Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Inflamm Res ; 71(2): 227-241, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34993560

RESUMO

OBJECTIVE: Trimetazidine (TMZ) exerts a strong inhibitory effect on ischemia/reperfusion (I/R) injury. Inflammation plays a key role in I/R injury. We hypothesized that TMZ may protect cardiomyocytes from I/R injury by inhibiting inflammation. METHODS: The left anterior descending coronary artery was ligated for 30 min followed by 6 h of reperfusion to establish a model of I/R injury. H9c2 cardiomyocytes were subjected to 2 h of hypoxia and 3 h of normoxic conditions to establish a model of hypoxia/reoxygenation (H/R) injury. We monitored the change in pyroptosis by performing Western blot analysis, microscopy and ELISA. RESULTS: I/R and H/R treatment stimulated gasdermin D-N domain (GSDMD-N) expression in cardiomyocytes (sham onefold vs. I/R 2.5-fold; control onefold vs. H/R 2.0-fold). Moreover, TMZ increased the viability of H9c2 cardiomyocytes subjected to H/R treatment (H/R 65.0% vs. H/R + TMZ 85.3%) and reduced the infarct size in vivo (I/R 47.0% vs. I/R + TMZ 28.3%). H/R and I/R treatment increased the levels of TLR4, MyD88, phospho-NF-κB p65 and the NLRP3 inflammasome; however, TMZ reduced the expression of these proteins. Additionally, TMZ inhibited noncanonical inflammasome signaling induced by I/R injury. CONCLUSIONS: In summary, TMZ alleviated pyroptosis induced by myocardial I/R injury through the TLR4/MyD88/NF-κB/NLRP3 inflammasome pathway. Therefore, TMZ represents an alternative treatment for myocardial I/R injury.


Assuntos
Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Proteínas de Ligação a Fosfato/antagonistas & inibidores , Proteínas Citotóxicas Formadoras de Poros/antagonistas & inibidores , Piroptose/efeitos dos fármacos , Trimetazidina/farmacologia , Animais , Masculino , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , NF-kappa B/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas de Ligação a Fosfato/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/fisiologia
2.
Int J Neurosci ; 132(1): 1-12, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32672480

RESUMO

MATERIALS AND METHODS: Locomotor outcomes in perforin-deficient (Pfp-/-) mice and wild-type littermate controls were measured after severe compression injury of the lower thoracic spinal cord up to six weeks after injury. RESULTS: According to both the Basso mouse scale score and single frame motion analysis, motor recovery of Pfp-/- mice was similar to wild-type controls. Interestingly, immunohistochemical analysis of cell types at six weeks after injury showed enhanced cholinergic reinnervation of spinal motor neurons caudal to the lesion site and neurofilament-positive structures at the injury site in Pfp-/- mice, whereas numbers of microglia/macrophages and astrocytes were decreased compared with controls. CONCLUSIONS: We conclude that, although, loss of perforin does not change the locomotor outcome after injury, it beneficially affects diverse cellular features, such as number of axons, cholinergic synapses, astrocytes and microglia/macrophages at or caudal to the lesion site. Perforin's ability to contribute to Rag2's influence on locomotion was observed in mice doubly deficient in perforin and Rag2 which recovered better than Rag2-/- or Pfp-/- mice, suggesting that natural killer cells can cooperate with T- and B-cells in spinal cord injury.


Assuntos
Locomoção/fisiologia , Regeneração Nervosa/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Traumatismos da Medula Espinal/enzimologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/deficiência
3.
J Cell Mol Med ; 25(17): 8159-8168, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34369076

RESUMO

Pyroptosis is a new form of programmed cell death generated by some inflammasomes, piloting the cleavage of gasdermin (GSDM) and stimulation of dormant cytokines like IL-18 and IL-1ß; these reactions are narrowly linked to certain diseases like diabetic nephropathy and atherosclerosis. Doxorubicin, a typical anthracycline, and famous anticancer drug has emerged as a prominent medication in several cancer chemotherapies, although its application is accompanied with expending of dose-dependent, increasing, irreversible and continuing cardiotoxic side effects. However, the exact path that links the induced pyroptosis to the mechanism by which Doxorubicin (DOX) acts against breast cancer cells is still puzzling. The present study seeks to elucidate the potential link between DOX-induced cell death and pyroptosis in two human breast cancer cell lines (MDA-MB-231 and T47D). We proved that treatment with DOX reduced the cell viability in a dose-dependent way and induced pyroptosis morphology in MDA-MB-231 and T47D cells. Also, protein expression analyses revealed GSDME as a key regulator in DOX-induced pyroptosis and highlighted the related role of Caspase-3 activation. Furthermore, DOX treatments induced intracellular accumulation of ROS, stimulated the phosphorylation of JNK, and Caspase-3 activation, subsequently. In conclusion, the study suggests that GSDME triggered DOX-induced pyroptosis in the caspase-3 dependent reactions through the ROS/JNK signalling pathway. Additionally, it showed that the DOX-induced cardiotoxicity and pyroptosis in breast cancer cells can be minimized by reducing the protein level of GSDME; thus, these outcomes provide a new research target and implications for the anticancer investigations and therapeutic applications.


Assuntos
Neoplasias da Mama , Caspase 3/fisiologia , Doxorrubicina/farmacologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Piroptose/efeitos dos fármacos , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
4.
Biochem Soc Trans ; 49(6): 2697-2710, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34812891

RESUMO

Pyroptosis is a highly inflammatory and lytic type of programmed cell death (PCD) commenced by inflammasomes, which sense perturbations in the cytosolic environment. Recently, several ground-breaking studies have linked a family of pore-forming proteins known as gasdermins (GSDMs) to pyroptosis. The human genome encodes six GSDM proteins which have a characteristic feature of forming pores in the plasma membrane resulting in the disruption of cellular homeostasis and subsequent induction of cell death. GSDMs have an N-terminal cytotoxic domain and an auto-inhibitory C-terminal domain linked together through a flexible hinge region whose proteolytic cleavage by various enzymes releases the N-terminal fragment that can insert itself into the inner leaflet of the plasma membrane by binding to acidic lipids leading to pore formation. Emerging studies have disclosed the involvement of GSDMs in various modalities of PCD highlighting their role in diverse cellular and pathological processes. Recently, the cryo-EM structures of the GSDMA3 and GSDMD pores were resolved which have provided valuable insights into the pore formation process of GSDMs. Here, we discuss the current knowledge regarding the role of GSDMs in PCD, structural and molecular aspects of autoinhibition, and pore formation mechanism followed by a summary of functional consequences of gasdermin-induced membrane permeabilization.


Assuntos
Apoptose/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Animais , Humanos , Proteínas Citotóxicas Formadoras de Poros/química , Conformação Proteica
5.
Int J Mol Sci ; 21(14)2020 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-32664669

RESUMO

The aggregation and accumulation of amyloid-ß plaques and tau proteins in the brain have been central characteristics in the pathophysiology of Alzheimer's disease (AD), making them the focus of most of the research exploring potential therapeutics for this neurodegenerative disease. With success in interventions aimed at depleting amyloid-ß peptides being limited at best, a greater understanding of the physiological role of amyloid-ß peptides is needed. The development of amyloid-ß plaques has been determined to occur 10-20 years prior to AD symptom manifestation, hence earlier interventions might be necessary to address presymptomatic AD. Furthermore, recent studies have suggested that amyloid-ß peptides may play a role in innate immunity as an antimicrobial peptide. These findings, coupled with the evidence of pathogens such as viruses and bacteria in AD brains, suggests that the buildup of amyloid-ß plaques could be a response to the presence of viruses and bacteria. This has led to the foundation of the antimicrobial hypothesis for AD. The present review will highlight the current understanding of amyloid-ß, and the role of bacteria and viruses in AD, and will also explore the therapeutic potential of antimicrobial and antiviral drugs in Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anti-Infecciosos/uso terapêutico , Antivirais/uso terapêutico , Encéfalo/microbiologia , Encéfalo/virologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/microbiologia , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/fisiologia , Animais , Anti-Infecciosos/farmacologia , Antivirais/farmacologia , Infecções Bacterianas/complicações , Infecções Bacterianas/tratamento farmacológico , Barreira Hematoencefálica , Ensaios Clínicos como Assunto , Citocinas/metabolismo , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Humanos , Imunidade Inata , Inflamação , Camundongos , Camundongos Knockout , Neuroglia/metabolismo , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Terapias em Estudo , Viroses/complicações , Viroses/tratamento farmacológico
6.
Blood ; 127(17): 2113-21, 2016 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-26864340

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory disease. Inherited forms of HLH are caused by biallelic mutations in several effectors of granule-dependent lymphocyte-mediated cytotoxicity. A small proportion of patients with a so-called "secondary" form of HLH, which develops in the aftermath of infection, autoimmunity, or cancer, carry a monoallelic mutation in one or more HLH-associated genes. Although this observation suggests that HLH may have a polygenic mode of inheritance, the latter is very difficult to prove in humans. In order to determine whether the accumulation of partial genetic defects in lymphocyte-mediated cytotoxicity can contribute to the development of HLH, we generated mice that were doubly or triply heterozygous for mutations in HLH-associated genes, those coding for perforin, Rab27a, and syntaxin-11. We found that the accumulation of monoallelic mutations did indeed increase the risk of developing HLH immunopathology after lymphocytic choriomeningitis virus infection. In mechanistic terms, the accumulation of heterozygous mutations in the two degranulation genes Rab27a and syntaxin-11, impaired the dynamics and secretion of cytotoxic granules at the immune synapse of T lymphocytes. In addition, the accumulation of heterozygous mutations within the three genes impaired natural killer lymphocyte cytotoxicity in vivo. The genetic defects can be ranked in terms of the severity of the resulting HLH manifestations. Our results form the basis of a polygenic model of the occurrence of secondary HLH.


Assuntos
Linfo-Histiocitose Hemofagocítica/genética , Herança Multifatorial , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Qa-SNARE/genética , Proteínas rab de Ligação ao GTP/genética , Animais , Degranulação Celular/genética , Cruzamentos Genéticos , Citotoxicidade Imunológica/genética , Dosagem de Genes , Predisposição Genética para Doença , Heterozigoto , Sinapses Imunológicas/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Mutação , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Proteínas Qa-SNARE/fisiologia , Organismos Livres de Patógenos Específicos , Linfócitos T Citotóxicos/imunologia , Proteínas rab de Ligação ao GTP/fisiologia , Proteínas rab27 de Ligação ao GTP
7.
Nat Rev Immunol ; 6(12): 940-52, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17124515

RESUMO

The granule exocytosis pathway of cytotoxic lymphocytes is crucial for immune surveillance and homeostasis. The trafficking of granule components, including the membrane-disruptive protein perforin, to the immunological synapse leads to the delivery of granule proteases (granzymes) into the target cell and its destruction through apoptosis. Several independent molecular abnormalities associated with defects of either granule trafficking or perforin function can cause cytotoxic lymphocyte dysfunction. In humans, inherited perforin mutations result in severe immune dysregulation that manifests as familial haemophagocytic lymphohistiocytosis. This Review describes recent progress in defining the structure, function, biochemistry and cell biology of perforin.


Assuntos
Exocitose , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Animais , Morte Celular/genética , Exocitose/genética , Homeostase/imunologia , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Mutação , Perforina , Polimorfismo Genético , Proteínas Citotóxicas Formadoras de Poros/genética , Estrutura Terciária de Proteína
8.
PLoS Pathog ; 10(5): e1004142, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24854422

