Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2314201121, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38635631

RESUMO

To effectively protect the host from viral infection while avoiding excessive immunopathology, the innate immune response must be tightly controlled. However, the precise regulation of antiviral innate immunity and the underlying mechanisms remain unclear. Here, we find that sirtuin3 (SIRT3) interacts with mitochondrial antiviral signaling protein (MAVS) to catalyze MAVS deacetylation at lysine residue 7 (K7), which promotes MAVS aggregation, as well as TANK-binding kinase I and IRF3 phosphorylation, resulting in increased MAVS activation and enhanced type I interferon signaling. Consistent with these findings, loss of Sirt3 in mice and zebrafish renders them more susceptible to viral infection compared to their wild-type (WT) siblings. However, Sirt3 and Sirt5 double-deficient mice exhibit the same viral susceptibility as their WT littermates, suggesting that loss of Sirt5 in Sirt3-deficient mice may counteract the increased viral susceptibility displayed in Sirt3-deficient mice. Thus, we not only demonstrate that SIRT3 positively regulates antiviral immunity in vitro and in vivo, likely via MAVS, but also uncover a previously unrecognized mechanism by which SIRT3 acts as an accelerator and SIRT5 as a brake to orchestrate antiviral innate immunity.


Assuntos
Sirtuína 3 , Sirtuínas , Viroses , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Imunidade Inata , Lisina , Sirtuína 3/genética , Sirtuínas/genética , Peixe-Zebra , Proteínas de Peixe-Zebra
2.
EMBO J ; 39(11): e103285, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32301534

RESUMO

RLR-mediated type I IFN production plays a pivotal role in innate antiviral immune responses, where the signaling adaptor MAVS is a critical determinant. Here, we show that MAVS is a physiological substrate of SIRT5. Moreover, MAVS is succinylated upon viral challenge, and SIRT5 catalyzes desuccinylation of MAVS. Mass spectrometric analysis indicated that Lysine 7 of MAVS is succinylated. SIRT5-catalyzed desuccinylation of MAVS at Lysine 7 diminishes the formation of MAVS aggregation after viral infection, resulting in the inhibition of MAVS activation and leading to the impairment of type I IFN production and antiviral gene expression. However, the enzyme-deficient mutant of SIRT5 (SIRT5-H158Y) loses its suppressive role on MAVS activation. Furthermore, we show that Sirt5-deficient mice are resistant to viral infection. Our study reveals the critical role of SIRT5 in limiting RLR signaling through desuccinylating MAVS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Agregados Proteicos , Sirtuínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Substituição de Aminoácidos , Animais , Regulação da Expressão Gênica , Células HCT116 , Células HEK293 , Humanos , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Sirtuínas/genética
3.
New Phytol ; 242(3): 1257-1274, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38481385

RESUMO

Plant pathogenic fungi elaborate numerous detoxification strategies to suppress host reactive oxygen species (ROS), but their coordination is not well-understood. Here, we show that Sirt5-mediated protein desuccinylation in Magnaporthe oryzae is central to host ROS detoxification. SIRT5 encodes a desuccinylase important for virulence via adaptation to host oxidative stress. Quantitative proteomics analysis identified a large number of succinylated proteins targeted by Sirt5, most of which were mitochondrial proteins involved in oxidative phosphorylation, TCA cycle, and fatty acid oxidation. Deletion of SIRT5 resulted in hypersuccinylation of detoxification-related enzymes, and significant reduction in NADPH : NADP+ and GSH : GSSG ratios, disrupting redox balance and impeding invasive growth. Sirt5 desuccinylated thioredoxin Trx2 and glutathione peroxidase Hyr1 to activate their enzyme activity, likely by affecting proper folding. Altogether, this work demonstrates the importance of Sirt5-mediated desuccinylation in controlling fungal process required for detoxifying host ROS during M. oryzae infection.


Assuntos
Ascomicetos , Magnaporthe , Oryza , Espécies Reativas de Oxigênio/metabolismo , Lisina/metabolismo , Estresse Oxidativo , Ascomicetos/metabolismo , Proteínas Fúngicas/metabolismo , Oryza/metabolismo , Doenças das Plantas/microbiologia
4.
BMC Cancer ; 24(1): 386, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532359

RESUMO

Chordoma is a relatively rare and locally aggressive malignant tumor. Sirtuin (SIRT)5 plays pivotal roles in various tumors, but the role of SIRT5 in chordoma has not been found. This study was performed to investigate the regulatory effects of SIRT5 on cell proliferation, migration, and invasion and the underlying mechanism in chordoma. A xenograft tumor mouse model was established to assess tumor growth. Reverse transcription-quantitative polymerase chain reaction was used to analyze the mRNA levels of SIRT5 and c-myc. The effects of SIRT5 and c-myc on cell proliferation, migration, and invasion of chordoma cells were detected by cell counting kit-8, colony formation, and Transwell assays. The interaction between SIRT5 and c-myc was evaluated by co-immunoprecipitation (IP) assay. The succinylation of c-myc was analyzed by IP and Western blot. The results showed that SIRT5 expression was upregulated in chordoma tissues and cells. SIRT5 interacted with c-myc to inhibit the succinylation of c-myc at K369 site in human embryonic kidney (HEK)-293T cells. Silencing of SIRT5 suppressed the cell proliferation, migration, and invasion of chordoma cells, while the results were reversed after c-myc overexpression. Moreover, silencing SIRT5 suppressed tumor growth in mice. These findings suggested that SIRT5 promoted the malignant advancement of chordoma by regulating the desuccinylation of c-myc.


Assuntos
Cordoma , Sirtuínas , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células , Sirtuínas/genética , Sirtuínas/metabolismo
5.
BMC Cardiovasc Disord ; 24(1): 464, 2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39210272

RESUMO

Myocardial infarction (MI) is a prevalent form of ischemic heart disease, significantly contributing to heart disease-related deaths worldwide. This condition is primarily caused by myocardial ischemic-reperfusion injury (MIRI). Sirtuin 5 (SIRT5) is a desuccinylase known for its ability to reduce protein succinylation. Recent studies have highlighted the potential role of SIRT5 in various human diseases, including MIRI. This study aims to investigate the specific role of SIRT5 in modulating autophagy and cardiomyocyte death in a MIRI model, as well as to identify the downstream protein targets of SIRT5. Initially, we established a hypoxia/reoxygenation (H/R)-induced MIRI cell model to measure SIRT5 expression and assess its functions. Our results indicated that H/R induction led to a downregulation of SIRT5 expression, decreased autophagy, and increased cell death. Notably, overexpression of SIRT5 effectively promoted autophagy and inhibited cell death in the MIRI cell model. Mechanistically, SIRT5 was found to directly interact with the target of myb1 membrane trafficking protein (TOM1) at the K48 site, inducing its desuccinylation and stabilization. Further rescue assays revealed that TOM1 knockdown reversed the changes in autophagy and apoptosis caused by SIRT5 overexpression in the MIRI cell model. In vivo experiments demonstrated that SIRT5 alleviated myocardial injury in MI models. In conclusion, this study uncovers the role of SIRT5-mediated desuccinylation of TOM1 in regulating autophagy-related cell death in MIRI, providing new insights into potential therapeutic strategies for MI.


Assuntos
Autofagia , Modelos Animais de Doenças , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Transdução de Sinais , Sirtuínas , Sirtuínas/metabolismo , Sirtuínas/genética , Miócitos Cardíacos/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/enzimologia , Animais , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/genética , Masculino , Camundongos Endogâmicos C57BL , Apoptose , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/genética , Linhagem Celular , Humanos
6.
Clin Exp Pharmacol Physiol ; 51(9): e13909, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39038854

RESUMO

Diabetic kidney disease (DKD) is a complication of diabetic mellitus. New treatments need to be developed. This study aimed to investigate the effects of quercetin-4'-O-ß-D-glucopyranoside (QODG) on podocyte injury. Podocytes were cultured in high glucose (HG) medium, treated with QODG, and overexpressing or knocking down SIRT5. Oxidative stress indicators were assessed using corresponding kits. Pyroptosis was detected by flow cytometry and western blot analysis. Succinylation modification was detected using immunoprecipitation (IP) and western blot analysis. The interaction between NEK7 and NLRP3 was determined by co-IP. The results indicated that QODG inhibited oxidative stress and pyroptosis of podocytes induced by HG. Besides, QODG suppressed succinylation levels in HG-induced podocytes, with the upregulation of SIRT5. Knockdown of SIRT5 reversed the effects of QODG on oxidative stress and pyroptosis. Moreover, SIRT5 inhibited the succinylation of NEK7 and the interaction between NLRP3 and NEK7. In conclusion, QODG upregulates SIRT5 to inhibit the succinylation modification of NEK7, impedes the interaction between NEK7 and NLRP3, and then inhibits the pyroptosis and oxidative stress injury of podocytes under HG conditions. The findings suggested that QODG has the potential to treat DKD and explore a novel underlying mechanism of QODG function.


Assuntos
Quinases Relacionadas a NIMA , Podócitos , Sirtuínas , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Podócitos/patologia , Quinases Relacionadas a NIMA/metabolismo , Sirtuínas/metabolismo , Sirtuínas/genética , Animais , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Glucosídeos/farmacologia , Linhagem Celular
7.
Artigo em Inglês | MEDLINE | ID: mdl-39145876

RESUMO

PURPOSE: Preeclampsia (PE) is a pregnancy-specific syndrome with increasing maternal and perinatal morbidity and mortality. Succinylation, a post-translational modification event, has been found in various diseases. However, the role of succinylation in PE has not been explored. This study aimed to investigate the effect of succinylation on PE and the underlying mechanisms. METHODS: Thirty-two PE patients and 32 normal pregnancy volunteers were recruited. Human extravasated trophoblast cells (HTR-8/SVneo) were used in in vitro study. RT-qPCR was performed to detect the expression of succinylation-related mRNAs. The cell proliferation, invasion, and migration were assessed using cell counting kit-8, ethynyldeoxyuridine, transwell, and wound healing assays. Co-immunoprecipitation and dual-luciferase reporter assays were performed to analyze the interaction between sirtuin (SIRT)5 and homeobox box 3 (HOXB3). RESULTS: SIRT5 was increased in the placental tissues of PE patients. SIRT5 inhibition increased cell proliferation, invasion, and migration in HTR-8/SVneo cells. Mechanistic investigations indicated that HOXB3 was a downstream regulatory target of SIRT5-mediated desuccinylation. Rescue experiments further verified that silencing of HOXB3 inhibited cell proliferation, invasion, and migration. Additionally, HOXB3 deficiency reversed the activation of the Notch and ß-catenin signaling pathway induced by SIRT5 inhibition. CONCLUSION: SIRT5 inhibited the trophoblast cell proliferation, invasion, and migration to promote PE through suppressing Notch and ß-catenin signaling pathway activation via desuccinylating HOXB3.

8.
Acta Pharmacol Sin ; 43(9): 2373-2385, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35046516

RESUMO

The protein arginine methyltransferase 5 (PRMT5), which is highly expressed in tumour tissues, plays a crucial role in cancer development. However, the mechanism by which PRMT5 promotes cancer growth is poorly understood. Here, we report that PRMT5 contributes to lipid metabolism reprogramming, tumour growth and metastasis depending on the SIRT7-mediated desuccinylation of PRMT5 K387 in tumours. Mass spectrometric analysis identified PRMT5 lysine 387 as its succinylation site. Moreover, the desuccinylation of PRMT5 K387 enhances the methyltransferase activity of PRMT5. SIRT7 catalyses the desuccinylation of PRMT5 in cells. The SIRT7-mediated dessuccinylation of PRMT5 lysine 387 fails to bind to STUB1, decreasing PRMT5 ubiquitination and increasing the interaction between PRMT5 and Mep50, which promotes the formation of the PRMT5-Mep50 octamer. The PRMT5-Mep50 octamer increases PRMT5 methyltransferase activity, leading to arginine methylation of SREBP1a. The symmetric dimethylation of SREBP1a increases the levels of cholesterol, fatty acid, and triglyceride biogenesis in the cells, escaping degradation through the ubiquitin-proteasome pathway. Functionally, the desuccinylation of PRMT5 K387 promotes lipid metabolism reprogramming, tumour growth and metastasis in vitro and in vivo in tumours.


Assuntos
Neoplasias , Sirtuínas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Humanos , Metabolismo dos Lipídeos , Lisina , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Sirtuínas/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
9.
Molecules ; 27(21)2022 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-36363992

RESUMO

FucoPol is an acylated polysaccharide with demonstrated valuable functional properties that include a shear thinning fluid behaviour, a film-forming capacity, and an emulsion forming and stabilizing capacity. In this study, the different conditions (concentration, temperature, and time) for alkaline treatment were investigated to deacylate FucoPol. Complete deacetylation and desuccinylation was achieved with 0.02 M NaOH, at 60 °C for 15 min, with no significant impact on the biopolymer's sugar composition, pyruvate content, and molecular mass distribution. FucoPol depyruvylation by acid hydrolysis was attempted, but it resulted in a very low polymer recovery. The effect of the ionic strength, pH, and temperature on the deacetylated/desuccinylated polysaccharide, d-FucoPol, was evaluated, as well as its emulsion and film-forming capacity. d-FucoPol aqueous solutions maintained the shear thinning behaviour characteristic of FucoPol, but the apparent viscosity decreased significantly. Moreover, contrary to FucoPol, whose solutions were not affected by the media's ionic strength, the d-FucoPol solutions had a significantly higher apparent viscosity for a higher ionic strength. On the other hand, the d-FucoPol solutions were not affected by the pH in the range of 3.6-11.5, while FucoPol had a decreased viscosity for acidic pH values and for a pH above 10.5. Although d-FucoPol displayed an emulsification activity for olive oil similar to that of FucoPol (98 ± 0%) for an oil-to-water ratio of 2:3, the emulsions were less viscous. The d-FucoPol films were flexible, with a higher Young's modulus (798 ± 152 MPa), a stress at the break (22.5 ± 2.5 MPa), and an elongation at the break (9.3 ± 0.7%) than FucoPol (458 ± 32 MPa, 15.5 ± 0.3 MPa and 8.1 ± 1.0%, respectively). Given these findings, d-FucoPol arises as a promising novel biopolymer, with distinctive properties that may render it useful for utilization as a suspending or emulsifier agent, and as a barrier in coatings and packaging films.


Assuntos
Fucose , Polissacarídeos , Emulsões , Polissacarídeos/química , Viscosidade , Biopolímeros , Reologia
10.
J Cell Mol Med ; 25(15): 7101-7109, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34160884

RESUMO

Protein post-translational modifications (PTMs) of histones are ubiquitous regulatory mechanisms involved in many biological processes, including replication, transcription, DNA damage repair and ontogenesis. Recently, many short-chain acylation histone modifications have been identified by mass spectrometry (MS). Lysine succinylation (Ksuc or Ksucc) is a newly identified histone PTM that changes the chemical environment of histones and is similar to other acylation modifications; lysine succinylation appears to accumulate at transcriptional start sites and to correlate with gene expression. Although numerous studies are ongoing, there is a lack of reviews on the Ksuc of histones. Here, we review lysine succinylation sites on histones, including the chemical characteristics and the mechanism by which lysine succinylation influences nucleosomal structure, chromatin dynamics and several diseases and then discuss lysine succinylation regulation to identify theoretical and experimental proof of Ksuc on histones and in diseases to inspire further research into histone lysine succinylation as a target of disease treatment in the future.


Assuntos
Código das Histonas , Nucleossomos/metabolismo , Succinatos/metabolismo , Animais , Humanos
11.
J Transl Med ; 19(1): 521, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930316

RESUMO

BACKGROUND: The aim of this study was to investigate the biological functions and underlying mechanisms of SIRT5 in clear cell renal cell carcinoma (ccRCC). METHODS: SIRT5 expression data in The Cancer Genome Atlas Kidney Clear Cell Carcinoma (TCGA-KIRC) were selected, and the correlations between SIRT5 expression and various clinicopathological parameters were analysed. SIRT5 expression in ccRCC tissues was examined using immunohistochemistry. Stable cell lines with SIRT5 knockdown were established. In vitro and in vivo experiments were conducted to investigate the functional roles of SIRT5 in the cellular biology of ccRCC, including cell viability assays, wound healing assays, soft agar colony formation assays, Transwell invasion assays, qRT-PCR, and Western blotting. In addition, microarrays, rescue experiments and Western blotting were used to investigate the molecular mechanisms underlying SIRT5 functions. RESULTS: SIRT5 expression was downregulated in ccRCC compared with normal tissues, which correlated with a poor prognosis of ccRCC. SIRT5 knockdown significantly increased cell proliferation, migration and invasion in vitro. In vivo experiments revealed that SIRT5 knockdown promoted ccRCC tumorigenesis and metastasis. Mechanistically, SIRT5 deglycosylated PDHA1 at K351 and increased PDC activity, thereby altering the metabolic crosstalk with the TCA cycle and inhibiting the Warburg effect. SIRT5 overexpression was related to low succinylation of PDHA1. CONCLUSIONS: Downregulated SIRT5 expression in ccRCC accelerated the Warburg effect through PDHA1 hypersuccinylation and induced tumorigenesis and progression, indicating that SIRT5 may become a potential target for ccRCC therapy.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Sirtuínas , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Neoplasias Renais/patologia , Sirtuínas/genética , Sirtuínas/metabolismo
12.
Exp Cell Res ; 382(2): 111483, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31247190

RESUMO

SIRT5 has a wide range of functions in different cellular processes such as glycolysis, TCA cycle and antioxidant defense, which mediates lysine desuccinylation, deglutarylation and demalonylation. Recent evidences have implicated that SIRT5 is a potential suppressor of gastric cancer (GC). However, the underlying mechanism of SIRT5 in gastric cancer is still unclear. Here, we show that SIRT5 expression is significantly decreased in human GC tissues. Functional analysis demonstrates that SIRT5 inhibits cell growth in vitro and in vivo, arrests the cell cycle in G1/S transition, and suppresses migration and invasion of GC cells via regulating epithelial-to-mesenchymal transition. Mechanistically, we demonstrate that there is the direct interaction between SIRT5 and 2-oxoglutarate dehydrogenase (OGDH), and desuccinylation of OGDH by SIRT5 inhibits the activity of OGDH complex. Further studies of the relationship between SIRT5 and OGDH show OGDH inhibition by succinyl phosphonate (SP) or siRNA suppresses the increase in cell growth and migration induced by SIRT5 deletion. Moreover, SIRT5 decreases mitochondrial membrane potential (ΔΨm), ATP products and increases the ROS levels and NADP/NADPH ratio in GC cells through the inhibition of OGDH complex activity. Therefore, SIRT5 suppresses GC cell growth and migration through desuccinylating OGDH and inhibiting OGDH complex activity to disturb mitochondrial functions and redox status.


Assuntos
Movimento Celular , Complexo Cetoglutarato Desidrogenase/metabolismo , Sirtuínas/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Nus , Mitocôndrias/metabolismo , Invasividade Neoplásica , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Ácido Succínico/metabolismo
13.
Biochem J ; 476(2): 211-223, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30523058

RESUMO

Histone modification is a ubiquitous regulatory mechanism involved in a variety of biological processes, including gene expression, DNA damage repair, cell differentiation, and ontogenesis. Succinylation sites on histones have been identified and may have functional consequences. Here, we demonstrate that human sirtuin 5 (Sirt5) catalyzes the sequence-selective desuccinylation of numerous histone succinyl sites. Structural studies of Sirt5 in complex with four succinyl peptides indicate an essential role for the conserved main chain hydrogen bonds formed by the succinyl lysine (0), +1, and +3 sites for substrate-enzyme recognition. Furthermore, biochemical assays reveal that the proline residue at the +1 site of the histone succinylation substrate is unfavorable for Sirt5 interaction. Our findings illustrate the molecular mechanism underlying the sequence-selective desuccinylase activity of Sirt5 and provide insights for further studies of the biological functions associated with histone succinylation and Sirt5.


Assuntos
Histonas/química , Peptídeos/química , Processamento de Proteína Pós-Traducional , Sirtuínas/química , Ácido Succínico/química , Histonas/genética , Histonas/metabolismo , Humanos , Peptídeos/genética , Peptídeos/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo , Relação Estrutura-Atividade , Ácido Succínico/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(16): 4320-5, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27051063

RESUMO

Cellular metabolites, such as acyl-CoA, can modify proteins, leading to protein posttranslational modifications (PTMs). One such PTM is lysine succinylation, which is regulated by sirtuin 5 (SIRT5). Although numerous proteins are modified by lysine succinylation, the physiological significance of lysine succinylation and SIRT5 remains elusive. Here, by profiling acyl-CoA molecules in various mouse tissues, we have discovered that different tissues have different acyl-CoA profiles and that succinyl-CoA is the most abundant acyl-CoA molecule in the heart. This interesting observation has prompted us to examine protein lysine succinylation in different mouse tissues in the presence and absence of SIRT5. Protein lysine succinylation predominantly accumulates in the heart whenSirt5is deleted. Using proteomic studies, we have identified many cardiac proteins regulated by SIRT5. Our data suggest that ECHA, a protein involved in fatty acid oxidation, is a major enzyme that is regulated by SIRT5 and affects heart function.Sirt5knockout (KO) mice have lower ECHA activity, increased long-chain acyl-CoAs, and decreased ATP in the heart under fasting conditions.Sirt5KO mice develop hypertrophic cardiomyopathy, as evident from the increased heart weight relative to body weight, as well as reduced shortening and ejection fractions. These findings establish that regulating heart metabolism and function is a major physiological function of lysine succinylation and SIRT5.


Assuntos
Acil Coenzima A/metabolismo , Cardiomegalia/metabolismo , Ácidos Graxos/metabolismo , Miocárdio/metabolismo , Processamento de Proteína Pós-Traducional , Sirtuínas/metabolismo , Acil Coenzima A/genética , Acilação , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Ácidos Graxos/genética , Metabolômica/métodos , Camundongos , Camundongos Knockout , Miocárdio/patologia , Oxirredução , Proteômica/métodos , Sirtuínas/genética
15.
Bioorg Med Chem Lett ; 25(8): 1671-1674, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25818461

RESUMO

A fluorogenic assay for SIRT5 has been developed to screen their small molecule modulators based on the recent discovery that SIRT5 is a demalonylase and desuccinylase. However, this assay uses a fluorogenic peptide containing 7-amino-4-methylcoumarin (AMC), which becomes the cause of false positive hits from the screening. To overcome this, we have developed an alternative method called a FRET-based assay, which will be reliable and useful for screening SIRT5 modulators in a high-throughput format since no AMC group present in this assay.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Sirtuínas/metabolismo , Sequência de Aminoácidos , Cumarínicos/química , Ensaios Enzimáticos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Cinética , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Sirtuínas/antagonistas & inibidores , Especificidade por Substrato , p-Dimetilaminoazobenzeno/análogos & derivados , p-Dimetilaminoazobenzeno/química
16.
Immun Inflamm Dis ; 12(8): e1341, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39092715

RESUMO

BACKGROUND: Sirtuin 7 (SIRT7) is pivotal in diverse diseases progression. Importantly, SIRT7 is associated with melanin production. However, whether SIRT7 regulates vitiligo is unclear. Therefore, we aimed to investigate the effects of SIRT7 on pigmentation and the modification of glucose 6-phosphate dehydrogenase (G6PD). METHODS: After knockdown SIRT7 and G6PD, pigmentation of melanocytes was evaluated using commercial kits, immunofluorescence, and Western blot analysis. The succinylation of G6PD mediated by SIRT7 was analyzed using co-immunoprecipitation, immunofluorescence, Western blot analysis, and cycloheximide-chase experiment. RESULTS: We found that SIRT7 was highly expressed in vitiligo skin lesions. Knockdown of SIRT7 increased tyrosinase activity, melanin content, and the levels of α-melanocyte-stimulating hormone, MITF, TYR, TRP1, and TRP2. Additionally, SIRT7 directly interacted with G6PD. Silenced SIRT7 promoted the succinylation of G6PD and enhanced its protein stability. G6PD knockdown reversed the effect of reduced SIRT7 expression on melanin production. CONCLSUION: Silencing of SIRT7 promotes pigmentation of melanocytes by succinylating G6PD, suggesting that SIRT7-mediated G6PD desuccinylation may promote vitiligo progression.


Assuntos
Progressão da Doença , Glucosefosfato Desidrogenase , Melaninas , Melanócitos , Sirtuínas , Vitiligo , Vitiligo/metabolismo , Vitiligo/patologia , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Sirtuínas/metabolismo , Sirtuínas/genética , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Melaninas/metabolismo , Melaninas/biossíntese
17.
FEBS Lett ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-39031546

RESUMO

SIRT5, one of the mammalian sirtuins, specifically recognizes succinyl-lysine residues on proteins and catalyzes the desuccinylation reaction. In this study, we characterized SIRT5 mutants with hydrophobic amino acid substitutions at Q140 and N141, in addition to the catalytic residue H158, known as an active site residue, by the Michaelis-Menten analysis and X-ray crystallography. Kinetic analysis showed that the catalytic efficiency (kcat/Km) of the Q140L and N141V mutants decreased to 0.02 times and 0.0038 times that of the wild-type SIRT5, respectively, with the activity of the N141V mutant becoming comparable to that of the H158M mutant. Our findings indicate that N141 contributes significantly to the desuccinylation reaction.

18.
J Neuroimmune Pharmacol ; 19(1): 17, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38717643

RESUMO

In our previous study, we concluded that sirtuin 5 (SIRT5) was highly expressed in microglia following ischaemic stroke, which induced excessive neuroinflammation and neuronal injury. Therefore, SIRT5-targeting interventions should reduce neuroinflammation and protect against ischaemic brain injury. Here, we showed that treatment with a specific SIRT5 inhibitor, MC3482, alleviated microglia-induced neuroinflammation and improved long-term neurological function in a mouse model of stroke. The mice were administrated with either vehicle or 2 mg/kg MC3482 daily for 7 days via lateral ventricular injection following the onset of middle cerebral artery occlusion. The outcome was assessed by a panel of tests, including a neurological outcome score, declarative memory, sensorimotor tests, anxiety-like behavior and a series of inflammatory factors. We observed a significant reduction of infarct size and inflammatory factors, and the improvement of long-term neurological function in the early stages during ischaemic stroke when the mice were treated with MC3482. Mechanistically, the administration of MC3482 suppressed the desuccinylation of annexin-A1, thereby promoting its membrane recruitment and extracellular secretion, which in turn alleviated neuroinflammation during ischaemic stroke. Based on our findings, MC3482 offers promise as an anti-ischaemic stroke treatment that targets directly the disease's underlying factors.


Assuntos
Anexina A1 , AVC Isquêmico , Microglia , Doenças Neuroinflamatórias , Sirtuínas , Animais , Masculino , Camundongos , Anexina A1/efeitos dos fármacos , Anexina A1/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Sirtuínas/antagonistas & inibidores , Sirtuínas/metabolismo , Regulação para Cima/efeitos dos fármacos
19.
Methods Mol Biol ; 2589: 303-316, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36255633

RESUMO

The class III histone deacetylase (HDACs) also known as sirtuins (SIRTs 1-7) are ubiquitously expressed, but SIRT7 mainly resides as nucleolar protein. In this chapter a couple of methods are described that are used to detect modulation of SIRT7 in response to DNA damage. SIRT7 is localized in the nucleoli and binds to the chromatin after DNA damage. Therefore, a protocol was optimized by our lab for chromatin fractionation. By this method, the movement of SIRT7 can be detected from the soluble part (cytosol+nucleoplasm) to the solid part (chromatin) of the cell. Change of SIRT7 expression levels, in different cells or after different treatment, can be detected by isolating whole-cell lysate followed by Western blotting. For analyzing binding of SIRT7 to other substrates, we have also optimized manual immunoprecipitation assays by using 1% NP40 buffer. This protocol is very helpful to pull down SIRT7 and associated proteins by using a single buffer. SIRT7 is a deacetylase, and its deacetylation activity can be checked both inside the cell by in vivo deacetylation assay and outside the cell by in vitro deacetylation assays. Recently it was also discovered that SIRT7 has desuccinylase activity which can be detected by histone desuccinylation assay. This chapter provides the methodology of SIRT7 detection in the whole cell lysate, binding of SIRT7 to the chromatin and other proteins for performing deacetylation and desuccinylation activity.


Assuntos
Histonas , Sirtuínas , Histonas/metabolismo , Cromatina , Dano ao DNA , Sirtuínas/metabolismo , Histona Desacetilases/metabolismo
20.
Cells ; 12(6)2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36980194

RESUMO

Sirtuin 5 (SIRT5) is a predominantly mitochondrial enzyme catalyzing the removal of glutaryl, succinyl, malonyl, and acetyl groups from lysine residues through a NAD+-dependent deacylase mechanism. SIRT5 is an important regulator of cellular homeostasis and modulates the activity of proteins involved in different metabolic pathways such as glycolysis, tricarboxylic acid (TCA) cycle, fatty acid oxidation, electron transport chain, generation of ketone bodies, nitrogenous waste management, and reactive oxygen species (ROS) detoxification. SIRT5 controls a wide range of aspects of myocardial energy metabolism and plays critical roles in heart physiology and stress responses. Moreover, SIRT5 has a protective function in the context of neurodegenerative diseases, while it acts as a context-dependent tumor promoter or suppressor. In addition, current research has demonstrated that SIRT5 is implicated in the SARS-CoV-2 infection, although opposing conclusions have been drawn in different studies. Here, we review the current knowledge on SIRT5 molecular actions under both healthy and diseased settings, as well as its functional effects on metabolic targets. Finally, we revise the potential of SIRT5 as a therapeutic target and provide an overview of the currently reported SIRT5 modulators, which include both activators and inhibitors.


Assuntos
COVID-19 , Neoplasias , Sirtuínas , Humanos , COVID-19/metabolismo , Redes e Vias Metabólicas , Neoplasias/metabolismo , SARS-CoV-2/metabolismo , Sirtuínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA