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.
Int Immunopharmacol ; 100: 108106, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34530204

RESUMO

BACKGROUND: NOD-like receptor protein 3 (NLRP3) inflammasome activation has emerged as a crucial contributor to sepsis-induced lung injury. Geranylgeranyl diphosphate synthase 1 (GGPPS1) reportedly exerts the pro-inflammatory capability via activation of NLRP3 inflammasome. However, little is known about the role and mechanism of GGPPS1 in sepsis-induced lung injury. METHODS: Mice underwent cecal ligation and puncture (CLP) surgery to establish the in vivo model of sepsis. The lung injury of mice was assessed by analyzing the histological changes, the lung wet/dry ratio, PaO2/FiO2 ratio, myeloperoxidase (MPO) activity, total protein content, total cell, and polymorphonuclear leukocyte counts. Mouse alveolar macrophages MH-S were exposed to LPS for developing in vitro model of sepsis. The mRNA and protein expression levels of GGPPS1, beclin-1, and autophagy and inflammasome-related genes were detected using quantitative reverse transcription-polymerase chain reaction and western blot assays. Enzyme-linked immunosorbent assay was conducted to determine the levels of interleukin (IL)-1ß and IL-18. RESULTS: We successfully established sepsis-induced acute lung injury in vivo by CLP surgery. GGPPS1 was upregulated in the lung tissues of CLP-induced septic mice. The activation of autophagy and NLRP3 inflammasome were found in the lung tissues of CLP-induced septic mice. The addition of exogenous GGPP (synthesis products catalyzed by GGPPS1) and autophagic inhibitor 3-MA aggravated sepsis-induced hypoxemia, alveolar inflammatory response, intrapulmonary hemorrhage, and pulmonary edema, as evidenced by increased lung injury score, lung wet/dry weight ratio, MPO activity, total protein content, total cell, and PMNs counts, and decreased PaO2/FiO2 ratio. While NLRP3 inhibitor MCC950 exerted the opposite effects. Additionally, administration of exogenous GGPP could inhibit the activation of autophagy, enhance the activity of NLRP3 inflammasome, and the production of IL-1ß and IL-18. Inhibition of autophagy by 3-MA treatment also promoted the activity of NLRP3 inflammasome and the production of IL-1ß and IL-18. While MCC950 restrained the activity of NLRP3 inflammasome, but did not affect the activation of autophagy. Notably, the expression of GGPPS1 was unaltered in CLP-induced mice following GGPP, 3-MA, or MCC950 treatment. Moreover, GGPPS1 was upregulated in MH-S cells stimulated with LPS, and GGPPS1 knockdown enhanced the activation of autophagy and inhibited the activity of NLRP3 inflammasome in vitro. Importantly, depletion of GGPPS1 could alleviate LPS-induced inflammatory response by inducing autophagy-dependent NLRP3 inflammasome inhibition. CONCLUSION: GGPPS1 knockdown suppressed NLRP3 inflammasome activity via promoting autophagy and then attenuated sepsis-induced acute lung injury, revealing a novel target for treating sepsis-induced lung injury.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Autofagia , Farnesiltranstransferase/deficiência , Inflamassomos/metabolismo , Pulmão/enzimologia , Macrófagos Alveolares/enzimologia , Complexos Multienzimáticos/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Sepse/enzimologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/prevenção & controle , Adenina/análogos & derivados , Adenina/toxicidade , Animais , Anti-Inflamatórios/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Farnesiltranstransferase/genética , Furanos/farmacologia , Técnicas de Silenciamento de Genes , Indenos/farmacologia , Inflamassomos/antagonistas & inibidores , Inflamassomos/genética , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Camundongos Endogâmicos C57BL , Complexos Multienzimáticos/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fosfatos de Poli-Isoprenil/toxicidade , Sepse/imunologia , Sepse/patologia , Sepse/prevenção & controle , Transdução de Sinais , Sulfonamidas/farmacologia
2.
J Mol Cell Biol ; 13(2): 116-127, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33340314

RESUMO

Dystocia is a serious problem for pregnant women, and it increases the cesarean section rate. Although uterine dysfunction has an unknown etiology, it is responsible for cesarean delivery and clinical dystocia, resulting in neonatal morbidity and mortality; thus, there is an urgent need for novel therapeutic agents. Previous studies indicated that statins, which inhibit the mevalonate (MVA) pathway of cholesterol synthesis, can reduce the incidence of preterm birth, but the safety of statins for pregnant women has not been thoroughly evaluated. Therefore, to unambiguously examine the function of the MVA pathway in pregnancy and delivery, we employed a genetic approach by using myometrial cell-specific deletion of geranylgeranyl pyrophosphate synthase (Ggps1) mice. We found that Ggps1 deficiency in myometrial cells caused impaired uterine contractions, resulting in disrupted embryonic placing and dystocia. Studies of the underlying mechanism suggested that Ggps1 is required for uterine contractions to ensure successful parturition by regulating RhoA prenylation to activate the RhoA/Rock2/p-MLC pathway. Our work indicates that perturbing the MVA pathway might result in problems during delivery for pregnant females, but modifying protein prenylation with supplementary farnesyl pyrophosphate or geranylgeranyl pyrophosphate might be a strategy to avoid side effects.


Assuntos
Distocia/etiologia , Distocia/fisiopatologia , Farnesiltranstransferase/deficiência , Predisposição Genética para Doença , Complexos Multienzimáticos/deficiência , Contração Uterina/genética , Animais , Biomarcadores , Modelos Animais de Doenças , Distocia/metabolismo , Farnesiltranstransferase/metabolismo , Feminino , Estudos de Associação Genética , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Infertilidade/genética , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/metabolismo , Organogênese/genética , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Fenótipo , Gravidez , Ligação Proteica , Transdução de Sinais , Útero/embriologia , Útero/metabolismo , Útero/fisiopatologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Biol Chem ; 400(12): 1617-1627, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31120854

RESUMO

Geranylgeranyl diphosphate synthase (GGPPS) is an enzyme that catalyzes the synthesis of geranylgeranyl pyrophosphate (GGPP). GGPPS is implicated in many disorders, but its role in idiopathic pulmonary fibrosis (IPF) remains unclear. This study aimed to investigate the role of GGPPS in IPF. We established bleomycin-induced lung injury in a lung-specific GGPPS-deficient mouse (GGPPS-/-) and detected GGPPS expression in lung tissues by Western blot and immunohistochemistry analysis. We found that GGPPS expression increased during lung injury and fibrosis in mice induced by bleomycin, and GGPPS deficiency augmented lung fibrosis. GGPPS deficiency activated lung fibroblast by facilitating transforming growth factor ß1 while antagonizing bone morphogenetic protein 4 signaling. Notably, the supplementation of exogenous GGPP mitigated lung fibrosis in GGPPS-/- mice induced by bleomycin. In conclusion, our findings suggest that GGPPS provides protection against pulmonary fibrosis and that the restoration of protein geranylgeranylation may benefit statin-induced lung injury.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Farnesiltranstransferase/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Complexos Multienzimáticos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Farnesiltranstransferase/deficiência , Inibidores de Hidroximetilglutaril-CoA Redutases , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos/deficiência
4.
J Pathol ; 246(3): 277-288, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29971772

RESUMO

Patients with obesity have a high prevalence of non-alcoholic fatty liver disease (NAFLD) and, in parallel, increased susceptibility to fibrosis/cirrhosis/hepatocellular carcinoma (HCC). Herein, we report that a high-fat diet (HFD) can augment glycolysis and then accelerate NAFLD-fibrosis progression by downregulating the expression of geranylgeranyl diphosphate synthase (GGPPS), which is a critical enzyme in the mevalonate pathway. Long-term HFD overloading decreases GGPPS expression in mice, which shifts the fuel preference from fatty acids towards glucose. Liver-specific Ggpps deficiency drives the Warburg effect by impairing mitochondrial function, and then induces hepatic inflammation, thus exacerbating fibrosis. Ggpps deficiency also enhances the hyperfarnesylation of liver kinase B1, and promotes metabolic reprogramming by regulating 5'-AMP-activated protein kinase activity. Clinical data further imply that GGPPS expression can predict the stage of NAFLD and recurrence of NAFLD-associated HCC. We conclude that the level of GGPPS is a susceptibility factor for NAFLD-fibrosis progression, and requires more stringent surveillance to ensure early prediction and precision of treatment of NAFLD-related HCC. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Metabolismo Energético , Farnesiltranstransferase/metabolismo , Ácidos Graxos/metabolismo , Glucose/metabolismo , Hepatócitos/enzimologia , Cirrose Hepática/enzimologia , Fígado/enzimologia , Hepatopatia Gordurosa não Alcoólica/enzimologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Farnesiltranstransferase/deficiência , Farnesiltranstransferase/genética , Glicólise , Hepatócitos/patologia , Humanos , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Hepáticas/patologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Oxirredução , Prenilação de Proteína , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
5.
J Bone Miner Metab ; 36(2): 133-147, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28357594

RESUMO

Bone fracture healing is achieved through the proliferation and differentiation of stem cells, while bone marrow stem cells (BMSCs) contribute to endochondral ossification. During fracture healing, mesenchymal progenitor cells first form a cartilaginous blastema that becomes vascularized to recruit precursor cells of osteoblasts through the bone morphogenetic protein 2 (Bmp2)/Smad-dependent Runx2 pathway. Statins deplete geranylgeranyl diphosphate (GGPP), which participates in the regulation of BMSCs differentiation, through the inhibition of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase, leading to impaired protein geranylgeranylation, which strongly impacts the bone synthesis induced by Bmp2. Accordingly, we would like to investigate the role of geranylgeranyl diphosphate synthase 1 (Ggps1) in bone fracture via endochondral ossification in mice. We used a Cre-loxP system, namely the tamoxifen-inducible Collagen 2-CreERT2 Ggps1 fl/fl, to eliminate specifically the Ggps1 activity in chondrocytes of 8-10-week-old mice. We found that the endochondral bone formation, calcification and vasculogenesis of the bony callus were accelerated in fractures in Ggps1-/-mice. Together, the results of this study confirm that the specific deletion of Ggps1, using the Collagen 2-CreERT2 mice, will accelerate the fracture healing process by activating the Bmp2/Smad-dependent Runx2 pathway. In addition, we managed to improve the fracture healing process by inhibiting the Ggps1 activity and its related products with statin drugs.


Assuntos
Condrócitos/metabolismo , Farnesiltranstransferase/metabolismo , Consolidação da Fratura , Técnicas de Silenciamento de Genes , Complexos Multienzimáticos/metabolismo , Osteogênese , Animais , Biomarcadores/metabolismo , Fenômenos Biomecânicos , Proteína Morfogenética Óssea 2/metabolismo , Calo Ósseo/metabolismo , Calo Ósseo/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Farnesiltranstransferase/deficiência , Fêmur/irrigação sanguínea , Fêmur/diagnóstico por imagem , Fêmur/patologia , Fêmur/fisiopatologia , Consolidação da Fratura/genética , Regulação da Expressão Gênica , Antígeno Ki-67/metabolismo , Masculino , Camundongos Knockout , Complexos Multienzimáticos/deficiência , Neovascularização Fisiológica , Osteogênese/genética , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo
6.
J Pathol ; 235(5): 672-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25385233

RESUMO

G protein-regulated cell function is crucial for cardiomyocytes, and any deregulation of its gene expression or protein modification can lead to pathological cardiac hypertrophy. Herein, we report that protein prenylation, a lipidic modification of G proteins that facilitates their association with the cell membrane, might control the process of cardiomyocyte hypertrophy. We found that geranylgeranyl diphosphate synthase (GGPPS), a key enzyme involved in protein prenylation, played a critical role in postnatal heart growth by regulating cardiomyocyte size. Cardiac-specific knockout of GGPPS in mice led to spontaneous cardiac hypertrophy, beginning from week 4, accompanied by the persistent enlargement of cardiomyocytes. This hypertrophic effect occurred by altered prenylation of G proteins. Evaluation of the prenylation, membrane association and hydrophobicity showed that Rheb was hyperactivated and increased mTORC1 signalling pathway after GGPPS deletion. Protein farnesylation or mTORC1 inhibition blocked GGPPS knockdown-induced mTORC1 activation and suppressed the larger neonatal rat ventricle myocyte size and cardiomyocyte hypertrophy in vivo, demonstrating a central role of the FPP-Rheb-mTORC1 axis for GGPPS deficiency-induced cardiomyocyte hypertrophy. The sustained cardiomyocyte hypertrophy progressively provoked cardiac decompensation and dysfunction, ultimately causing heart failure and adult death. Importantly, GGPPS was down-regulated in the hypertrophic hearts of mice subjected to transverse aortic constriction (TAC) and in failing human hearts. Moreover, HPLC-MS/MS detection revealed that the myocardial farnesyl diphosphate (FPP):geranylgeranyl diphosphate (GGPP) ratio was enhanced after pressure overload. Our observations conclude that the alteration of protein prenylation promotes cardiomyocyte hypertrophic growth, which acts as a potential cause for pathogenesis of heart failure and may provide a new molecular target for hypertrophic heart disease clinical therapy.


Assuntos
Cardiomegalia/enzimologia , Farnesiltranstransferase/deficiência , Insuficiência Cardíaca/enzimologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Miócitos Cardíacos/enzimologia , Neuropeptídeos/metabolismo , Prenilação de Proteína , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Cardiomegalia/tratamento farmacológico , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Linhagem Celular , Modelos Animais de Doenças , Progressão da Doença , Farnesiltranstransferase/genética , Feminino , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Complexos Multiproteicos/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Neuropeptídeos/genética , Inibidores de Proteínas Quinases/farmacologia , Prenilação de Proteína/efeitos dos fármacos , Interferência de RNA , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Fatores de Tempo , Transfecção , Função Ventricular Esquerda
7.
J Biol Chem ; 288(50): 35952-60, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24136196

RESUMO

Isoprenoids and prenylated proteins have been implicated in the pathophysiology of Alzheimer disease (AD), including amyloid-ß precursor protein metabolism, Tau phosphorylation, synaptic plasticity, and neuroinflammation. However, little is known about the relative importance of the two protein prenyltransferases, farnesyltransferase (FT) and geranylgeranyltransferase-1 (GGT), in the pathogenesis of AD. In this study, we defined the impact of deleting one copy of FT or GGT on the development of amyloid-ß (Aß)-associated neuropathology and learning/memory impairments in APPPS1 double transgenic mice, a well established model of AD. Heterozygous deletion of FT reduced Aß deposition and neuroinflammation and rescued spatial learning and memory function in APPPS1 mice. Heterozygous deletion of GGT reduced the levels of Aß and neuroinflammation but had no impact on learning and memory. These results document that farnesylation and geranylgeranylation play differential roles in AD pathogenesis and suggest that specific inhibition of protein farnesylation could be a potential strategy for effectively treating AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Cognição , Farnesiltranstransferase/deficiência , Farnesiltranstransferase/genética , Deleção de Genes , Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Memória , Camundongos , Camundongos Transgênicos , Proteólise
8.
J Lipid Res ; 53(1): 77-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22039581

RESUMO

Protein farnesyltransferase (FTase) and protein geranylgeranyltransferase-I (GGTase-I) add 15- or 20-carbon lipids, respectively, to proteins that terminate with a CaaX motif. These posttranslational modifications of proteins with lipids promote protein interactions with membrane surfaces in cells, but the in vivo importance of the CaaX prenyltransferases and the protein lipidation reactions they catalyze remain incompletely defined. One study concluded that a deficiency of FTase was inconsequential in adult mice and led to little or no tissue pathology. To assess the physiologic importance of the CaaX prenyltransferases, we used conditional knockout alleles and an albumin-Cre transgene to produce mice lacking FTase, GGTase-I, or both enzymes in hepatocytes. The hepatocyte-specific FTase knockout mice survived but exhibited hepatocellular disease and elevated transaminases. Mice lacking GGTase-I not only had elevated transaminases but also had dilated bile cannaliculi, hyperbilirubinemia, hepatosplenomegaly, and reduced survival. Of note, GGTase-I-deficient hepatocytes had a rounded shape and markedly reduced numbers of actin stress fibers. Hepatocyte-specific FTase/GGTase-I double-knockout mice closely resembled mice lacking GGTase-I alone, but the disease was slightly more severe. Our studies refute the notion that FTase is dispensable and demonstrate that GGTase-I is crucial for the vitality of hepatocytes.


Assuntos
Alquil e Aril Transferases/deficiência , Dimetilaliltranstransferase/deficiência , Farnesiltranstransferase/deficiência , Hepatócitos/enzimologia , Hepatopatias/fisiopatologia , Prenilação de Proteína/efeitos dos fármacos , Animais , Fígado/patologia , Fígado/fisiopatologia , Hepatopatias/patologia , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...