RESUMO

Septic pneumonias resulting from bacterial infections of the lung are a leading cause of human death worldwide. Little is known about the capacity of CD8 T cell-mediated immunity to combat these infections and the types of effector functions that may be most effective. Pneumonic plague is an acutely lethal septic pneumonia caused by the Gram-negative bacterium Yersinia pestis. We recently identified a dominant and protective Y. pestis antigen, YopE69-77, recognized by CD8 T cells in C57BL/6 mice. Here, we use gene-deficient mice, Ab-mediated depletion, cell transfers, and bone marrow chimeric mice to investigate the effector functions of YopE69-77-specific CD8 T cells and their relative contributions during pulmonary Y. pestis infection. We demonstrate that YopE69-77-specific CD8 T cells exhibit perforin-dependent cytotoxicity in vivo; however, perforin is dispensable for YopE69-77-mediated protection. In contrast, YopE69-77-mediated protection is severely impaired when production of TNFα and IFNγ by CD8 T cells is simultaneously ablated. Interestingly, TNFα is absolutely required at the time of challenge infection and can be provided by either T cells or non-T cells, whereas IFNγ provided by T cells prior to challenge appears to facilitate the differentiation of optimally protective CD8 T cells. We conclude that cytokine production, not cytotoxicity, is essential for CD8 T cell-mediated control of pulmonary Y. pestis infection and we suggest that assays detecting Ag-specific TNFα production in addition to antibody titers may be useful correlates of vaccine efficacy against plague and other acutely lethal septic bacterial pneumonias.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/genética , Interferon gama/fisiologia , Peste/imunologia , Pneumonia Bacteriana/imunologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Yersinia pestis/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/complicações , Peste/genética , Pneumonia Bacteriana/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Fator de Necrose Tumoral alfa/genética
9.
J Immunol ; 190(3): 1351-9, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23275602

RESUMO

Tumor-targeted vaccines represent a strategy to enhance the graft-versus-leukemia effect after allogeneic blood and marrow transplantation (BMT). We have previously shown that graft-versus-host disease (GVHD) can negatively impact quantitative responses to vaccines. Using a minor histocompatibility Ag-mismatched BMT (B6 → B6 × C3H.SW) followed by adoptive transfer of HY-specific T cells and HY-expressing dendritic cells, we assessed whether GVHD induced by donor lymphocyte infusion (DLI) affects the persistence, proliferation, and survival of vaccine-responding, nonalloantigen reactive T cells. Both CD8(+) and CD4(+) HY-specific T cells undergo less vaccine-driven proliferation in allogeneic recipients with GVHD. Although vaccine-responding CD8(+) T cells show decreased IFN-γ and CD107a production, CD4(+) T cells exhibit increased programmed death 1 and T cell Ig mucin-like domain 3 expression. In addition, the degree of apoptosis in vaccine-responding CD8(+) T cells was higher in the presence of GVHD, but there was no difference in CD4(+) T cell apoptosis. Using Fas ligand-deficient or TRAIL-deficient DLI had no impact on apoptosis of HY-specific T cells. However, perforin-deficient alloreactive DLI induced significantly less apoptosis of vaccine-responding CD8(+) T cells and resulted in enhanced tumor protection. Thus, diminished vaccine responses during GVHD result from impaired proliferation of CD8(+) and CD4(+) T cells responding to vaccination, with an additional contribution from perforin-mediated CD8(+) T cell apoptosis. These results provide important insights toward optimizing vaccine responses after allogeneic BMT.


Assuntos
Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/imunologia , Imunoterapia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Subpopulações de Linfócitos T/imunologia , Vacinação , Transferência Adotiva , Animais , Animais Congênicos , Vacinas Anticâncer/uso terapêutico , Carcinoma de Células de Transição/imunologia , Carcinoma de Células de Transição/patologia , Carcinoma de Células de Transição/terapia , Divisão Celular , Técnicas de Cocultura , Proteína Ligante Fas/deficiência , Proteína Ligante Fas/imunologia , Feminino , Antígeno H-Y/imunologia , Epitopos Imunodominantes/imunologia , Transfusão de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antígenos de Histocompatibilidade Menor/imunologia , Quimera por Radiação , Organismos Livres de Patógenos Específicos , Ligante Indutor de Apoptose Relacionado a TNF/deficiência , Ligante Indutor de Apoptose Relacionado a TNF/imunologia
10.
Subcell Biochem ; 80: 3-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798004

RESUMO

Membranes are essential in defining the border and ensuring function of all living cells. As such they are vulnerable and have been a preferred target of attack throughout evolution. The most powerful way of damaging a membrane is through the insertion of pore-forming proteins. Research over the last decades shows that such proteins are produced by bacteria to attack bacterial or eukaryotic cells, vertebrates to kill invading organisms or infected cells, and by eukaryotic cells to "kill" mitochondria and trigger apoptosis. The breadth of effect of these proteins is bringing together, in a very exciting way, research communities that used to be unaware of each other.


Assuntos
Toxinas Bacterianas/metabolismo , Membrana Celular/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Animais , Bactérias/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacologia , Humanos , Perforina/química , Perforina/farmacologia , Perforina/fisiologia , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/farmacologia
11.
Subcell Biochem ; 80: 197-220, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798013

RESUMO

Perforin (PFN) is the key pore-forming molecule in the cytotoxic granules of immune killer cells. Expressed only in killer cells, PFN is the rate-limiting molecule for cytotoxic function, delivering the death-inducing granule serine proteases (granzymes) into target cells marked for immune elimination. In this chapter we describe our current understanding of how PFN accomplishes this task. We discuss where PFN is expressed and how its expression is regulated, the biogenesis and storage of PFN in killer cells and how they are protected from potential damage, how it is released, how it delivers Granzymes into target cells and the consequences of PFN deficiency.


Assuntos
Citotoxicidade Imunológica , Neoplasias/imunologia , Perforina/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Vírus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Humanos , Imunidade Celular/fisiologia , Perforina/química , Proteínas Citotóxicas Formadoras de Poros/química
12.
Subcell Biochem ; 80: 161-95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798012

RESUMO

The cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins that are produced by numerous Gram-positive bacterial pathogens. These toxins are released in the extracellular environment as water-soluble monomers or dimers that bind to cholesterol-rich membranes and assemble into large pore complexes. Depending upon their concentration, the nature of the host cell and membrane (cytoplasmic or intracellular) they target, the CDCs can elicit many different cellular responses. Among the CDCs, listeriolysin O (LLO), which is a major virulence factor of the facultative intracellular pathogen Listeria monocytogenes, is involved in several stages of the intracellular lifecycle of the bacterium and displays unique characteristics. It has long been known that following L. monocytogenes internalization into host cells, LLO disrupts the internalization vacuole, enabling the bacterium to replicate into the host cell cytosol. LLO is then used by cytosolic bacteria to spread from cell to cell, avoiding bacterial exposure to the extracellular environment. Although LLO is continuously produced during the intracellular lifecycle of L. monocytogenes, several processes limit its toxicity to ensure the survival of infected cells. It was previously thought that LLO activity was limited to mediating vacuolar escape during bacterial entry and cell to cell spreading. This concept has been challenged by compelling evidence suggesting that LLO secreted by extracellular L. monocytogenes perforates the host cell plasma membrane, triggering important host cell responses. This chapter provides an overview of the well-established intracellular activity of LLO and the multiple roles attributed to LLO secreted by extracellular L. monocytogenes.


Assuntos
Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Listeria monocytogenes/patogenicidade , Animais , Toxinas Bacterianas/química , Colesterol/metabolismo , Citotoxinas/química , Citotoxinas/metabolismo , Citotoxinas/fisiologia , Regulação Bacteriana da Expressão Gênica , Proteínas de Choque Térmico/química , Proteínas Hemolisinas/química , Humanos , Listeria monocytogenes/genética , Modelos Moleculares , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Fatores de Virulência/química , Fatores de Virulência/fisiologia
13.
Subcell Biochem ; 80: 271-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798017

RESUMO

Proteins with membrane-attack complex/perforin (MACPF) domains are found in almost all kingdoms of life, and they have a variety of biological roles, including defence and attack, organism development, and cell adhesion and signalling. The distribution of these proteins in fungi appears to be restricted to some Pezizomycotina and Basidiomycota species only, in correlation with another group of proteins with unknown biological function, known as aegerolysins. These two protein groups coincide in only a few species, and they might operate in concert as cytolytic bi-component pore-forming agents. Representative proteins here include pleurotolysin B, which has a MACPF domain, and the aegerolysin-like protein pleurotolysin A, and the very similar ostreolysin A, which have been purified from oyster mushroom (Pleurotus ostreatus). These have been shown to act in concert to perforate natural and artificial lipid membranes with high cholesterol and sphingomyelin content. The aegerolysin-like proteins provide the membrane cholesterol/sphingomyelin selectivity and recruit oligomerised pleurotolysin B molecules, to create a membrane-inserted pore complex. The resulting protein structure has been imaged with electron microscopy, and it has a 13-meric rosette-like structure, with a central lumen that is ~4-5 nm in diameter. The opened transmembrane pore is non-selectively permeable for ions and smaller neutral solutes, and is a cause of cytolysis of a colloid-osmotic type. The biological significance of these proteins for the fungal life-style is discussed.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Proteínas Fúngicas/fisiologia , Proteínas Hemolisinas/fisiologia , Perforina/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Sequência de Aminoácidos , Animais , Complexo de Ataque à Membrana do Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Humanos , Dados de Sequência Molecular , Perforina/química , Filogenia , Pleurotus/genética , Pleurotus/patogenicidade , Proteínas Citotóxicas Formadoras de Poros/química , Multimerização Proteica/fisiologia , Homologia de Sequência de Aminoácidos
14.
Am J Transplant ; 14(3): 580-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24730048

RESUMO

Memory T (Tm) cells pose a major barrier to long-term transplant survival. Whether regulatory T cells (Tregs)can control them remains poorly defined. Previously,we established that double-negative (DN) Tregs suppress effector T (Teff) cells. Here, we demonstrate that DNTregs effectively suppress CD4+/CD8+Teff and CD8+Tm but not CD4+Tm cells, whereas the suppression on CD8+Tm is abrogated by perforin (PFN) deficiency in DNTregs. Consistently, in a BALB/c to B6-Rag1-/-skin transplantation, transfer of DN Tregs suppressed the rejection mediated by CD4þ/CD8+Teff and CD8+Tmcells (76.0±4.9, 87.5±5.0 and 63.0±4.7 days, respectively)but not CD4þTmcells (25.3±1.4 days). Both CD8þ effector memory T and central memory T compartments significantly reduced after DN Treg transfer. CD4+Tm highly expresses granzyme B (GzmB) inhibitor serine protease inhibitor-6 (Spi6). Spi6 deficiency renders CD4þTm susceptible to DN Treg suppression. In addition,transfer of WT DN Tregs, but not PFN-/-DN Tregs,inhibited the skin allograft rejection mediated by Spi6-/-CD4þTm(75.5±7.9 days). In conclusion, CD4+ and CD8+Tm cells differentially respond toDNTregs' suppression.The GzmB resistance conferred by Spi6 in CD4þTm cells might hint at the physiological significance of Tmpersistence


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Granzimas/fisiologia , Memória Imunológica/imunologia , Proteínas de Membrana/fisiologia , Serpinas/fisiologia , Transplante de Pele , Linfócitos T Reguladores/imunologia , Animais , Western Blotting , Proteína Ligante Fas/fisiologia , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Citotóxicas Formadoras de Poros/fisiologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Cancer Immunol Immunother ; 63(6): 571-80, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24658838

RESUMO

One alternative approach for the treatment of lung cancer might be the activation of the immune system using vaccination strategies. However, most of clinical vaccination trials for lung cancer did not reach their primary end points, suggesting that lung cancer is of low immunogenicity. To provide additional experimental information about this important issue, we investigated which type of immune cells contributes to the protection from lung cancer development. Therefore, A/J mice induced for lung adenomas/ adenocarcinomas by the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) were depleted of CD4+ or CD8+ T cells, CD11b+ macrophages, Gr-1+ neutrophils and asialo GM1+ natural killer (NK) cells. Subsequent analysis of tumour growth showed an increase in tumour number only in mice depleted of NK cells. Further asking by which mechanism NK cells suppressed tumour development, we neutralized several death ligands of the tumour necrosis factor (TNF) family known to be involved in NK cell-mediated cytotoxicity. However neither depletion of TNF-α, TNF-related apoptosis-inducing ligand, TNF-like weak inducer of apoptosis or FasL alone nor in combination induced an augmentation of tumour burden. To show whether an alternative cell death pathway is involved, we next generated A/J mice deficient for perforin. After challenging with NNK, mice deficient for perforin showed an increase in tumour number and volume compared to wild-type A/J mice. In summary, our data suggest that NK cells and perforin-mediated cytolysis are critically involved in the protection from lung cancer giving promise for further immunotherapeutic strategies for this disease.


Assuntos
Adenocarcinoma/imunologia , Apoptose , Citotoxicidade Imunológica/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/imunologia , Nitrosaminas/toxicidade , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/patologia , Animais , Carcinógenos/toxicidade , Feminino , Fatores de Transcrição Forkhead/metabolismo , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos A , Linfócitos T/imunologia , Linfócitos T/patologia , Fator de Necrose Tumoral alfa/metabolismo
16.
Blood ; 119(7): 1713-6, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-22186995

RESUMO

Mutations in the perforin gene (PRF1) are a common cause of the fatal immune dysregulation disorder, familial hemophagocytic lymphohistiocytosis (type 2 FHL, FHL2). Here we report a female infant born with biallelic PRF1 mutations: a novel substitution, D49N, and a previously identified in-frame deletion, K285del. We assessed the effects of each mutation on the cytotoxicity of human NK cells in which the expression of endogenous perforin was ablated with miR30-based short hairpin (sh) RNAs. Both mutations were detrimental for function, thereby explaining the clinically severe presentation and rapidly fatal outcome. We demonstrate that D49N exerts its deleterious effect by generating an additional (third) N-linked glycosylation site, resulting in protein misfolding and degradation in the killer cell. Our data provide a rationale for treating some cases of type 2 familial hemophagocytic lymphohistiocytosis, based on the pharmacologic inhibition or modification of glycosylation.


Assuntos
Doenças do Sistema Imunitário/genética , Linfócitos/metabolismo , Mutação de Sentido Incorreto/fisiologia , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Sequência de Bases , Células Cultivadas , Análise Mutacional de DNA , Evolução Fatal , Feminino , Glicosilação , Células HEK293 , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/patologia , Recém-Nascido , Linfócitos/imunologia , Linfócitos/patologia , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/imunologia , Linhagem , Perforina , Proteínas Citotóxicas Formadoras de Poros/fisiologia
17.
Scand J Immunol ; 79(6): 395-403, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24684620

RESUMO

Infection with murine gammaherpesvirus 68 has become an accepted model for studying the virus/host interactions with regard to gammaherpesvirus infections. Previous studies using gene-deficient mice have revealed that neither IFNγ nor perforin is essential in controlling the outcome of infection or the virus load during chronic infection in C57BL/6 mice. However, pronounced multiorgan fibrosis and splenic atrophy are observed in mice lacking IFNγ or the IFNγ receptor. To study the interplay between perforin and IFNγ in controlling the virus-induced pathology and the viral load during chronic gammaherpesvirus infection, we infected IFNγ/perforin double-deficient C57BL/6 mice and followed the course of infection. While absence of perforin prevented the splenic atrophy in IFNγ-deficient mice, fibrosis did not disappear. Moreover, double-deficient mice developed extreme splenomegaly, were unable to control the viral load and displayed chronic immune activation. Thus, IFNγ and perforin act in concert to minimize pathology and control the viral load in mice chronically infected with MHV68. Furthermore, while certain aspect of the virus-induced pathology in IFNγ-deficient mice may be alleviated in double-deficient mice, other aspects are exaggerated, and the normal architecture of the spleen is completely destroyed. We believe that these findings add to the understanding of the virus/host interaction during chronic gammaherpes virus infection.


Assuntos
Infecções por Herpesviridae/imunologia , Interferon gama/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Rhadinovirus , Animais , Quimiocina CXCL1/sangue , Citocinas/sangue , Feminino , Infecções por Herpesviridae/patologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interferon/fisiologia , Receptor de Interferon gama
18.
J Immunol ; 189(5): 2099-109, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22869906

RESUMO

An effective immune response against influenza A infection depends on the generation of virus-specific T cells. NK cells are one of the first-line defenses against influenza A infection. We set out to delineate the role of NK cells in T cell immunity using a murine model of influenza A infection with A/PR/8/34. We show that early T cell recruitment mainly occurs in the posterior mediastinal lymph node (pMLN). Depletion of NK cells significantly impaired both dendritic cell (DC) and T cell recruitment into the pMLN. A similar reduction of T cell recruitment was observed when migration was blocked by pertussis toxin, suggesting that migration of pulmonary NK cells and DCs regulates cell recruitment to the pMLN. T cell recruitment was dependent on IFN-γ, and transfer of IFN-γ-competent naive NK cells into IFN-γ-/- mice restored T cell recruitment, whereas IFN-γ-deficient NK cells failed to do so. In addition, NK cell depletion reduced the uptake and transport of influenza A virus by DCs, and significantly impaired the virus-specific T cell response. Both IFN-γ-/- and perforin-/- mice showed reduced viral Ag transport by DCs, suggesting that the ability of NK cells to influence virus transport depends on IFN-γ and perforin. In summary, our data suggest that NK cells play a critical role in the initiation and shaping of the T cell response after influenza A infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Interferon gama/fisiologia , Células Matadoras Naturais/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Animais , Apoptose/imunologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Linhagem Celular Tumoral , Células Dendríticas/patologia , Células Dendríticas/virologia , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Células Matadoras Naturais/virologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia
19.
Eur J Immunol ; 42(6): 1488-99, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22678903

RESUMO

Perforin-deficient (PKO) mice serve as models for familial hemophagocytic lympho-histiocytosis, a uniformly fatal disease associated with viral infection of perforin-deficient humans. Naïve perforin-deficient BALB/c mice survive while vaccinated PKO mice containing virus-specific memory CD8(+) T cells rapidly succumb to lymphocytic choriomeningitis virus (LCMV) infection. Thus, vaccination converts a nonlethal persistent infection into a fatal disease mediated by virus-specific memory CD8(+) T cells. Here, we determine the extent to which vaccination-induced mortality in PKO mice following LCMV challenge is due to differences in vaccine modalities, the quantity or epitope specificity of memory CD8(+) T cells. We show that LCMV-induced mortality in immune PKO mice is independent of vaccine modalities and that the starting number of memory CD8(+) T cells specific to the immunodominant epitope NP(118-126) dictates the magnitude of secondary CD8(+) T-cell expansion, the inability to regulate production of CD8(+) T-cell-derived IFN-γ, and mortality in the vaccinated PKO mice. Importantly, mortality is determined by the epitope specificity of memory CD8(+) T cells and the associated degree of functional exhaustion and cytokine dysregulation but not the absolute magnitude of CD8(+) T-cell expansion. These data suggest that deeper understanding of the parameters that influence the outcome of vaccine-induced diseases would aid rational vaccine design to minimize adverse outcomes after infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T , Memória Imunológica , Coriomeningite Linfocítica/mortalidade , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas Citotóxicas Formadoras de Poros/deficiência , Vacinação/efeitos adversos , Vacinas Virais/efeitos adversos , Transferência Adotiva , Animais , Citocinas/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Vacinas Virais/imunologia
20.
Blood ; 118(22): 5794-8, 2011 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21881043

RESUMO

Familial hemophagocytic lymphohistiocytosis (HLH) is a rare primary immunodeficiency disorder characterized by defects in cell-mediated cytotoxicity that results in fever, hepatosplenomegaly, and cytopenias. Familial HLH is well recognized in children but rarely diagnosed in adults. We conducted a retrospective review of genetic and immunologic test results in patients who developed HLH in adulthood. Included in our study were 1531 patients with a clinical diagnosis of HLH; 175 patients were 18 years or older. Missense and splice-site sequence variants in PRF1, MUNC13-4, and STXBP2 were found in 25 (14%) of the adult patients. The A91V-PRF1 genotype was found in 12 of these patients (48%). The preponderance of hypomorphic mutations in familial HLH-causing genes correlates with the later-onset clinical symptoms and the more indolent course in adult patients. We conclude that late-onset familial HLH occurs more commonly than was suspected previously.


Assuntos
Linfo-Histiocitose Hemofagocítica/genética , Proteínas de Membrana/genética , Proteínas Munc18/genética , Mutação , Proteínas Citotóxicas Formadoras de Poros/genética , Adolescente , Adulto , Idade de Início , Idoso , Análise Mutacional de DNA , Feminino , Frequência do Gene , Humanos , Linfo-Histiocitose Hemofagocítica/epidemiologia , Masculino , Proteínas de Membrana/fisiologia , Pessoa de Meia-Idade , Proteínas Munc18/fisiologia , Mutação/fisiologia , Perforina , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA