Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Nat Immunol ; 24(1): 30-41, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36443515

RESUMO

Inflammasome complexes are pivotal in the innate immune response. The NLR family pyrin domain containing protein 3 (NLRP3) inflammasome is activated in response to a broad variety of cellular stressors. However, a primary and converging sensing mechanism by the NLRP3 receptor initiating inflammasome assembly remains ill defined. Here, we demonstrate that NLRP3 inflammasome activators primarily converge on disruption of endoplasmic reticulum-endosome membrane contact sites (EECS). This defect causes endosomal accumulation of phosphatidylinositol 4-phosphate (PI4P) and a consequent impairment of endosome-to-trans-Golgi network trafficking (ETT), necessary steps for endosomal recruitment of NLRP3 and subsequent inflammasome activation. Lowering endosomal PI4P levels prevents endosomal association of NLRP3 and inhibits inflammasome activation. Disruption of EECS or ETT is sufficient to enhance endosomal PI4P levels, to recruit NLRP3 to endosomes and to potentiate NLRP3 inflammasome activation. Mice with defects in ETT in the myeloid compartment are more susceptible to lipopolysaccharide-induced sepsis. Our study thus identifies a distinct cellular mechanism leading to endosomal NLRP3 recruitment and inflammasome activation.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Camundongos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Imunidade Inata , Proteínas de Transporte/metabolismo , Endossomos/metabolismo
2.
Cell ; 136(2): 235-48, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-19135240

RESUMO

Dysfunction and loss of insulin-producing pancreatic beta cells represent hallmarks of diabetes mellitus. Here, we show that mice lacking the mitogen-activated protein kinase (MAPK) p38delta display improved glucose tolerance due to enhanced insulin secretion from pancreatic beta cells. Deletion of p38delta results in pronounced activation of protein kinase D (PKD), the latter of which we have identified as a pivotal regulator of stimulated insulin exocytosis. p38delta catalyzes an inhibitory phosphorylation of PKD1, thereby attenuating stimulated insulin secretion. In addition, p38delta null mice are protected against high-fat-feeding-induced insulin resistance and oxidative stress-mediated beta cell failure. Inhibition of PKD1 reverses enhanced insulin secretion from p38delta-deficient islets and glucose tolerance in p38delta null mice as well as their susceptibility to oxidative stress. In conclusion, the p38delta-PKD pathway integrates regulation of the insulin secretory capacity and survival of pancreatic beta cells, pointing to a pivotal role for this pathway in the development of overt diabetes mellitus.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína Quinase 13 Ativada por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Animais , Exocitose , Feminino , Glucose/metabolismo , Complexo de Golgi/metabolismo , Secreção de Insulina , Masculino , Camundongos , Proteína Quinase 13 Ativada por Mitógeno/genética , Fosfolipases Tipo C/metabolismo
3.
EMBO Rep ; 21(7): e50645, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32419206

RESUMO

In contrast to highly specific sensor molecules of the innate immune system, the NLRP3 receptor detects a broad variety of danger signals including pathogens. Sensing triggers intracellular NLRP3 inflammasome complex assembly to induce an inflammatory response with the primary aim to eliminate pathogens. However, several of them have developed distinct strategies to hijack NLRP3-dependent immunity. In this issue of EMBO Reports, Zhang and colleagues demonstrate that reovirus infection of airway epithelial cells promotes EphA2-dependent phosphorylation of NLRP3 that impedes the recruitment of other inflammasome components necessary for its activation [1]. This potentially uncovers a mechanism that may lead to reduced viral clearance in the lung, eventually contributing to life-threatening respiratory disease.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas de Transporte/metabolismo , Células Epiteliais/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fosforilação
4.
EMBO Rep ; 20(9): e48235, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31353801

RESUMO

Caspase-4, the cytosolic LPS sensor, and gasdermin D, its downstream effector, constitute the non-canonical inflammasome, which drives inflammatory responses during Gram-negative bacterial infections. It remains unclear whether other proteins regulate cytosolic LPS sensing, particularly in human cells. Here, we conduct a genome-wide CRISPR/Cas9 screen in a human monocyte cell line to identify genes controlling cytosolic LPS-mediated pyroptosis. We find that the transcription factor, IRF2, is required for pyroptosis following cytosolic LPS delivery and functions by directly regulating caspase-4 levels in human monocytes and iPSC-derived monocytes. CASP4, GSDMD, and IRF2 are the only genes identified with high significance in this screen highlighting the simplicity of the non-canonical inflammasome. Upon IFN-γ priming, IRF1 induction compensates IRF2 deficiency, leading to robust caspase-4 expression. Deficiency in IRF2 results in dampened inflammasome responses upon infection with Gram-negative bacteria. This study emphasizes the central role of IRF family members as specific regulators of the non-canonical inflammasome.


Assuntos
Caspases Iniciadoras/metabolismo , Fator Regulador 2 de Interferon/metabolismo , Caspases Iniciadoras/genética , Morte Celular/efeitos dos fármacos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/fisiologia , Humanos , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 2 de Interferon/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Monócitos/metabolismo , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo , Células U937
5.
Gastroenterology ; 157(2): 537-551.e9, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30978357

RESUMO

BACKGROUND & AIMS: The mechanisms of hepatitis C virus (HCV) infection, liver disease progression, and hepatocarcinogenesis are only partially understood. We performed genomic, proteomic, and metabolomic analyses of HCV-infected cells and chimeric mice to learn more about these processes. METHODS: Huh7.5.1dif (hepatocyte-like cells) were infected with culture-derived HCV and used in RNA sequencing, proteomic, metabolomic, and integrative genomic analyses. uPA/SCID (urokinase-type plasminogen activator/severe combined immunodeficiency) mice were injected with serum from HCV-infected patients; 8 weeks later, liver tissues were collected and analyzed by RNA sequencing and proteomics. Using differential expression, gene set enrichment analyses, and protein interaction mapping, we identified pathways that changed in response to HCV infection. We validated our findings in studies of liver tissues from 216 patients with HCV infection and early-stage cirrhosis and paired biopsy specimens from 99 patients with hepatocellular carcinoma, including 17 patients with histologic features of steatohepatitis. Cirrhotic liver tissues from patients with HCV infection were classified into 2 groups based on relative peroxisome function; outcomes assessed included Child-Pugh class, development of hepatocellular carcinoma, survival, and steatohepatitis. Hepatocellular carcinomas were classified according to steatohepatitis; the outcome was relative peroxisomal function. RESULTS: We quantified 21,950 messenger RNAs (mRNAs) and 8297 proteins in HCV-infected cells. Upon HCV infection of hepatocyte-like cells and chimeric mice, we observed significant changes in levels of mRNAs and proteins involved in metabolism and hepatocarcinogenesis. HCV infection of hepatocyte-like cells significantly increased levels of the mRNAs, but not proteins, that regulate the innate immune response; we believe this was due to the inhibition of translation in these cells. HCV infection of hepatocyte-like cells increased glucose consumption and metabolism and the STAT3 signaling pathway and reduced peroxisome function. Peroxisomes mediate ß-oxidation of very long-chain fatty acids; we found intracellular accumulation of very long-chain fatty acids in HCV-infected cells, which is also observed in patients with fatty liver disease. Cells in livers from HCV-infected mice had significant reductions in levels of the mRNAs and proteins associated with peroxisome function, indicating perturbation of peroxisomes. We found that defects in peroxisome function were associated with outcomes and features of HCV-associated cirrhosis, fatty liver disease, and hepatocellular carcinoma in patients. CONCLUSIONS: We performed combined transcriptome, proteome, and metabolome analyses of liver tissues from HCV-infected hepatocyte-like cells and HCV-infected mice. We found that HCV infection increases glucose metabolism and the STAT3 signaling pathway and thereby reduces peroxisome function; alterations in the expression levels of peroxisome genes were associated with outcomes of patients with liver diseases. These findings provide insights into liver disease pathogenesis and might be used to identify new therapeutic targets.


Assuntos
Hepacivirus/patogenicidade , Hepatite C Crônica/patologia , Hepatócitos/patologia , Fígado/patologia , Animais , Linhagem Celular Tumoral , Conjuntos de Dados como Assunto , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Glucose/metabolismo , Hepatite C Crônica/metabolismo , Hepatite C Crônica/virologia , Hepatócitos/transplante , Hepatócitos/virologia , Humanos , Fígado/citologia , Fígado/virologia , Metabolômica , Camundongos , Peroxissomos/metabolismo , Peroxissomos/patologia , Proteômica , Fator de Transcrição STAT3/metabolismo , Quimeras de Transplante
6.
Diabetes Obes Metab ; 20 Suppl 2: 104-115, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230186

RESUMO

Regulated insulin secretion from pancreatic ß-cells is a major process maintaining glucose homeostasis in mammals. Enhancing insulin release in response to chronic nutrient overload and obesity-related insulin resistance (pre-diabetes) requires several adaptive cellular mechanisms maintaining ß-cell health under such stresses. Once these mechanisms are overwhelmed, ß-cell failure occurs leading to full-blown Type 2 Diabetes (T2D). Nutrient-dependent macroautophagy represents one such adaptive mechanism in ß-cells. While macroautophagy levels are high and protective in ß-cells in pre-diabetes, they decrease at later stages contributing to ß-cell failure. However, mechanisms compromising macroautophagy in ß-cells remain poorly understood. In this review, we discuss how recently discovered signalling cascades that emanate from the limiting membrane of lysosomes contribute to changes in macroautophagy flux in physiology and disease. In particular, these mechanisms are put into context with ß-cell function highlighting most recently described links between nutrient-dependent lysosomal signalling pathways and insulin secretion. Understanding these mechanisms in response to metabolic stress might pave the way for development of more tailored treatment strategies aimed at preserving ß-cell health.


Assuntos
Células Secretoras de Insulina/fisiologia , Lisossomos/fisiologia , Nutrientes/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Autofagia/fisiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Metabolismo Energético/fisiologia , Humanos , Insulina/metabolismo , Secreção de Insulina/fisiologia , Membranas Intracelulares/enzimologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais/fisiologia
7.
Handb Exp Pharmacol ; 233: 195-220, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25903413

RESUMO

Low-grade inflammation is an established pathological condition that contributes to the development of obesity, insulin resistance and type 2 diabetes. Metabolic inflammation is dependent on multiple signalling events. In an overnutrition state, canonical inflammatory pathways are induced by inflammatory cytokines and lipid species. They can also be triggered through inflammasome activation as well as through cellular stress provoked by the unfolded protein response at the endoplasmic reticulum as well as by reactive oxygen species. In this chapter, we summarize the current knowledge about signalling events within the cell and describe how they impact on metabolic inflammation and whole-body metabolism. We particularly highlight the interplay between different signalling pathways that link low-grade inflammation responses to the inactivation of the insulin receptor pathway, ultimately leading to insulin resistance, a hallmark of type 2 diabetes.


Assuntos
Inflamação/etiologia , Transdução de Sinais/fisiologia , Animais , Citocinas/fisiologia , Humanos , Quinase I-kappa B/fisiologia , Resistência à Insulina , Sistema de Sinalização das MAP Quinases , Ativação de Macrófagos
8.
Eur J Immunol ; 43(3): 716-22, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23254454

RESUMO

Interleukin (IL)-1α is a potent proinflammatory cytokine that has been implicated in the development of atherosclerosis. We investigated whether a vaccine inducing IL-1α neutralizing antibodies could interfere with disease progression in a murine model of atherosclerosis. We immunized Apolipoprothin E (ApoE)-deficient mice with a vaccine (IL-1α-C-Qß) consisting of full-length, native IL-1α chemically conjugated to virus-like particles derived from the bacteriophage Qß. ApoE(-/-) mice were administered six injections of IL-1α-C-Qß or nonconjugated Qß over a period of 160 days while being maintained on a western diet. Atherosclerosis was measured in the descending aorta and in cross-sections at the aortic root. Macrophage infiltration in the aorta was measured using CD68. Expression levels of VCAM-1, ICAM-1, and MCP-1 were quantified by RT-PCR. Immunization against IL-1α reduced plaque progression in the descending aorta by 50% and at the aortic root by 37%. Macrophage infiltration in the aorta was reduced by 22%. Inflammation was also reduced in the adventitia, with a decrease of 54% in peri-aortic infiltrate score and reduced expression levels of VCAM-1 and ICAM-1. Active immunization targeting IL-1α reduced both the inflammatory reaction in the plaque as well as plaque progression. In summary, vaccination against IL-1α protected ApoE(-/-) mice against disease, suggesting that this may be a potential treatment option for atherosclerosis.


Assuntos
Aterosclerose/imunologia , Interleucina-1alfa/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Anticorpos/imunologia , Anticorpos Neutralizantes/imunologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/prevenção & controle , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Placa Aterosclerótica/genética , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem
9.
EMBO Rep ; 13(11): 1012-20, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-22964757

RESUMO

Adipose tissue is the largest compartment in the mammalian body for storing energy as fat, providing an important reservoir of fuel for maintaining whole body energy homeostasis. Herein, we identify the transcriptional cofactor hairless (HR) to be required for white adipogenesis. Moreover, forced expression of HR in non-adipogenic precursor cells induces adipogenic gene expression and enhances adipocyte formation under permissive conditions. HR exerts its proadipogenic effects by regulating the expression of PPARγ, one of the central adipogenic transcription factors. In conclusion, our data provide a new mechanism required for white adipogenesis.


Assuntos
Adipócitos Brancos/citologia , Adipogenia/genética , Regulação da Expressão Gênica no Desenvolvimento , PPAR gama/metabolismo , Fatores de Transcrição/metabolismo , Células 3T3 , Adipócitos Brancos/metabolismo , Animais , Diferenciação Celular , Camundongos , Camundongos Knockout , Mutação , PPAR gama/genética , Fatores de Transcrição/genética , Transcrição Gênica
10.
Nat Genet ; 30(2): 158-66, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11818961

RESUMO

The Jun and JunB components of the AP-1 transcription factor are known to have antagonistic functions. Here we show, by a knock-in strategy and a transgenic complementation approach, that Junb can substitute for absence of Jun during mouse development. Junb can rescue both liver and cardiac defects in Jun-null mice in a manner dependent on gene dosage. JunB restores the expression of genes regulated by Jun/Fos, but not those regulated by Jun/ATF, thereby rescuing Jun-dependent defects in vivo as well as in primary fibroblasts and fetal hepatoblasts in vitro. Thus, the transcriptionally less active JunB has the potential to substitute for Jun, indicating that the spatial and temporal regulation of expression of the transcription factor AP-1 may be more important than the coding sequence of its components.


Assuntos
Divisão Celular/genética , Divisão Celular/fisiologia , Desenvolvimento Embrionário e Fetal/genética , Desenvolvimento Embrionário e Fetal/fisiologia , Genes jun , Proteínas Proto-Oncogênicas c-jun/fisiologia , Animais , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes fos , Cardiopatias Congênitas/genética , Fígado/anormalidades , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/fisiologia , Proteína Supressora de Tumor p53/metabolismo
11.
Sci Immunol ; 8(90): eadf4699, 2023 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-38134241

RESUMO

Immune cells sense the microenvironment to fine-tune their inflammatory responses. Patients with cryopyrin-associated periodic syndrome (CAPS), caused by mutations in the NLRP3 gene, develop autoinflammation triggered by nonantigenic cues such as from the environment. However, the underlying mechanisms are poorly understood. Here, we uncover that KCNN4, a calcium-activated potassium channel, links PIEZO-mediated mechanotransduction to NLRP3 inflammasome activation. Yoda1, a PIEZO1 agonist, lowered the threshold for NLRP3 inflammasome activation. PIEZO-mediated sensing of stiffness and shear stress increased NLRP3-dependent inflammation. Myeloid-specific deletion of PIEZO1/2 protected mice from gouty arthritis. Mechanistically, activation of PIEZO1 triggers calcium influx, which activates KCNN4 to evoke potassium efflux and promotes NLRP3 inflammasome activation. Activation of PIEZO signaling was sufficient to activate the inflammasome in cells expressing CAPS-causing NLRP3 mutants via KCNN4. Last, pharmacological inhibition of KCNN4 alleviated autoinflammation in cells of patients with CAPS and in mice bearing a CAPS mutation. Thus, PIEZO-dependent mechanical inputs boost inflammation in NLRP3-dependent diseases, including CAPS.


Assuntos
Síndromes Periódicas Associadas à Criopirina , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Animais , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Mecanotransdução Celular , Síndromes Periódicas Associadas à Criopirina/genética , Inflamação , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Canais Iônicos/genética
12.
Nat Metab ; 5(6): 1045-1058, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37277610

RESUMO

Hypothalamic AgRP/NPY neurons are key players in the control of feeding behaviour. Ghrelin, a major orexigenic hormone, activates AgRP/NPY neurons to stimulate food intake and adiposity. However, cell-autonomous ghrelin-dependent signalling mechanisms in AgRP/NPY neurons remain poorly defined. Here we show that calcium/calmodulin-dependent protein kinase ID (CaMK1D), a genetic hot spot in type 2 diabetes, is activated upon ghrelin stimulation and acts in AgRP/NPY neurons to mediate ghrelin-dependent food intake. Global Camk1d-knockout male mice are resistant to ghrelin, gain less body weight and are protected against high-fat-diet-induced obesity. Deletion of Camk1d in AgRP/NPY, but not in POMC, neurons is sufficient to recapitulate above phenotypes. In response to ghrelin, lack of CaMK1D attenuates phosphorylation of CREB and CREB-dependent expression of the orexigenic neuropeptides AgRP/NPY in fibre projections to the paraventricular nucleus (PVN). Hence, CaMK1D links ghrelin action to transcriptional control of orexigenic neuropeptide availability in AgRP neurons.


Assuntos
Diabetes Mellitus Tipo 2 , Grelina , Camundongos , Animais , Masculino , Grelina/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Camundongos Knockout , Ingestão de Alimentos , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/metabolismo
13.
Dev Cell ; 12(6): 887-900, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17543862

RESUMO

Faithful cell-cycle progression is tightly controlled by the ubiquitin-proteasome system. Here we identify a human Cullin 3-based E3 ligase (Cul3) which is essential for mitotic division. In a complex with the substrate-specific adaptors KLHL9 and KLHL13, Cul3 is required for correct chromosome alignment in metaphase, proper midzone and midbody formation, and completion of cytokinesis. This Cul3-based E3 ligase removes components of the chromosomal passenger complex from mitotic chromosomes and allows their accumulation on the central spindle during anaphase. Aurora B directly binds to the substrate-recognition domain of KLHL9 and KLHL13 in vitro, and coimmunoprecipitates with the Cul3 complex during mitosis. Moreover, Aurora B is ubiquitylated in a Cul3-dependent manner in vivo, and by reconstituted Cul3/KLHL9/KLHL13 ligase in vitro. We thus propose that the Cul3/KLHL9/KLHL13 E3 ligase controls the dynamic behavior of Aurora B on mitotic chromosomes, and thereby coordinates faithful mitotic progression and completion of cytokinesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromossomos Humanos , Proteínas Culina/metabolismo , Citocinese , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Aurora Quinase B , Aurora Quinases , Proteínas de Ciclo Celular/genética , Proteínas Culina/genética , Células HeLa , Humanos , Proteínas Serina-Treonina Quinases/genética , Fuso Acromático , Ubiquitina/metabolismo , Complexos Ubiquitina-Proteína Ligase/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
14.
EMBO Rep ; 11(11): 834-40, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20930846

RESUMO

Mitogen-activated protein kinase (MAPK) signalling occurs in response to almost any change in the extracellular or intracellular milieu that affects the metabolism of the cell, organ or the entire organism. MAPK-dependent signal transduction is required for physiological metabolic adaptation, but inappropriate MAPK signalling contributes to the development of several interdependent pathological traits, collectively known as metabolic syndrome. Metabolic syndrome leads to life-threatening clinical consequences, such as type 2 diabetes. This Review provides an overview of the MAPK-signalling mechanisms that underly basic cellular metabolism, discussing their link to disease.


Assuntos
Células/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estresse Fisiológico , Adipócitos/enzimologia , Animais , Células/enzimologia , Humanos , Insulina/metabolismo
15.
Nat Med ; 11(12): 1330-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16311603

RESUMO

Abdominal aortic aneurysm (AAA) is a common disease among elderly people that, when surgical treatment is inapplicable, results in progressive expansion and rupture of the aorta with high mortality. Although nonsurgical treatment for AAA is much awaited, few options are available because its molecular pathogenesis remains elusive. Here, we identify JNK as a proximal signaling molecule in the pathogenesis of AAA. Human AAA tissue showed a high level of phosphorylated JNK. We show that JNK programs a gene expression pattern in different cell types that cooperatively enhances the degradation of the extracellular matrix while suppressing biosynthetic enzymes of the extracellular matrix. Selective inhibition of JNK in vivo not only prevented the development of AAA but also caused regression of established AAA in two mouse models. Thus, JNK promotes abnormal extracellular matrix metabolism in the tissue of AAA and may represent a therapeutic target.


Assuntos
Antracenos/farmacologia , Aneurisma da Aorta Abdominal/tratamento farmacológico , Aneurisma da Aorta Abdominal/prevenção & controle , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Adenoviridae , Animais , Aorta/química , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Proteínas da Matriz Extracelular/biossíntese , Vetores Genéticos , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Análise em Microsséries , Extratos de Tecidos/metabolismo
16.
EMBO Mol Med ; 14(4): e14841, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35263037

RESUMO

Small-Cell Lung Cancer (SCLC) is an aggressive neuroendocrine malignancy with a poor prognosis. Here, we focus on the neuroendocrine SCLC subtypes, SCLC-A and SCLC-N, whose transcription addiction was driven by ASCL1 and NEUROD1 transcription factors which target E-box motifs to activate up to 40% of total genes, the promoters of which are maintained in a steadily open chromatin environment according to ATAC and H3K27Ac signatures. This leverage is used by the marine agent lurbinectedin, which preferentially targets the CpG islands located downstream of the transcription start site, thus arresting elongating RNAPII and promoting its degradation. This abrogates the expression of ASCL1 and NEUROD1 and of their dependent genes, such as BCL2, INSM1, MYC, and AURKA, which are responsible for relevant SCLC tumorigenic properties such as inhibition of apoptosis and cell survival, as well as for a part of its neuroendocrine features. In summary, we show how the transcription addiction of these cells becomes their Achilles's heel, and how this is effectively exploited by lurbinectedin as a novel SCLC therapeutic endeavor.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Carbolinas , Compostos Heterocíclicos de 4 ou mais Anéis , Neoplasias Pulmonares , Proteínas Repressoras , Carcinoma de Pequenas Células do Pulmão , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carbolinas/farmacologia , Linhagem Celular Tumoral , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo
17.
J Cell Biol ; 172(6): 847-60, 2006 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-16533945

RESUMO

Separase is a protease whose liberation from its inhibitory chaperone Securin triggers sister chromatid disjunction at anaphase onset in yeast by cleaving cohesin's kleisin subunit. We have created conditional knockout alleles of the mouse Separase and Securin genes. Deletion of both copies of Separase but not Securin causes embryonic lethality. Loss of Securin reduces Separase activity because deletion of just one copy of the Separase gene is lethal to embryos lacking Securin. In embryonic fibroblasts, Separase depletion blocks sister chromatid separation but does not prevent other aspects of mitosis, cytokinesis, or chromosome replication. Thus, fibroblasts lacking Separase become highly polyploid. Hepatocytes stimulated to proliferate in vivo by hepatectomy also become unusually large and polyploid in the absence of Separase but are able to regenerate functional livers. Separase depletion in bone marrow causes aplasia and the presumed death of hematopoietic cells other than erythrocytes. Destruction of sister chromatid cohesion by Separase may be a universal feature of mitosis in eukaryotic cells.


Assuntos
Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Segregação de Cromossomos/genética , Período de Replicação do DNA/genética , Endopeptidases/genética , Mitose/genética , Anáfase/genética , Animais , Proteínas de Transporte/genética , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Desenvolvimento Embrionário/genética , Feminino , Fibroblastos , Genes Letais/genética , Células-Tronco Hematopoéticas/metabolismo , Hepatócitos , Regeneração Hepática/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Poliploidia , Securina , Separase , Coesinas
18.
Nat Med ; 9(12): 1484-90, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14625544

RESUMO

Genetic susceptibility and autoimmunity triggered by microbial infections are factors implicated in the pathogenesis of dilated cardiomyopathy, the most common cause of heart failure in young patients. Here we show that dendritic cells (DCs) loaded with a heart-specific self peptide induce CD4+ T-cell-mediated myocarditis in nontransgenic mice. Toll-like receptor (TLR) stimulation, in concert with CD40 triggering of self peptide-loaded dendritic cells, was shown to be required for disease induction. After resolution of acute myocarditis, DC-immunized mice developed heart failure, and TLR stimulation of these mice resulted in relapse of inflammatory infiltrates. Injection of damaged, syngeneic cardiomyocytes also induced myocarditis in mice if TLRs were activated in vivo. DC-induced myocarditis provides a unifying theory as to how tissue damage and activation of TLRs during infection can induce autoimmunity, relapses and cardiomyopathy.


Assuntos
Autoimunidade , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/imunologia , Células Dendríticas/imunologia , Adaptação Fisiológica , Animais , Autoantígenos/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/metabolismo , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Humanos , Imunidade Inata , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Modelos Imunológicos , Fragmentos de Peptídeos/imunologia , Receptores de Superfície Celular/metabolismo , Receptores Toll-Like , Miosinas Ventriculares/imunologia
19.
Cell Rep ; 35(7): 109129, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34010649

RESUMO

Mitochondria are highly dynamic organelles subjected to fission and fusion events. During mitosis, mitochondrial fission ensures equal distribution of mitochondria to daughter cells. If and how this process can actively drive mitotic progression remains largely unknown. Here, we discover a pathway linking mitochondrial fission to mitotic progression in mammalian cells. The mitochondrial fission factor (MFF), the main mitochondrial receptor for the Dynamin-related protein 1 (DRP1), is directly phosphorylated by Protein Kinase D (PKD) specifically during mitosis. PKD-dependent MFF phosphorylation is required and sufficient for mitochondrial fission in mitotic but not in interphasic cells. Phosphorylation of MFF is crucial for chromosome segregation and promotes cell survival by inhibiting adaptation of the mitotic checkpoint. Thus, PKD/MFF-dependent mitochondrial fission is critical for the maintenance of genome integrity during cell division.


Assuntos
Proteínas Mitocondriais/genética , Mitose/fisiologia , Proteína Quinase C/metabolismo , Animais , Humanos , Camundongos , Transdução de Sinais
20.
Nat Commun ; 12(1): 5862, 2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34615873

RESUMO

NLRP3 controls the secretion of inflammatory cytokines IL-1ß/18 and pyroptosis by assembling the inflammasome. Upon coordinated priming and activation stimuli, NLRP3 recruits NEK7 within hetero-oligomers that nucleate ASC and caspase-1 filaments, but the apical molecular mechanisms underlying inflammasome assembly remain elusive. Here we show that NEK7 recruitment to NLRP3 is controlled by the phosphorylation status of NLRP3 S803 located within the interaction surface, in which NLRP3 S803 is phosphorylated upon priming and later dephosphorylated upon activation. Phosphomimetic substitutions of S803 abolish NEK7 recruitment and inflammasome activity in macrophages in vitro and in vivo. In addition, NLRP3-NEK7 binding is also essential for NLRP3 deubiquitination by BRCC3 and subsequently inflammasome assembly, with NLRP3 phosphomimetic mutants showing enhanced ubiquitination and degradation than wildtype NLRP3. Finally, we identify CSNK1A1 as the kinase targeting NLRP3 S803. Our findings thus reveal NLRP3 S803 phosphorylation status as a druggable apical molecular mechanism controlling inflammasome assembly.


Assuntos
Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/química , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Caseína Quinase II , Caseína Quinase Ialfa , Caspase 1/metabolismo , Citocinas/metabolismo , Enzimas Desubiquitinantes , Células HEK293 , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Quinases Relacionadas a NIMA/metabolismo , Fosforilação , Piroptose , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA