Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 141(19): 2330-2342, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-36706356

RESUMO

Familial forms of the severe immunoregulatory disease hemophagocytic lymphohistiocytosis (HLH) arise from biallelic mutations in the PRF1, UNC13D, STXBP2, and STX11 genes. Early and accurate diagnosis of the disease is important to determine the most appropriate treatment option, including potentially curative stem cell transplantation. The diagnosis of familial HLH (FHL) is traditionally based on finding biallelic mutations in patients with HLH symptoms and reduced natural killer (NK)-cell cytotoxicity. However, patients often have a low NK-cell count or receive immunosuppressive therapies that may render the NK-cell cytotoxicity assay unreliable. Furthermore, to fully understand the nature of a disease it is critical to directly assess the effect of mutations on cellular function; this will help to avoid instances in which carriers of innocuous mutations may be recommended for invasive procedures including transplantation. To overcome this diagnostic problem, we have developed a rapid and robust method that takes advantage of the functional equivalence of the human and mouse orthologues of PRF1, UNC13D, STX11, and STXBP2 proteins. By knocking out endogenous mouse genes in CD8+ T cells and simultaneously replacing them with their mutated human orthologues, we can accurately assess the effect of mutations on cell function. The wide dynamic range of this novel system allowed us to understand the basis of, otherwise cryptic, cases of FHL or HLH and, in some instances, to demonstrate that previously reported mutations are unlikely to cause FHL. This novel approach provides valuable new information to enable more accurate diagnosis and treatment of patients with HLH or FHL who inherit mutations of undetermined pathogenicity.


Assuntos
Linfo-Histiocitose Hemofagocítica , Humanos , Animais , Camundongos , Linfo-Histiocitose Hemofagocítica/diagnóstico , Linfo-Histiocitose Hemofagocítica/genética , Proteínas Citotóxicas Formadoras de Poros , Perforina/genética , Genótipo , Mutação , Fenótipo , Proteínas de Membrana/genética , Proteínas Munc18/genética
2.
Blood ; 139(12): 1833-1849, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35081253

RESUMO

Niemann-Pick disease type C1 (NP-C1) is a rare lysosomal storage disorder resulting from mutations in an endolysosomal cholesterol transporter, NPC1. Despite typically presenting with pronounced neurological manifestations, NP-C1 also resembles long-term congenital immunodeficiencies that arise from impairment of cytotoxic T lymphocyte (CTL) effector function. CTLs kill their targets through exocytosis of the contents of lysosome-like secretory cytotoxic granules (CGs) that store and ultimately release the essential pore-forming protein perforin and proapoptotic serine proteases, granzymes, into the synapse formed between the CTL and target cell. We discovered that NPC1 deficiency increases CG lipid burden, impairs autophagic flux through stalled trafficking of the transcription factor EB (TFEB), and dramatically reduces CTL cytotoxicity. Using a variety of immunological and cell biological techniques, we found that the cytotoxic defect arises specifically from impaired perforin pore formation. We demonstrated defects of CTL function of varying severity in patients with NP-C1, with the greatest losses of function associated with the most florid and/or earliest disease presentations. Remarkably, perforin function and CTL cytotoxicity were restored in vitro by promoting lipid clearance with therapeutic 2-hydroxypropyl-ß-cyclodextrin; however, restoration of autophagy through TFEB overexpression was ineffective. Overall, our study revealed that NPC1 deficiency has a deleterious impact on CTL (but not natural killer cell) cytotoxicity that, in the long term, may predispose patients with NP-C1 to atypical infections and impaired immune surveillance more generally.


Assuntos
Doença de Niemann-Pick Tipo A , Doença de Niemann-Pick Tipo C , Colesterol/metabolismo , Granzimas , Humanos , Doença de Niemann-Pick Tipo C/metabolismo , Perforina/genética , Linfócitos T Citotóxicos/metabolismo
3.
Front Immunol ; 13: 931820, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36618385

RESUMO

When killing through the granule exocytosis pathway, cytotoxic lymphocytes release key effector molecules into the immune synapse, perforin and granzymes, to initiate target cell killing. The pore-forming perforin is essential for the function of cytotoxic lymphocytes, as its pores disrupt the target cell membrane and allow diffusion of pro-apoptotic serine proteases, granzyme, into the target cell, where they initiate various cell death cascades. Unlike human perforin, the detection of its murine counterpart in a live cell system has been problematic due its relatively low expression level and the lack of sensitive antibodies. The lack of a suitable methodology to visualise murine perforin secretion into the synapse hinders the study of the cytotoxic lymphocyte secretory machinery in murine models of human disease. Here, we describe a novel recombinant technology, whereby a short ALFA-tag sequence has been fused with the amino-terminus of a mature murine perforin, and this allowed its detection by the highly specific FluoTag®-X2 anti-ALFA nanobodies using both Total Internal Reflection Fluorescence (TIRF) microscopy of an artificial synapse, and confocal microscopy of the physiological immune synapse with a target cell. This methodology can have broad application in the field of cytotoxic lymphocyte biology and for the many models of human disease.


Assuntos
Sinapses Imunológicas , Perforina , Linfócitos T Citotóxicos , Animais , Camundongos , Morte Celular , Membrana Celular/metabolismo , Granzimas/metabolismo , Perforina/metabolismo
4.
Faraday Discuss ; 232(0): 236-255, 2021 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-34545865

RESUMO

Perforin is a pore forming protein used by cytotoxic T lymphocytes to remove cancerous or virus-infected cells during the immune response. During the response, the lymphocyte membrane becomes refractory to perforin function by accumulating densely ordered lipid rafts and externalizing negatively charged lipid species. The dense membrane packing lowers the capacity of perforin to bind, and the negatively charged lipids scavenge any residual protein before pore formation. Using atomic force microscopy on model membrane systems, we here provide insight into the molecular basis of perforin lipid specificity.


Assuntos
Lipídeos , Linfócitos T Citotóxicos , Perforina , Proteínas Citotóxicas Formadoras de Poros
5.
Methods Enzymol ; 629: 291-306, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31727246

RESUMO

Perforin is an indispensable effector protein of primary cytotoxic lymphocytes (CTL or NK cells) that typically defend the host against virus infection, or gene-modified (chimeric antigen receptor-CAR) anticancer T cells. Perforin's pore-forming activity is necessary for the delivery of proapoptotic serine proteases, granzymes, into the cytosol of infected or cancerous target cells. The complete loss of perforin function is detrimental for the function of cytotoxic lymphocytes, and leads to fatal immune dysregulation in infants and predisposes the carriers of hypomorphic perforin mutations to various chronic inflammatory sequelae and blood cancers. Here, we describe several optimized and validated functional assays using purified effector proteins and cytotoxic lymphocytes that enable detailed analysis of perforin-mediated target cell death pathways.


Assuntos
Microscopia Intravital/métodos , Perforina/metabolismo , Animais , Apoptose/imunologia , Eritrócitos , Granzimas/imunologia , Granzimas/metabolismo , Hemólise/imunologia , Microscopia Intravital/instrumentação , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos Transgênicos , Pressão Osmótica , Perforina/genética , Perforina/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Ovinos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Imagem com Lapso de Tempo/instrumentação , Imagem com Lapso de Tempo/métodos
6.
Nat Commun ; 10(1): 5396, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31776337

RESUMO

Killer T cells (cytotoxic T lymphocytes, CTLs) maintain immune homoeostasis by eliminating virus-infected and cancerous cells. CTLs achieve this by forming an immunological synapse with their targets and secreting a pore-forming protein (perforin) and pro-apoptotic serine proteases (granzymes) into the synaptic cleft. Although the CTL and the target cell are both exposed to perforin within the synapse, only the target cell membrane is disrupted, while the CTL is invariably spared. How CTLs escape unscathed remains a mystery. Here, we report that CTLs achieve this via two protective properties of their plasma membrane within the synapse: high lipid order repels perforin and, in addition, exposed phosphatidylserine sequesters and inactivates perforin. The resulting resistance of CTLs to perforin explains their ability to kill target cells in rapid succession and to survive these encounters. Furthermore, these mechanisms imply an unsuspected role for plasma membrane organization in protecting cells from immune attack.


Assuntos
Lipídeos de Membrana/química , Células T Matadoras Naturais/imunologia , Linfócitos T Citotóxicos/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Morte Celular , Linhagem Celular , Membrana Celular/química , Membrana Celular/metabolismo , Colesterol/metabolismo , Lipídeos de Membrana/metabolismo , Camundongos Transgênicos , Perforina/metabolismo , Fosfatidilserinas/metabolismo , Linfócitos T Citotóxicos/química , Linfócitos T Citotóxicos/imunologia
7.
J Exp Med ; 212(3): 307-17, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25732304

RESUMO

Failure of cytotoxic T lymphocytes (CTLs) or natural killer (NK) cells to kill target cells by perforin (Prf)/granzyme (Gzm)-induced apoptosis causes severe immune dysregulation. In familial hemophagocytic lymphohistiocytosis, Prf-deficient infants suffer a fatal "cytokine storm" resulting from macrophage overactivation, but the link to failed target cell death is not understood. We show that prolonged target cell survival greatly amplifies the quanta of inflammatory cytokines secreted by CTLs/NK cells and that interferon-γ (IFN-γ) directly invokes the activation and secondary overproduction of proinflammatory IL-6 from naive macrophages. Furthermore, using live cell microscopy to visualize hundreds of synapses formed between wild-type, Prf-null, or GzmA/B-null CTLs/NK cells and their targets in real time, we show that hypersecretion of IL-2, TNF, IFN-γ, and various chemokines is linked to failed disengagement of Prf- or Gzm-deficient lymphocytes from their targets, with mean synapse time increased fivefold, from ∼8 to >40 min. Surprisingly, the signal for detachment arose from the dying target cell and was caspase dependent, as delaying target cell death with various forms of caspase blockade also prevented their disengagement from fully competent CTLs/NK cells and caused cytokine hypersecretion. Our findings provide the cellular mechanism through which failed killing by lymphocytes causes systemic inflammation involving recruitment and activation of myeloid cells.


Assuntos
Citocinas/metabolismo , Células Matadoras Naturais/metabolismo , Linfócitos T Citotóxicos/metabolismo , Animais , Sinalização do Cálcio , Caspases/metabolismo , Sobrevivência Celular , Feminino , Granzimas/genética , Granzimas/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Perforina/genética , Perforina/metabolismo , Fatores de Tempo
8.
J Immunol ; 191(5): 2328-34, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23885110

RESUMO

The effective engagement of cytotoxic lymphocytes (CLs) with their target cells is essential for the removal of virus-infected and malignant cells from the body. The spatiotemporal properties that define CL engagement and killing of target cells remain largely uncharacterized due to a lack of biological reporters. We have used a novel live cell microscopy technique to visualize the engagement of primary human and mouse CL with their targets and the subsequent delivery of the lethal hit. Extensive quantitative real-time analysis of individual effector-target cell conjugates demonstrated that a single effector calcium flux event was sufficient for the degranulation of human CLs, resulting in the breach of the target cell membrane by perforin within 65-100 s. In contrast, mouse CLs demonstrated distinct calcium signaling profiles leading to degranulation: whereas mouse NKs required a single calcium flux event, CD8(+) T cells typically required several calcium flux events before perforin delivery. Irrespective of their signaling profile, every target cell that was damaged by perforin died by apoptosis. To our knowledge, we demonstrate for the first time that perforin pore delivery is unidirectional, occurring exclusively on the target cell membrane, but sparing the killer cell. Despite this, the CTL membrane was not intrinsically perforin resistant, as intact CTLs presented as targets to effector CTLs were capable of being killed by perforin-dependent mechanisms. Our results highlight the remarkable efficiency and specificity of perforin pore delivery by CLs.


Assuntos
Sinapses Imunológicas/imunologia , Células Matadoras Naturais/imunologia , Microscopia Confocal/métodos , Perforina/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Degranulação Celular/imunologia , Células Cultivadas , Humanos , Sinapses Imunológicas/metabolismo , Células Matadoras Naturais/metabolismo , Camundongos , Linfócitos T Citotóxicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...