Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 631(8019): 207-215, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38926576

RESUMEN

Pyroptosis is a lytic cell death mode that helps limit the spread of infections and is also linked to pathology in sterile inflammatory diseases and autoimmune diseases1-4. During pyroptosis, inflammasome activation and the engagement of caspase-1 lead to cell death, along with the maturation and secretion of the inflammatory cytokine interleukin-1ß (IL-1ß). The dominant effect of IL-1ß in promoting tissue inflammation has clouded the potential influence of other factors released from pyroptotic cells. Here, using a system in which macrophages are induced to undergo pyroptosis without IL-1ß or IL-1α release (denoted Pyro-1), we identify unexpected beneficial effects of the Pyro-1 secretome. First, we noted that the Pyro-1 supernatants upregulated gene signatures linked to migration, cellular proliferation and wound healing. Consistent with this gene signature, Pyro-1 supernatants boosted migration of primary fibroblasts and macrophages, and promoted faster wound closure in vitro and improved tissue repair in vivo. In mechanistic studies, lipidomics and metabolomics of the Pyro-1 supernatants identified the presence of both oxylipins and metabolites, linking them to pro-wound-healing effects. Focusing specifically on the oxylipin prostaglandin E2 (PGE2), we find that its synthesis is induced de novo during pyroptosis, downstream of caspase-1 activation and cyclooxygenase-2 activity; further, PGE2 synthesis occurs late in pyroptosis, with its release dependent on gasdermin D pores opened during pyroptosis. As for the pyroptotic metabolites, they link to immune cell infiltration into the wounds, and polarization to CD301+ macrophages. Collectively, these data advance the concept that the pyroptotic secretome possesses oxylipins and metabolites with tissue repair properties that may be harnessed therapeutically.


Asunto(s)
Macrófagos , Oxilipinas , Piroptosis , Secretoma , Cicatrización de Heridas , Animales , Femenino , Humanos , Ratones , Caspasa 1/metabolismo , Movimiento Celular , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Dinoprostona/biosíntesis , Dinoprostona/metabolismo , Fibroblastos/metabolismo , Fibroblastos/citología , Gasderminas/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta , Lipidómica , Macrófagos/metabolismo , Macrófagos/citología , Ratones Endogámicos C57BL , Oxilipinas/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Secretoma/metabolismo , Cicatrización de Heridas/fisiología
2.
Autophagy ; 19(11): 2958-2971, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37615626

RESUMEN

Macroautophagy/autophagy is a cellular recycling program regulating cell survival and controlling inflammatory responses in a context-dependent manner. Here, we demonstrate that keratinocyte-selective ablation of Atg16l1, an essential autophagy mediator, results in exacerbated inflammatory and neoplastic skin responses. In addition, mice lacking keratinocyte autophagy exhibit precocious onset of hair follicle growth, indicating altered activation kinetics of hair follicle stem cells (HFSCs). These HFSCs also exhibit expanded potencies in an autophagy-deficient context as shown by de novo hair follicle formation and improved healing of abrasion wounds. ATG16L1-deficient keratinocytes are markedly sensitized to apoptosis. Compound deletion of RIPK3-dependent necroptotic and CASP8-dependent apoptotic responses or of TNFRSF1A/TNFR1 reveals that the enhanced sensitivity of autophagy-deficient keratinocytes to TNF-dependent cell death is driving altered activation of HFSCs. Together, our data demonstrate that keratinocyte autophagy dampens skin inflammation and tumorigenesis but curtails HFSC activation by restraining apoptotic responses.Abbreviations: ATG16L1: autophagy related 16 like 1; DMBA: 2,4-dimethoxybenzaldehyde; DP: dermal papilla; EpdSCs: epidermal stem cells; Gas6: growth arrest specific 6; HF: hair follicle; HFSC: hair follicle stem cell; IFE: interfollicular epidermis; KRT5: keratin 5; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; PMK: primary mouse keratinocyte; RIPK3: receptor-interacting serine-threonine kinase 3; scRNAseq: single-cell RNA-sequencing; SG: sebaceous gland; TEWL: transepidermal water loss; TPA: 12-O-tetradecanoylphorbol-13-acetate; TNF: tumor necrosis factor; TNFRSF1A/TNFR1: tumor necrosis factor receptor superfamily, member 1a; UMAP: uniform manifold approximation and projection.

4.
Nat Commun ; 13(1): 3676, 2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35760796

RESUMEN

Immunogenic cell death significantly contributes to the success of anti-cancer therapies, but immunogenicity of different cell death modalities widely varies. Ferroptosis, a form of cell death that is characterized by iron accumulation and lipid peroxidation, has not yet been fully evaluated from this perspective. Here we present an inducible model of ferroptosis, distinguishing three phases in the process-'initial' associated with lipid peroxidation, 'intermediate' correlated with ATP release and 'terminal' recognized by HMGB1 release and loss of plasma membrane integrity-that serves as tool to study immune cell responses to ferroptotic cancer cells. Co-culturing ferroptotic cancer cells with dendritic cells (DC), reveals that 'initial' ferroptotic cells decrease maturation of DC, are poorly engulfed, and dampen antigen cross-presentation. DC loaded with ferroptotic, in contrast to necroptotic, cancer cells fail to protect against tumor growth. Adding ferroptotic cancer cells to immunogenic apoptotic cells dramatically reduces their prophylactic vaccination potential. Our study thus shows that ferroptosis negatively impacts antigen presenting cells and hence the adaptive immune response, which might hinder therapeutic applications of ferroptosis induction.


Asunto(s)
Ferroptosis , Neoplasias , Muerte Celular , Células Dendríticas , Humanos , Peroxidación de Lípido/fisiología
5.
Nature ; 606(7915): 776-784, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35614212

RESUMEN

Chronic non-healing wounds are a major complication of diabetes, which affects 1 in 10 people worldwide. Dying cells in the wound perpetuate the inflammation and contribute to dysregulated tissue repair1-3. Here we reveal that the membrane transporter SLC7A11 acts as a molecular brake on efferocytosis, the process by which dying cells are removed, and that inhibiting SLC7A11 function can accelerate wound healing. Transcriptomics of efferocytic dendritic cells in mouse identified upregulation of several SLC7 gene family members. In further analyses, pharmacological inhibition of SLC7A11, or deletion or knockdown of Slc7a11 using small interfering RNA enhanced efferocytosis in dendritic cells. Slc7a11 was highly expressed in dendritic cells in skin, and single-cell RNA sequencing of inflamed skin showed that Slc7a11 was upregulated in innate immune cells. In a mouse model of excisional skin wounding, inhibition or loss of SLC7A11 expression accelerated healing dynamics and reduced the apoptotic cell load in the wound. Mechanistic studies revealed a link between SLC7A11, glucose homeostasis and diabetes. SLC7A11-deficient dendritic cells were dependent on aerobic glycolysis using glucose derived from glycogen stores for increased efferocytosis; also, transcriptomics of efferocytic SLC7A11-deficient dendritic cells identified increased expression of genes linked to gluconeogenesis and diabetes. Further, Slc7a11 expression was higher in the wounds of diabetes-prone db/db mice, and targeting SLC7A11 accelerated their wound healing. The faster healing was also linked to the release of the TGFß family member GDF15 from efferocytic dendritic cells. In sum, SLC7A11 is a negative regulator of efferocytosis, and removing this brake improves wound healing, with important implications for wound management in diabetes.


Asunto(s)
Sistema de Transporte de Aminoácidos y+ , Células Dendríticas , Diabetes Mellitus , Fagocitosis , Cicatrización de Heridas , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Animales , Células Dendríticas/citología , Células Dendríticas/inmunología , Diabetes Mellitus/inmunología , Gluconeogénesis , Glucosa , Glucólisis , Factor 15 de Diferenciación de Crecimiento , Ratones
6.
Nat Commun ; 12(1): 5913, 2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34625556

RESUMEN

OTULIN is a deubiquitinase that specifically cleaves linear ubiquitin chains. Here we demonstrate that the ablation of Otulin selectively in keratinocytes causes inflammatory skin lesions that develop into verrucous carcinomas. Genetic deletion of Tnfr1, knockin expression of kinase-inactive Ripk1 or keratinocyte-specific deletion of Fadd and Mlkl completely rescues mice with OTULIN deficiency from dermatitis and tumorigenesis, thereby identifying keratinocyte cell death as the driving force for inflammation. Single-cell RNA-sequencing comparing non-lesional and lesional skin reveals changes in epidermal stem cell identity in OTULIN-deficient keratinocytes prior to substantial immune cell infiltration. Keratinocytes lacking OTULIN display a type-1 interferon and IL-1ß response signature, and genetic or pharmacologic inhibition of these cytokines partially inhibits skin inflammation. Finally, expression of a hypomorphic mutant Otulin allele, previously shown to cause OTULIN-related autoinflammatory syndrome in humans, induces a similar inflammatory phenotype, thus supporting the importance of OTULIN for restraining skin inflammation and maintaining immune homeostasis.


Asunto(s)
Endopeptidasas/metabolismo , Queratinocitos/metabolismo , Piel/metabolismo , Animales , Muerte Celular/genética , Citocinas/metabolismo , Endopeptidasas/genética , Proteína de Dominio de Muerte Asociada a Fas , Técnicas de Sustitución del Gen , Homeostasis , Inflamación/patología , Interferón Tipo I , Interleucina-1beta , Ratones , Necroptosis , Fragmentos de Péptidos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Piel/patología , Células Madre/metabolismo , Análisis de Sistemas , Ubiquitina/metabolismo
8.
Curr Biol ; 31(1): R20-R22, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33434480

RESUMEN

New work shows that the glycocalyx meshwork on the surface of macrophages prevents phagocytic receptors from binding their ligands by two means - electrostatic charge and steric hindrance. Components of this barrier are present on pathogenic and malignant targets that elude phagocytosis.


Asunto(s)
Glicocálix , Fagocitosis , Ligandos , Macrófagos , Fagocitos
9.
Nat Commun ; 11(1): 1817, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286311

RESUMEN

Dendritic cells (DCs) constitute a specialized population of immune cells that present exogenous antigen (Ag) on major histocompatibility complex (MHC) class I molecules to initiate CD8 + T cell responses against pathogens and tumours. Although cross-presentation depends critically on the trafficking of Ag-containing intracellular vesicular compartments, the molecular machinery that regulates vesicular transport is incompletely understood. Here, we demonstrate that mice lacking Kif5b (the heavy chain of kinesin-1) in their DCs exhibit a major impairment in cross-presentation and thus a poor in vivo anti-tumour response. We find that kinesin-1 critically regulates antigen cross-presentation in DCs, by controlling Ag degradation, the endosomal pH, and MHC-I recycling. Mechanistically, kinesin-1 appears to regulate early endosome maturation by allowing the scission of endosomal tubulations. Our results highlight kinesin-1's role as a molecular checkpoint that modulates the balance between antigen degradation and cross-presentation.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/metabolismo , Endosomas/metabolismo , Cinesinas/metabolismo , Ácidos/metabolismo , Animales , Antígenos/metabolismo , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Proliferación Celular , Endocitosis , Antígenos de Histocompatibilidad Clase I/metabolismo , Cinesinas/deficiencia , Ratones Noqueados , Ratones Transgénicos , Microtúbulos/metabolismo , Neoplasias/patología , Ovalbúmina/inmunología , Solubilidad
10.
J Cell Sci ; 133(5)2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32079661

RESUMEN

Toll-like receptor 7 (TLR7) is an endosomal receptor that recognizes single-stranded RNA from viruses. Its trafficking and activation is regulated by the endoplasmic reticulum (ER) chaperone UNC93B1 and lysosomal proteases. UNC93B1 also modulates major histocompatibility complex class II (MHCII) antigen presentation, and deficiency in MHCII protein diminishes TLR9 signaling. These results indicate a link between proteins that regulate both innate and adaptive responses. Here, we report that TLR7 resides in lysosomes and interacts with the MHCII-chaperone molecule, the invariant chain (Ii) or CD74, in B cells. In the absence of CD74, TLR7 displays both ER and lysosomal localization, leading to an increase in pro-inflammatory cytokine production. Furthermore, stimulation with TLR7 but not TLR9, is inefficient in boosting antigen presentation in Ii-deficient cells. In contrast, in B cells lacking TLR7 or mutated for UNC93B1, which are able to trigger TLR7 activation, antigen presentation is enhanced. This suggests that TLR7 signaling in B cells is controlled by the Ii chain.


Asunto(s)
Proteínas de Transporte de Membrana , Receptor Toll-Like 7 , Antígenos de Diferenciación de Linfocitos B/genética , Linfocitos B/metabolismo , Antígenos de Histocompatibilidad Clase II , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
11.
Methods Mol Biol ; 1988: 289-296, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31147948

RESUMEN

Proteases generate peptides that bind to MHC class II molecules to interact with a wide diversity of CD4+ T cells. They are expressed in dedicated organelles: endosomes and lysosomes of professional antigen-presenting cells (pAPCs) such as B cells, macrophages, and dendritic cells. The identification of endosomal proteases which produce antigenic peptides is important for example for better vaccination and to prevent autoimmune diseases. Here, we describe a panel of techniques (in vitro digestion assays of protein with recombinant proteases or purified endosomes/lysosomes, T cell stimulation) to monitor the production of MHC class II ligands.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/metabolismo , Biología Molecular/métodos , Péptido Hidrolasas/metabolismo , Animales , Presentación de Antígeno/inmunología , Electroforesis en Gel de Poliacrilamida , Endosomas/metabolismo , Ligandos , Lisosomas/metabolismo , Ratones , Biosíntesis de Proteínas
12.
Cell Mol Gastroenterol Hepatol ; 6(4): 477-493.e1, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30364784

RESUMEN

Background & Aims: Microvillus inclusion disease (MVID) is a congenital intestinal malabsorption disorder caused by defective apical vesicular transport. Existing cellular models do not fully recapitulate this heterogeneous pathology. The aim of this study was to characterize 3-dimensional intestinal organoids that continuously generate polarized absorptive cells as an accessible and relevant model to investigate MVID. Methods: Intestinal organoids from Munc18-2/Stxbp2-null mice that are deficient for apical vesicular transport were subjected to enterocyte-specific differentiation protocols. Lentiviral rescue experiments were performed using human MUNC18-2 variants. Apical trafficking and microvillus formation were characterized by confocal and transmission electron microscopy. Spinning disc time-lapse microscopy was used to document the lifecycle of microvillus inclusions. Results: Loss of Munc18-2/Stxbp2 recapitulated the pathologic features observed in patients with MUNC18-2 deficiency. The defects were fully restored by transgenic wild-type human MUNC18-2 protein, but not the patient variant (P477L). Importantly, we discovered that the MVID phenotype was correlated with the degree of enterocyte differentiation: secretory vesicles accumulated already in crypt progenitors, while differentiated enterocytes showed an apical tubulovesicular network and enlarged lysosomes. Upon prolonged enterocyte differentiation, cytoplasmic F-actin-positive foci were observed that further progressed into classic microvillus inclusions. Time-lapse microscopy showed their dynamic formation by intracellular maturation or invagination of the apical or basolateral plasma membrane. Conclusions: We show that prolonged enterocyte-specific differentiation is required to recapitulate the entire spectrum of MVID. Primary organoids can provide a powerful model for this heterogeneous pathology. Formation of microvillus inclusions from multiple membrane sources showed an unexpected dynamic of the enterocyte brush border.


Asunto(s)
Diferenciación Celular , Enterocitos/patología , Intestinos/patología , Síndromes de Malabsorción/metabolismo , Microvellosidades/patología , Mucolipidosis/metabolismo , Proteínas Munc18/deficiencia , Proteínas Munc18/metabolismo , Organoides/metabolismo , Actinas/metabolismo , Animales , Núcleo Celular/metabolismo , Enterocitos/metabolismo , Humanos , Lisosomas/metabolismo , Síndromes de Malabsorción/patología , Ratones Noqueados , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Mucolipidosis/patología , Organoides/patología , Organoides/ultraestructura
13.
Nat Commun ; 8(1): 1640, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29158474

RESUMEN

Dendritic cells (DC) have the unique ability to present exogenous antigens via the major histocompatibility complex class I pathway to stimulate naive CD8+ T cells. In DCs with a non-functional mutation in Unc93b1 (3d mutation), endosomal acidification, phagosomal maturation, antigen degradation, antigen export to the cytosol and the function of the store-operated-Ca2+-entry regulator STIM1 are impaired. These defects result in compromised antigen cross-presentation and anti-tumor responses in 3d-mutated mice. Here, we show that UNC93B1 interacts with the calcium sensor STIM1 in the endoplasmic reticulum, a critical step for STIM1 oligomerization and activation. Expression of a constitutively active STIM1 mutant, which no longer binds UNC93B1, restores antigen degradation and cross-presentation in 3d-mutated DCs. Furthermore, ablation of STIM1 in mouse and human cells leads to a decrease in cross-presentation. Our data indicate that the UNC93B1 and STIM1 cooperation is important for calcium flux and antigen cross-presentation in DCs.


Asunto(s)
Presentación de Antígeno , Calcio/metabolismo , Células Dendríticas/inmunología , Proteínas de Transporte de Membrana/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Animales , Antígenos/inmunología , Antígenos/metabolismo , Células Cultivadas , Reactividad Cruzada , Células Dendríticas/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/inmunología
14.
Nat Commun ; 8(1): 1852, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29176619

RESUMEN

Antigen cross-presentation by dendritic cells (DC) stimulates cytotoxic T cell activation to promote immunity to intracellular pathogens, viruses and cancer. Phagocytosed antigens generate potent T cell responses, but the signalling and trafficking pathways regulating their cross-presentation are unclear. Here, we show that ablation of the store-operated-Ca2+-entry regulator STIM1 in mouse myeloid cells impairs cross-presentation and DC migration in vivo and in vitro. Stim1 ablation reduces Ca2+ signals, cross-presentation, and chemotaxis in mouse bone-marrow-derived DCs without altering cell differentiation, maturation or phagocytic capacity. Phagosomal pH homoeostasis and ROS production are unaffected by STIM1 deficiency, but phagosomal proteolysis and leucyl aminopeptidase activity, IRAP recruitment, as well as fusion of phagosomes with endosomes and lysosomes are all impaired. These data suggest that STIM1-dependent Ca2+ signalling promotes the delivery of endolysosomal enzymes to phagosomes to enable efficient cross-presentation.


Asunto(s)
Presentación de Antígeno/fisiología , Células Dendríticas/fisiología , Fagosomas/fisiología , Molécula de Interacción Estromal 1/metabolismo , Animales , Calcio/metabolismo , Movimiento Celular/fisiología , Cistinil Aminopeptidasa/metabolismo , Células Dendríticas/inmunología , Retículo Endoplásmico/metabolismo , Concentración de Iones de Hidrógeno , Ratones Noqueados , Fagocitosis/fisiología , Fagosomas/química , Especies Reactivas de Oxígeno/metabolismo , Molécula de Interacción Estromal 1/genética
15.
Nat Immunol ; 18(5): 509-518, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28319098

RESUMEN

The retention of intracellular Toll-like receptors (TLRs) in the endoplasmic reticulum prevents their activation under basal conditions. TLR9 is activated by sensing ligands in specific endosomal-lysosomal compartments. Here we identified IRAP+ endosomes as major cellular compartments for the early steps of TLR9 activation in dendritic cells (DCs). Both TLR9 and its ligand, the dinucleotide CpG, were present as cargo in IRAP+ endosomes. In the absence of the aminopeptidase IRAP, the trafficking of CpG and TLR9 to lysosomes and signaling via TLR9 were enhanced in DCs and in mice following bacterial infection. IRAP stabilized CpG-containing endosomes by interacting with the actin-nucleation factor FHOD4, which slowed the trafficking of TLR9 toward lysosomes. Thus, endosomal retention of TLR9 via the interaction of IRAP with the actin cytoskeleton is a mechanism that prevents hyper-activation of TLR9 in DCs.


Asunto(s)
Cistinil Aminopeptidasa/metabolismo , Citoesqueleto/metabolismo , Células Dendríticas/fisiología , Endosomas/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Células Cultivadas , Islas de CpG/genética , Cistinil Aminopeptidasa/genética , Células Dendríticas/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Oligodesoxirribonucleótidos/inmunología , Unión Proteica , Transducción de Señal
16.
Blood ; 128(1): 60-71, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27222478

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening syndrome, characterized by severe hyperinflammation and immunopathological manifestations in several tissues. These features result from organ infiltration by overactivated CD8 T-cells and macrophages, which produce high levels of pro-inflammatory cytokines, such as IFN-γ, TNF-α, IL-6, and IL-18. Recently, several Janus kinase 1/2 (JAK1/2) inhibitors, such as ruxolitinib, have been developed as immunosuppressive agents. They have proven beneficial effects in the treatment of myeloproliferative disorders and inflammatory conditions. To determine whether pharmacological inhibition of the JAK1/2 not only prevents the onset of HLH immunopathology but also is effective against existing HLH, cytotoxicity-impaired Prf1(-/-) and Rab27a(-/-) mice with full-blown HLH syndrome were treated with a clinically relevant dose of ruxolitinib. In vivo, ruxolitinib treatment suppressed signal transducer and activator of transcription 1 activation and led to recovery from HLH manifestations in both murine models. In the Prf1(-/-) mice, these beneficial effects were evidenced by a greater survival rate, and in both murine models, they were evidenced by the correction of blood cytopenia and a rapid decrease in serum IL-6 and TNF-α levels. During ruxolitinib treatment, liver tissue damage receded concomitantly with a decrease in the number of infiltrating inflammatory macrophages and an increase in the number of alternatively activated macrophages. In Rab27a(-/-) mice, central nervous system involvement was significantly reduced by ruxolitinib therapy. Our findings demonstrate that clinically relevant doses of the JAK1/2 inhibitor ruxolitinib suppresses the harmful consequences of macrophage overactivation characterizing HLH in 2 murine models. The results could be readily translated into the clinic for the treatment of primary, and perhaps even secondary, forms of HLH.


Asunto(s)
Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Linfohistiocitosis Hemofagocítica/tratamiento farmacológico , Linfohistiocitosis Hemofagocítica/enzimología , Pirazoles/farmacología , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hígado/enzimología , Linfohistiocitosis Hemofagocítica/genética , Macrófagos/enzimología , Ratones , Ratones Noqueados , Nitrilos , Perforina/genética , Perforina/metabolismo , Pirimidinas , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas rab27 de Unión a GTP
17.
Blood ; 127(17): 2113-21, 2016 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-26864340

RESUMEN

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.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Herencia Multifactorial , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Qa-SNARE/genética , Proteínas de Unión al GTP rab/genética , Animales , Degranulación de la Célula/genética , Cruzamientos Genéticos , Citotoxicidad Inmunológica/genética , Dosificación de Gen , Predisposición Genética a la Enfermedad , Heterocigoto , Sinapsis Inmunológicas/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Mutación , Proteínas Citotóxicas Formadoras de Poros/fisiología , Proteínas Qa-SNARE/fisiología , Organismos Libres de Patógenos Específicos , Linfocitos T Citotóxicos/inmunología , Proteínas de Unión al GTP rab/fisiología , Proteínas rab27 de Unión a GTP
18.
F1000Res ; 4(F1000 Faculty Rev): 930, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26594351

RESUMEN

The granule-dependent cytotoxic activity of T and natural killer lymphocytes has progressively emerged as an important effector pathway not only for host defence but also for immune regulation. The analysis of an early-onset, severe, primary immune dysregulatory syndrome known as hemophagocytic lymphohistiocytosis (HLH) has been decisive in highlighting this latter role and identifying key effectors on the basis of gene mutation analyses and mediators in the maturation and secretion of cytotoxic granules. Studies of cytotoxicity-deficient murine counterparts have helped to define primary HLH as a syndrome in which uncontrolled T-cell activation in response to lymphocytic choriomeningitis virus infection results in excessive macrophage activation and inflammation-associated cytopenia. Recent recognition of late-onset HLH, which occurs in a variety of settings, in association with hypomorphic, monoallelic mutations in genes encoding components of the granule-dependent cytotoxic pathway or even in the absence of such mutations has broadened our view about the mechanisms that underlie the perturbation of immune homeostasis. These findings have led to the development of a model in which disease occurs when a threshold is reached through the accumulation of genetic and environmental risk factors. Nevertheless, validation of this model will require further investigations.

19.
Blood ; 125(9): 1427-34, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25525117

RESUMEN

The impairment of cytotoxic activity of lymphocytes disturbs immune surveillance and leads to the development of hemophagocytic lymphohistiocytic syndrome (HLH). Although cytotoxic T lymphocyte (CTL) control of HLH development is well documented, the role for natural killer (NK)-cell effector functions in the pathogenesis of this immune disorder remains unclear. In this study, we specifically targeted a defect in cytotoxicity to either CTL or NK cells in mice so as to dissect the contribution of these lymphocyte subsets to HLH-like disease severity after lymphocytic choriomeningitis virus (LCMV) infection. We found that NK-cell cytotoxicity was sufficient to protect mice from the fatal outcome that characterizes HLH-like disease and was also sufficient to reduce HLH-like manifestations. Mechanistically, NK-cell cytotoxicity reduced tissue infiltration by inflammatory macrophages and downmodulated LCMV-specific T-cell responses by limiting hyperactivation of CTL. Interestingly, the critical protective effect of NK cells on HLH was independent of interferon-γ secretion and changes in viral load. Therefore our findings identify a crucial role of NK-cell cytotoxicity in limiting HLH-like immunopathology, highlighting the important role of NK cytotoxic activity in immune homeostasis.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Proliferación Celular , Células Cultivadas , Interferón gamma/metabolismo , Células Asesinas Naturales/patología , Células Asesinas Naturales/virología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Linfohistiocitosis Hemofagocítica/patología , Linfohistiocitosis Hemofagocítica/virología , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Bazo/patología , Bazo/virología , Carga Viral
20.
J Clin Invest ; 124(12): 5516-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25401470

RESUMEN

Innate immunity to viral infection involves induction of the type I IFN response; however, dysfunctional regulation of this pathway leads to inappropriate inflammation. Here, we evaluated a nonconsanguineous family of mixed European descent, with 4 members affected by systemic inflammatory and autoimmune conditions, including lupus, with variable clinical expression. We identified a germline dominant gain-of-function mutation in TMEM173, which encodes stimulator of type I IFN gene (STING), in the affected individuals. STING is a key signaling molecule in cytosolic DNA-sensing pathways, and STING activation normally requires dimerization, which is induced by 2'3' cyclic GMP-AMP (cGAMP) produced by the cGAMP synthase in response to cytosolic DNA. Structural modeling supported constitutive activation of the mutant STING protein based on stabilized dimerization. In agreement with the model predictions, we found that the STING mutant spontaneously localizes in the Golgi of patient fibroblasts and is constitutively active in the absence of exogenous 2'3'-cGAMP in vitro. Accordingly, we observed elevated serum IFN activity and a type I IFN signature in peripheral blood from affected family members. These findings highlight the key role of STING in activating both the innate and adaptive immune responses and implicate aberrant STING activation in features of human lupus.


Asunto(s)
Enfermedades Genéticas Congénitas/inmunología , Lupus Eritematoso Sistémico/inmunología , Mutación , Multimerización de Proteína/inmunología , Transducción de Señal/inmunología , Inmunidad Adaptativa/genética , Adulto , Anciano , Preescolar , Femenino , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Humanos , Inmunidad Innata/genética , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Masculino , Proteínas de la Membrana , Nucleótidos Cíclicos/genética , Nucleótidos Cíclicos/inmunología , Multimerización de Proteína/genética , Transducción de Señal/genética , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA