NOD mouse model of autoimmune diabetes. Granzyme A deficiency resulted in an increased incidence in diabetes associated with accumulation of ssDNA in immune cells and induction of an interferon response in pancreatic islets. Central tolerance to proinsulin in transgenic NOD mice was broken on a granzyme A-deficient background. We have identified a novel endogenous trigger for autoimmune diabetes and an in vivo role for granzyme A in maintaining immune tolerance.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Granzimas/fisiologia , Tolerância Imunológica , Interferon Tipo I/fisiologia , Animais , DNA de Cadeia Simples/metabolismo , Feminino , Granzimas/deficiência , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
9.
Arthritis Rheumatol ; 69(2): 320-334, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27598995

RESUMO

OBJECTIVE: Granzyme A (GzmA) levels are elevated in the plasma and synovium of patients with rheumatoid arthritis (RA), suggesting involvement of this protease in the pathogenesis of the disease. GzmA contributes to sepsis by regulating the production of proinflammatory cytokines. The purpose of this study was to evaluate the contribution of GzmA to the pathogenesis of RA in vivo and to examine the possibility that GzmA acting via tumor necrosis factor (TNF) stimulates osteoclastogenesis. METHODS: Inflammatory arthritis induced by type II collagen was evaluated in wild-type, GzmA-deficient, and perforin-deficient mice. The osteoclastogenic potential of GzmA was examined in vitro using bone marrow cells and colony-forming unit-granulocyte-macrophage (CFU-GM) cells and in vivo using GzmA-deficient mice. RESULTS: Gene deletion of GzmA attenuated collagen-induced arthritis, including serum levels of proinflammatory cytokines, joint damage, and bone erosion in affected mice, suggesting that osteoclast activity is reduced in the absence of GzmA. Accordingly, GzmA-treated bone marrow cells produced multinucleated cells that fulfilled the criteria for mature osteoclasts: tartrate-resistant acid phosphatase (TRAP) activity, ß integrin expression, calcitonin receptor expression, and resorptive activity on dentin slices. GzmA appeared to act without accessory cells, and its activity was not affected by osteoprotegerin, suggesting a minor contribution of RANKL. It also induced the expression and secretion of TNF. Neutralization of TNF or stimulation of CFU-GM cells from TNF-/- mice prevented GzmA-induced osteoclastogenesis. GzmA-deficient mice had reduced osteoclastogenesis in vivo (fewer calcitonin receptor-positive multinucleated cells and fewer transcripts for cathepsin K, matrix metalloproteinase 9, and TRAP in joints) and reduced serum levels of C-terminal telopeptide of type I collagen. CONCLUSION: GzmA contributes to the joint destruction of RA partly by promoting osteoclast differentiation.


Assuntos
Artrite Experimental/enzimologia , Artrite Experimental/etiologia , Artrite Reumatoide/enzimologia , Artrite Reumatoide/etiologia , Granzimas/fisiologia , Osteogênese/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL
11.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L507-16, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27343190

RESUMO

Streptococcus pneumoniae is the most common causative pathogen in community-acquired pneumonia (CAP). Granzyme A (GzmA) is a serine protease produced by a variety of cell types involved in the immune response. We sought to determine the role of GzmA on the host response during pneumococcal pneumonia. GzmA was measured in bronchoalveolar lavage fluid (BALF) harvested from CAP patients from the infected and contralateral uninfected side and in lung tissue slides from CAP patients and controls. In CAP patients, GzmA levels were increased in BALF obtained from the infected lung. Human lungs showed constitutive GzmA expression by both parenchymal and nonparenchymal cells. In an experimental setting, pneumonia was induced in wild-type (WT) and GzmA-deficient (GzmA(-/-)) mice by intranasal inoculation of S. pneumoniae In separate experiments, WT and GzmA(-/-) mice were treated with natural killer (NK) cell depleting antibodies. Upon infection with S. pneumoniae, GzmA(-/-) mice showed a better survival and lower bacterial counts in BALF and distant body sites compared with WT mice. Although NK cells showed strong GzmA expression, NK cell depletion did not influence bacterial loads in either WT or GzmA(-/-) mice. These results implicate that GzmA plays an unfavorable role in host defense during pneumococcal pneumonia by a mechanism that does not depend on NK cells.


Assuntos
Granzimas/fisiologia , Pneumonia Pneumocócica/enzimologia , Streptococcus pneumoniae/imunologia , Animais , Líquido da Lavagem Broncoalveolar , Estudos de Casos e Controles , Feminino , Humanos , Imunidade Celular , Células Matadoras Naturais/fisiologia , Pulmão/enzimologia , Pulmão/imunologia , Pulmão/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Infiltração de Neutrófilos , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia
12.
Artigo em Japonês | MEDLINE | ID: mdl-27212596

RESUMO

Development of acute experimental autoimmune encephalomyelitis (EAE) depends on Th17 cells expressing the nuclear factor NR4A2, which we have previously reported to be upregulated in peripheral blood T cells from patients of multiple sclerosis (MS). EAE induced in mice lacking NR4A2 in T cells showed a great reduction in Th17-mediated acute symptoms, whereas a late-onset disease independent of NR4A2 was still inducible. We identified cytotoxic T-cell-like CD4+ T cells expressing the T-box transcription factor Eomesodermin (Eomes) as a pathogenic component for the development of the late-onset disease. Furthermore, T cell-specific deletion of the Eomes gene or Eomes-specific RNA interference in vivo remarkably ameliorated the late-onset EAE. Intriguingly, similar Eomes-expressing CD4+ T cells are increased in the peripheral blood and cerebrospinal fluid only from patients with secondary-progressive MS accompanied by neurodegenerative symptoms, but not in relapsing-remitting MS. Mechanistic analysis revealed that granzyme B was secreted by Eomes-expressing CD4+ T cells and the activation of protease-activated receptor-1 by granzyme B is involved in the neuroinflammation observed in the late-onset EAE.


Assuntos
Esclerose Múltipla Crônica Progressiva/etiologia , Proteínas com Domínio T , Linfócitos T Auxiliares-Indutores , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Granzimas/fisiologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla Crônica Progressiva/genética , Esclerose Múltipla Crônica Progressiva/patologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Receptor PAR-1/fisiologia , Células Th17
13.
FEBS J ; 283(14): 2577-98, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26648190

RESUMO

This review aims to highlight the proteases required for regulated cell death mechanisms in animals and plants. The aim is to be incisive, and not inclusive of all the animal proteases that have been implicated in various publications. The review also aims to focus on instances when several publications from disparate groups have demonstrated the involvement of an animal protease, and also when there is substantial biochemical, mechanistic and genetic evidence. In doing so, the literature can be culled to a handful of proteases, covering most of the known regulated cell death mechanisms: apoptosis, regulated necrosis, necroptosis, pyroptosis and NETosis in animals. In plants, the literature is younger and not as extensive as for mammals, although the molecular drivers of vacuolar death, necrosis and the hypersensitive response in plants are becoming clearer. Each of these death mechanisms has at least one proteolytic component that plays a major role in controlling the pathway, and sometimes they combine in networks to regulate cell death/survival decision nodes. Some similarities are found among animal and plant cell death proteases but, overall, the pathways that they govern are kingdom-specific with very little overlap.


Assuntos
Morte Celular/fisiologia , Peptídeo Hidrolases/fisiologia , Animais , Apoptose/fisiologia , Caspases/fisiologia , Catálise , Granzimas/fisiologia , Humanos , Modelos Biológicos , Necrose , Peptídeo Hidrolases/classificação , Células Vegetais/enzimologia , Transdução de Sinais/fisiologia
14.
Infect Immun ; 83(2): 702-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25452549

RESUMO

In the livers of susceptible C57BL/6 (B6) mice infected with Leishmania donovani, CD8(+) T cell mechanisms are required for granuloma assembly, macrophage activation, intracellular parasite killing, and self-cure. Since gene expression of perforin and granzymes A and B (GzmA and GzmB), cytolytic proteins linked to CD8(+) cell effector function, was enhanced in infected liver tissue, B6 mice deficient in these granular proteins were used to gauge host defense roles. Neither perforin nor GzmA was required; however, mice deficient in GzmB (GzmB(-/-), GzmB cluster(-/-), and GzmA×B cluster double knockout [DKO] mice) showed both delayed granuloma assembly and initially impaired control of parasite replication. Since these two defects in B6 mice were limited to early-stage infection, innately resistant 129/Sv mice were also tested. In this genetic setting, expression of both innate and subsequent T (Th1) cell-dependent acquired resistance, including the self-cure phenotype, was entirely derailed in GzmA×B cluster DKO mice. These results, in susceptible B6 mice for GzmB and in resistant 129/Sv mice for GzmA and/or the GzmB cluster, point to granzyme-mediated host defense regulation in the liver in experimental visceral leishmaniasis.


Assuntos
Granzimas/fisiologia , Leishmania donovani/imunologia , Leishmaniose Visceral/imunologia , Fígado/imunologia , Macrófagos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Granuloma/metabolismo , Granzimas/genética , Imunidade Inata/genética , Imunidade Inata/imunologia , Inflamação/imunologia , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-10/biossíntese , Interleucina-18/biossíntese , Leishmaniose Visceral/parasitologia , Fígado/parasitologia , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina/biossíntese , Perforina/genética , Células Th1/imunologia
15.
Am J Transplant ; 15(1): 234-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25534448

RESUMO

The clonal expansion of effector T cells and subsequent generation of memory T cells are critical in determining the outcome of transplantation. While cytotoxic T lymphocytes induce direct cytolysis of target cells through secretion of Granzyme-B (GrB), they also express cytoplasmic serine protease inhibitor-6 (Spi6) to protect themselves from GrB that has leaked from granules. Here, we studied the role of GrB/Spi6 axis in determining clonal expansion of alloreactive CD8-T cells and subsequent generation of memory CD8-T cells in transplantation. CD8-T cells from Spi6(-/-) mice underwent more GrB mediated apoptosis upon alloantigen stimulation in vitro and in vivo following adoptive transfer into an allogeneic host. Interestingly, while OT1.Spi6(-/-) CD8 T cells showed significantly lower clonal expansion following skin transplants from OVA mice, there was no difference in the size of the effector memory CD8-T cells long after transplantation. Furthermore, lack of Spi6 resulted in a decrease of short-lived-effector-CD8-cells but did not impact the pool of memory-precursor-effector-CD8-cells. Similar results were found in heart transplant models. Our findings suggest that the final alloreactive CD8-memory-pool-size is independent from the initial clonal-proliferation as memory precursors express low levels of GrB and therefore are independent of Spi6 for survival. These data advance our understanding of memory T cells generation in transplantation and provide basis for Spi6 based strategies to target effector T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Transplante de Coração , Memória Imunológica/imunologia , Proteínas de Membrana/fisiologia , Serpinas/fisiologia , Transplante de Pele , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Sobrevivência Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Doença Enxerto-Hospedeiro/patologia , Granzimas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
16.
Med Hypotheses ; 83(6): 688-93, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25459135

RESUMO

Granzymes (GZMs) are a class of serine protease, found in cytoplasmic granules of cytotoxic T cells and natural killer cells. The main function of these proteins has been recognized as wiping out viral infections via inducing the apoptosis. This review will highlight inter and intra species differences of GZMs in terms of their functional and structure. These futures may help to device a strategy for isolation of human specific GZMs, which are needed for understating of their role in immune system and devising an effective immune therapy.


Assuntos
Granzimas/fisiologia , Animais , Apoptose , Domínio Catalítico , Citoplasma/metabolismo , Exocitose , Genômica , Humanos , Sistema Imunitário , Células Matadoras Naturais/enzimologia , Células Matadoras Naturais/imunologia , Camundongos , Dados de Sequência Molecular , Família Multigênica , Filogenia , Estrutura Secundária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Linfócitos T/enzimologia , Linfócitos T/imunologia
17.
Toxicol Appl Pharmacol ; 280(2): 264-71, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25168425

RESUMO

UNLABELLED: The role of the immune system, specifically NK, NKT and CD3 cells, in acetaminophen (APAP) induced liver injury remains inconsistently defined. In the present study, wild type (C57BL/6J) mice and granzyme B deficient (GrB -/-) mice were treated with acetaminophen to assess the role of the immune system in acute liver injury. Doses of acetaminophen that induced sub lethal liver injury in wild type mice unexpectedly produced fatal hepatotoxicity in granzyme B deficient (GrB -/-) mice. Analysis revealed that GrB -/- mice had an increased population of intrahepatic CD3 (+), CD4 (-), and CD8 (-) lymphocytes expressing the CD69 activation marker and Fas ligand. Depletion of these cells in the GrB -/- and wild type mice made them less susceptible to APAP injury, while depletion of NK1.1 (+) cells or both CD4 (+) and CD8 (+) T cells failed to provide the same hepatoprotection. Transfer of the GrB -/- IHLs further exacerbated liver injury and increased mortality in wild type mice but not in LRP/LPR mice, lacking fas expression. CONCLUSIONS: Acetaminophen toxicity is enhanced by the presence of activated, FasL expressing intrahepatic CD3 (+), CD4 (-), CD8 (-), NK1.1 (-) T cells. Depletion of these cells from GrB -/- mice and wild type mice greatly reduces mortality and improves the course of liver injury recovery.


Assuntos
Acetaminofen/toxicidade , Fígado/efeitos dos fármacos , Linfócitos T/imunologia , Alanina Transaminase/sangue , Animais , Antígenos Ly/análise , Complexo CD3/análise , Antígenos CD4/análise , Antígenos CD8/análise , Proteína Ligante Fas/análise , Granzimas/fisiologia , Fígado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Subfamília B de Receptores Semelhantes a Lectina de Células NK/análise
18.
Transplantation ; 98(4): 402-10, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-24918613

RESUMO

BACKGROUND: Protease inhibitor 9 (PI-9) is an intracellular serpin that specifically inhibits granzyme B, a cytotoxic serine protease found in the cytosolic granules of cytotoxic T lymphocytes and natural killer cells. Enhanced cortical expression of PI-9 has been observed in kidney allografts with subclinical rejection, suggesting that the tubular epithelial cell (TEC) expression of this protein may have a protective role and attenuate overt allograft rejection. METHODS AND RESULTS: We demonstrate that TEC express SPI-6 protein, the murine homolog of PI-9, basally with a modest increase after cytokine exposure. Tubular epithelial cell expression of SPI-6 blocks granzyme B-mediated death because TEC from SPI-6 null kidneys have increased susceptibility to cytotoxic CD8+ cells in vitro. The role of SPI-6 was tested in a mouse kidney transplant model using SPI-6 null or wild type donor kidneys (H-2) into nephrectomized recipients (H-2). SPI-6 null kidney recipients demonstrated reduced renal function at day 8 after transplantation compared to controls (creatinine, 113±23 vs. 28±3 µmol/L; n=5; P<0.01), consistent with observed tubular injury and extensive mononuclear cell infiltration. Loss of donor kidney SPI-6 shortened graft survival time (20±19 vs. 66±33 days; n=8-10; P<0.001). CONCLUSION: Our data show for the first time that resistance of kidney TEC to cytotoxic T-cell granzyme B-induced death in vitro and in vivo is mediated by the expression of SPI-6. We suggest that SPI-6 is an important endogenous mechanism to prevent rejection injury from perforin or granzyme B effectors and enhanced PI-9 or SPI-6 expressions by TEC may provide protection from diverse forms of inflammatory kidney injury and promote long-term allograft survival.


Assuntos
Sobrevivência de Enxerto , Granzimas/antagonistas & inibidores , Transplante de Rim , Túbulos Renais/patologia , Proteínas de Membrana/fisiologia , Serpinas/fisiologia , Aloenxertos , Animais , Células Cultivadas , Células Epiteliais/patologia , Rejeição de Enxerto , Granzimas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
19.
Endocrinology ; 155(8): 2909-23, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24840346

RESUMO

Estrogen has potent immunomodulatory effects on proinflammatory responses, which can be mediated by serine proteases. We now demonstrate that estrogen increased the extracellular expression and IL-12-induced activity of a critical member of serine protease family Granzyme A, which has been shown to possess a novel inflammatory persona. The inhibition of serine protease activity with inhibitor 4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride significantly diminished enhanced production of proinflammatory interferon-γ, IL-1ß, IL-1α, and Granzyme A activity even in the presence of a Th1-inducing cytokine, IL-12 from splenocytes from in vivo estrogen-treated mice. Inhibition of serine protease activity selectively promoted secretion of Th2-specific IL-4, nuclear phosphorylated STAT6A, signal transducer and activator of transcription (STAT)6A translocation, and STAT6A DNA binding in IL-12-stimulated splenocytes from estrogen-treated mice. Inhibition with 4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride reversed the down-regulation of Th2 transcription factors, GATA3 and c-Maf in splenocytes from estrogen-exposed mice. Although serine protease inactivation enhanced the expression of Th2-polarizing factors, it did not reverse estrogen-modulated decrease of phosphorylated STAT5, a key factor in Th2 development. Collectively, data suggest that serine protease inactivity augments the skew toward a Th2-like profile while down-regulating IL-12-induced proinflammatory Th1 biomolecules upon in vivo estrogen exposure, which implies serine proteases as potential regulators of inflammation. Thus, these studies may provide a potential mechanism underlying the immunomodulatory effect of estrogen and insight into new therapeutic strategies for proinflammatory and female-predominant autoimmune diseases.


Assuntos
Estrogênios/fisiologia , Granzimas/fisiologia , Mediadores da Inflamação/fisiologia , Interleucina-12/fisiologia , Inibidores de Serina Proteinase/farmacologia , Células Th2/metabolismo , Animais , Citocinas/metabolismo , Feminino , Fator de Transcrição GATA3/metabolismo , Granzimas/metabolismo , Interleucina-4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-maf/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT6/metabolismo , Sulfonas/farmacologia , Células Th2/enzimologia , Regulação para Cima/fisiologia
20.
Cancer Genomics Proteomics ; 11(2): 67-79, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24709544

RESUMO

Proteases are often overexpressed in tumor cells and/or the stromal compartment and can thus be exploited in tumor therapy to activate cytotoxic prodrugs as, for example, in cytolytic fusion proteins, and for tumor imaging. Specifically, we discuss cathepsin B-activated prodrug conjugates, antibody-directed prodrug therapy, protease-activated peptide-thapsigargin conjugates, protease-activated cytotoxic receptor ligands and other cytotoxic proteins, protease-mediated activation of anthrax toxin, granzyme B as a therapeutic principle in cytolytic fusion proteins, and tumor-imaging based on deregulated proteases.


Assuntos
Antineoplásicos/metabolismo , Neoplasias/tratamento farmacológico , Pró-Fármacos/metabolismo , Animais , Antineoplásicos/uso terapêutico , Catepsina B/fisiologia , Granzimas/fisiologia , Humanos , Metaloproteinases da Matriz/fisiologia , Pró-Fármacos/uso terapêutico , Proteólise , Serina Endopeptidases/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...