Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 470
Filtrer
1.
FASEB J ; 38(13): e23756, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38949649

RÉSUMÉ

Asthma is a chronic pulmonary disease with the worldwide prevalence. The structural alterations of airway walls, termed as "airway remodeling", are documented as the core contributor to the airway dysfunction during chronic asthma. Forkhead box transcription factor FOXK2 is a critical regulator of glycolysis, a metabolic reprogramming pathway linked to pulmonary fibrosis. However, the role of FOXK2 in asthma waits further explored. In this study, the chronic asthmatic mice were induced via ovalbumin (OVA) sensitization and repetitive OVA challenge. FOXK2 was upregulated in the lungs of OVA mice and downregulated after adenovirus-mediated FOXK2 silencing. The lung inflammation, peribronchial collagen deposition, and glycolysis in OVA mice were obviously attenuated after FOXK2 knockdown. Besides, the expressions of FOXK2 and SIRT2 in human bronchial epithelial cells (BEAS-2B) were increasingly upregulated upon TGF-ß1 stimulation and downregulated after FOXK2 knockdown. Moreover, the functional loss of FOXK2 remarkably suppressed TGF-ß1-induced epithelial-mesenchymal transition (EMT) and glycolysis in BEAS-2B cells, as manifested by the altered expressions of EMT markers and glycolysis enzymes. The glycolysis inhibitor 2-deoxy-d-glucose (2-DG) inhibited the EMT in TGF-ß1-induced cells, making glycolysis a driver of EMT. The binding of FOXK2 to SIRT2 was validated, and SIRT2 overexpression blocked the FOXK2 knockdown-mediated inhibition of EMT and glycolysis in TGF-ß1-treated cells, which suggests that FOXK2 regulates EMT and glycolysis in TGF-ß1-treated cells in a SIRT2-dependnet manner. Collectively, this study highlights the protective effect of FOXK2 knockdown on airway remodeling during chronic asthma.


Sujet(s)
Remodelage des voies aériennes , Asthme , Facteurs de transcription Forkhead , Glycolyse , Sirtuine-2 , Asthme/métabolisme , Asthme/anatomopathologie , Animaux , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Souris , Remodelage des voies aériennes/physiologie , Humains , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Transition épithélio-mésenchymateuse , Souris de lignée BALB C , Femelle , Facteur de croissance transformant bêta-1/métabolisme , Poumon/métabolisme , Poumon/anatomopathologie , Lignée cellulaire
2.
Cell Mol Immunol ; 21(7): 674-688, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38740925

RÉSUMÉ

The NLRP3 inflammasome functions as an inflammatory driver, but its relationship with lipid metabolic changes in early sepsis remains unclear. Here, we found that GITR expression in monocytes/macrophages was induced by lysophosphatidylcholine (LPC) and was positively correlated with the severity of sepsis. GITR is a costimulatory molecule that is mainly expressed on T cells, but its function in macrophages is largely unknown. Our in vitro data showed that GITR enhanced LPC uptake by macrophages and specifically enhanced NLRP3 inflammasome-mediated macrophage pyroptosis. Furthermore, in vivo studies using either cecal ligation and puncture (CLP) or LPS-induced sepsis models demonstrated that LPC exacerbated sepsis severity/lethality, while conditional knockout of GITR in myeloid cells or NLRP3/caspase-1/IL-1ß deficiency attenuated sepsis severity/lethality. Mechanistically, GITR specifically enhanced inflammasome activation by regulating the posttranslational modification (PTM) of NLRP3. GITR competes with NLRP3 for binding to the E3 ligase MARCH7 and recruits MARCH7 to induce deacetylase SIRT2 degradation, leading to decreasing ubiquitination but increasing acetylation of NLRP3. Overall, these findings revealed a novel role of macrophage-derived GITR in regulating the PTM of NLRP3 and systemic inflammatory injury, suggesting that GITR may be a potential therapeutic target for sepsis and other inflammatory diseases. GITR exacerbates LPC-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3. According to the model, LPC levels increase during the early stage of sepsis, inducing GITR expression on macrophages. GITR not only competes with NLRP3 for binding to the E3 ligase MARCH7 but also recruits MARCH7 to induce the degradation of the deacetylase SIRT2, leading to decreasing ubiquitination but increasing acetylation of NLRP3 and therefore exacerbating LPC-induced NLRP3 inflammasome activation, macrophage pyroptosis and systemic inflammatory injury.


Sujet(s)
Protéine associée au récepteur du TNF induit par les corticoïdes , Lysolécithine , Macrophages , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Maturation post-traductionnelle des protéines , Pyroptose , Sepsie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Animaux , Sepsie/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Lysolécithine/métabolisme , Souris , Protéine associée au récepteur du TNF induit par les corticoïdes/métabolisme , Inflammasomes/métabolisme , Mâle , Souris knockout , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Acétylation
3.
Exp Cell Res ; 439(1): 114068, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38750717

RÉSUMÉ

Acetylation, a critical regulator of diverse cellular processes, holds significant implications in various cancer contexts. Further understanding of the acetylation patterns of key cancer-driven proteins is crucial for advancing therapeutic strategies in cancer treatment. This study aimed to unravel the acetylation patterns of Engulfment and Cell Motility Protein 1 (ELMO1) and its relevance to the pathogenesis of colorectal cancer (CRC). Immunoprecipitation and mass spectrometry precisely identified lysine residue 505 (K505) as a central acetylation site in ELMO1. P300 emerged as the acetyltransferase for ELMO1 K505 acetylation, while SIRT2 was recognized as the deacetylase. Although K505 acetylation minimally affected ELMO1's localization and stability, it played a crucial role in mediating ELMO1-Dock180 interaction, thereby influencing Rac1 activation. Functionally, ELMO1 K505 acetylation proved to be a pivotal factor in CRC progression, exerting its influence on key cellular processes. Clinical analysis of CRC samples unveiled elevated ELMO1 acetylation in primary tumors, indicating a potential association with CRC pathologies. This work provides insights into ELMO1 acetylation and its significance in advancing potentially therapeutic interventions in CRC treatment.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Tumeurs colorectales , Protéine G rac1 , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Acétylation , Protéine G rac1/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Lignée cellulaire tumorale , Évolution de la maladie , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Mouvement cellulaire , Cellules HCT116
4.
Gynecol Endocrinol ; 40(1): 2353733, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38818662

RÉSUMÉ

BACKGROUND: Polycystic ovarian syndrome (PCOS) is a prevalent metabolic and endocrine condition in females of reproductive age. This work was to discover the underlying role of Dickkopf 1 (DKK1) and its putative regulating mechanism in P COS. METHODS: Mice recieved dehydroepiandrosterone (DHEA) injection to establish the in vivo P COS model.Hematoxylin and eosin (H&E) staining was performed for histological analysis. RT-qP CR and Western blotting were used to detect gene and protein expression. CCK-8 and flow cytometry assays were applied to detect cell viability and apoptosis. Co-immunoprecipitation (Co-IP) and immunoprecipitation (IP) were applied to assess association between DKK1 and SIRT2. RESULTS: In this work, DKK1 is downregulated in P COS rats. It was revealed that DKK1 knockdown induced apoptosis and suppressed proliferation in KGN cells, whereas DKK1 overexpression had exactly the opposite effects. In addition, DKK1 deactivates the T GF-ß1/SMad3 signaling pathway, thereby controlling KGN cell proliferation and apoptosis. Besides, SIRT2 inhibition reversed the impact of DKK1 overexpression on KGN cell proliferation and apoptosis. Furthermore, SIRT2 downregulated DKK1 expression by deacetylating DKK1 in KGN cells. DISCUSSION: Altogether, we concluded that SIRT2-induced deacetylation of DKK1 triggers T GF-ß1/Smad3 hyperactivation, thereby inhibiting proliferation and promoting apoptosis of KGN cells. The above results indicated that DKK1 might function as a latent target for P COS treatment.


Sujet(s)
Protéines et peptides de signalisation intercellulaire , Syndrome des ovaires polykystiques , Transduction du signal , Sirtuine-2 , Protéine Smad-3 , Facteur de croissance transformant bêta-1 , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/génétique , Femelle , Animaux , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines et peptides de signalisation intercellulaire/génétique , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Souris , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Rats , Apoptose , Acétylation , Prolifération cellulaire , Modèles animaux de maladie humaine , Humains
5.
Biogerontology ; 25(4): 705-737, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38619670

RÉSUMÉ

Polyalthia longifolia is well-known for its abundance of polyphenol content and traditional medicinal uses. Previous research has demonstrated that the methanolic extract of P. longifolia leaves (PLME, 1 mg/mL) possesses anti-aging properties in Saccharomyces cerevisiae BY611 yeast cells. Building on these findings, this study delves deeper into the potential antiaging mechanism of PLME, by analyzing the transcriptional responses of BY611 cells treated with PLME using RNA-sequencing (RNA-seq) technology. The RNA-seq analysis results identified 1691 significantly (padj < 0.05) differentially expressed genes, with 947 upregulated and 744 downregulated genes. Notably, the expression of three important aging-related genes, SIR2, SOD1, and SOD2, showed a significant difference following PLME treatment. The subsequent integration of these targeted genes with GO and KEGG pathway analysis revealed the multifaceted nature of PLME's anti-aging effects in BY611 yeast cells. Enriched GO and KEGG analysis showed that PLME treatment promotes the upregulation of SIR2, SOD1, and SOD2 genes, leading to a boosted cellular antioxidant defense system, reduced oxidative stress, regulated cell metabolism, and maintain genome stability. These collectively increased longevities in PLME-treated BY611 yeast cells and indicate the potential anti-aging action of PLME through the modulation of SIR2 and SOD genes. The present study provided novel insights into the roles of SIR2, SOD1, and SOD2 genes in the anti-aging effects of PLME treatment, offering promising interventions for promoting healthy aging.


Sujet(s)
Extraits de plantes , Feuilles de plante , Polyalthia , Saccharomyces cerevisiae , Protéines SIR de Saccharomyces cerevisiae , Sirtuine-2 , Superoxide dismutase , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Superoxide dismutase/métabolisme , Superoxide dismutase/génétique , Sirtuine-2/génétique , Sirtuine-2/métabolisme , Protéines SIR de Saccharomyces cerevisiae/génétique , Protéines SIR de Saccharomyces cerevisiae/métabolisme , Analyse de séquence d'ARN/méthodes , Méthanol/composition chimique , Vieillissement/effets des médicaments et des substances chimiques , Vieillissement/génétique , Régulation de l'expression des gènes fongiques/effets des médicaments et des substances chimiques , Superoxide dismutase-1/génétique , Superoxide dismutase-1/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme
6.
mBio ; 15(6): e0044524, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38682948

RÉSUMÉ

Histone deacetylation affects Candida albicans (C. albicans) pathogenicity by modulating virulence factor expression and DNA damage. The histone deacetylase Sir2 is associated with C. albicans plasticity and maintains genome stability to help C. albicans adapt to various environmental niches. However, whether Sir2-mediated chromatin modification affects C. albicans virulence is unclear. The purpose of our study was to investigate the effect of Sir2 on C. albicans pathogenicity and regulation. Here, we report that Sir2 is required for C. albicans pathogenicity, as its deletion affects the survival rate, fungal burden in different organs and the extent of tissue damage in a mouse model of disseminated candidiasis. We evaluated the impact of Sir2 on C. albicans virulence factors and revealed that the Sir2 null mutant had an impaired ability to adhere to host cells and was more easily recognized by the innate immune system. Comprehensive analysis revealed that the disruption of C. albicans adhesion was due to a decrease in cell surface hydrophobicity rather than the differential expression of adhesion genes on the cell wall. In addition, Sir2 affects the distribution and exposure of mannan and ß-glucan on the cell wall, indicating that Sir2 plays a role in preventing the immune system from recognizing C. albicans. Interestingly, our results also indicated that Sir2 helps C. albicans maintain metabolic activity under hypoxic conditions, suggesting that Sir2 contributes to C. albicans colonization at hypoxic sites. In conclusion, our findings provide detailed insights into antifungal targets and a useful foundation for the development of antifungal drugs. IMPORTANCE: Candida albicans (C. albicans) is the most common opportunistic fungal pathogen and can cause various superficial infections and even life-threatening systemic infections. To successfully propagate infection, this organism relies on the ability to express virulence-associated factors and escape host immunity. In this study, we demonstrated that the histone deacetylase Sir2 helps C. albicans adhere to host cells and escape host immunity by mediating cell wall remodeling; as a result, C. albicans successfully colonized and invaded the host in vivo. In addition, we found that Sir2 contributes to carbon utilization under hypoxic conditions, suggesting that Sir2 is important for C. albicans survival and the establishment of infection in hypoxic environments. In summary, we investigated the role of Sir2 in regulating C. albicans pathogenicity in detail; these findings provide a potential target for the development of antifungal drugs.


Sujet(s)
Candida albicans , Candidose , Paroi cellulaire , Échappement immunitaire , Sirtuine-2 , Candida albicans/génétique , Candida albicans/pathogénicité , Candida albicans/immunologie , Paroi cellulaire/métabolisme , Animaux , Candidose/microbiologie , Candidose/immunologie , Souris , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Facteurs de virulence/métabolisme , Facteurs de virulence/génétique , Virulence , Modèles animaux de maladie humaine , Délétion de gène , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Souris de lignée BALB C , Femelle
7.
EMBO Rep ; 25(5): 2441-2478, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38649663

RÉSUMÉ

Ago2 differentially regulates oncogenic and tumor-suppressive miRNAs in cancer cells. This discrepancy suggests a secondary event regulating Ago2/miRNA action in a context-dependent manner. We show here that a positive charge of Ago2 K212, that is preserved by SIR2-mediated Ago2 deacetylation in cancer cells, is responsible for the direct interaction between Ago2 and Caveolin-1 (CAV1). Through this interaction, CAV1 sequesters Ago2 on the plasma membranes and regulates miRNA-mediated translational repression in a compartment-dependent manner. Ago2/CAV1 interaction plays a role in miRNA-mediated mRNA suppression and in miRNA release via extracellular vesicles (EVs) from tumors into the circulation, which can be used as a biomarker of tumor progression. Increased Ago2/CAV1 interaction with tumor progression promotes aggressive cancer behaviors, including metastasis. Ago2/CAV1 interaction acts as a secondary event in miRNA-mediated suppression and increases the complexity of miRNA actions in cancer.


Sujet(s)
Protéines Argonaute , Cavéoline-1 , microARN , Métastase tumorale , Protéines Argonaute/métabolisme , Protéines Argonaute/génétique , microARN/métabolisme , microARN/génétique , Cavéoline-1/métabolisme , Cavéoline-1/génétique , Humains , Lignée cellulaire tumorale , Animaux , Régulation de l'expression des gènes tumoraux , Vésicules extracellulaires/métabolisme , Souris , Liaison aux protéines , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Sirtuine-2/métabolisme , Sirtuine-2/génétique
8.
J Biol Chem ; 300(5): 107273, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38588806

RÉSUMÉ

The stability of ribosomal DNA (rDNA) is maintained through transcriptional silencing by the NAD+-dependent histone deacetylase Sir2 in Saccharomyces cerevisiae. Alongside proteostasis, rDNA stability is a crucial factor regulating the replicative lifespan of S. cerevisiae. The unfolded protein response (UPR) is induced by misfolding of proteins or an imbalance of membrane lipid composition and is responsible for degrading misfolded proteins and restoring endoplasmic reticulum (ER) membrane homeostasis. Recent investigations have suggested that the UPR can extend the replicative lifespan of yeast by enhancing protein quality control mechanisms, but the relationship between the UPR and rDNA stability remains unknown. In this study, we found that the deletion of ARV1, which encodes an ER protein of unknown molecular function, activates the UPR by inducing lipid bilayer stress. In arv1Δ cells, the UPR and the cell wall integrity pathway are activated independently of each other, and the high osmolarity glycerol (HOG) pathway is activated in a manner dependent on Ire1, which mediates the UPR. Activated Hog1 translocates the stress response transcription factor Msn2 to the nucleus, where it promotes the expression of nicotinamidase Pnc1, a well-known Sir2 activator. Following Sir2 activation, rDNA silencing and rDNA stability are promoted. Furthermore, the loss of other ER proteins, such as Pmt1 or Bst1, and ER stress induced by tunicamycin or inositol depletion also enhance rDNA stability in a Hog1-dependent manner. Collectively, these findings suggest that the induction of the UPR enhances rDNA stability in S. cerevisiae by promoting the Msn2-Pnc1-Sir2 pathway in a Hog1-dependent manner.


Sujet(s)
ADN ribosomique , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Réponse aux protéines mal repliées , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , ADN ribosomique/métabolisme , ADN ribosomique/génétique , Double couche lipidique/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Mitogen-Activated Protein Kinases/métabolisme , Mitogen-Activated Protein Kinases/génétique , Nicotinamidase/métabolisme , Nicotinamidase/génétique , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Protéines SIR de Saccharomyces cerevisiae/métabolisme , Protéines SIR de Saccharomyces cerevisiae/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Réticulum endoplasmique/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Glycoprotéines membranaires
9.
Protein Sci ; 33(5): e4994, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38647411

RÉSUMÉ

Sirtuin 2 (SIRT2) is a class III histone deacetylase that is highly conserved from bacteria to mammals. We prepared and characterized the wild-type (WT) and mutant forms of the histone deacetylase (HDAC) domain of human SIRT2 (hSIRT2) using various biophysical methods and evaluated their deacetylation activity. We found that WT hSIRT2 HDAC (residues 52-357) forms a homodimer in a concentration-dependent manner with a dimer-monomer dissociation constant of 8.3 ± 0.5 µM, which was determined by mass spectrometry. The dimer was disrupted into two monomers by binding to the HDAC inhibitors SirReal1 and SirReal2. We also confirmed dimer formation of hSIRT2 HDAC in living cells using a NanoLuc complementation reporter system. Examination of the relationship between dimer formation and deacetylation activity using several mutants of hSIRT2 HDAC revealed that some non-dimerizing mutants exhibited deacetylation activity for the N-terminal peptide of histone H3, similar to the wild type. The hSIRT2 HDAC mutant Δ292-306, which lacks a SIRT2-specific disordered loop region, was identified to exist as a monomer with slightly reduced deacetylation activity; the X-ray structure of the mutant Δ292-306 was almost identical to that of the WT hSIRT2 HDAC bound to an inhibitor. These results indicate that hSIRT2 HDAC forms a dimer, but this is independent of deacetylation activity. Herein, we discuss insights into the dimer formation of hSIRT2 based on our biophysical experimental results.


Sujet(s)
Multimérisation de protéines , Sirtuine-2 , Humains , Sirtuine-2/métabolisme , Sirtuine-2/composition chimique , Sirtuine-2/génétique , Acétylation , Cellules HEK293
10.
Proc Natl Acad Sci U S A ; 121(18): e2319833121, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38648480

RÉSUMÉ

Sirt2 is a nicotinamide adenine dinucleotide (NAD+)-dependent protein lysine deacylase that can remove both acetyl group and long-chain fatty acyl groups from lysine residues of many proteins. It was reported to affect inflammatory bowel disease (IBD) symptoms in a mouse model. However, conflicting roles were reported, with genetic knockout aggravating while pharmacological inhibition alleviating IBD symptoms. These seemingly conflicting reports cause confusion and deter further efforts in developing Sirt2 inhibitors as a potential treatment strategy for IBD. We investigated these conflicting reports and elucidated the role of Sirt2 in the mouse model of IBD. We essentially replicated these conflicting results and confirmed that Sirt2 inhibitors' protective effect is not through off-targets as two very different Sirt2 inhibitors (TM and AGK2) showed similar protection in the IBD mouse model. We believe that the differential effects of inhibitors and knockout are due to the fact that the Sirt2 inhibitors only inhibit some but not all the activities of Sirt2. This hypothesis is confirmed by the observation that a PROTAC degrader of Sirt2 did not protect mice in the IBD model, similar to Sirt2 knockout. Our study provides an interesting example where genetic knockout and pharmacological inhibition do not align and emphasizes the importance of developing substrate-dependent inhibitors. Importantly, we showed that the effect of Sirt2 inhibition in IBD is through regulating the gut epithelium barrier by inhibiting Arf6-mediated endocytosis of E-cadherin, a protein important for the intestinal epithelial integrity. This mechanistic understanding further supports Sirt2 as a promising therapeutic target for treating IBD.


Sujet(s)
Colite , Muqueuse intestinale , Sirtuine-2 , Animaux , Humains , Souris , Cadhérines/métabolisme , Cadhérines/génétique , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/prévention et contrôle , Modèles animaux de maladie humaine , Furanes , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Souris de lignée C57BL , Souris knockout , Quinoléines , Sirtuine-2/métabolisme , Sirtuine-2/antagonistes et inhibiteurs , Sirtuine-2/génétique
11.
Gene ; 915: 148428, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38575099

RÉSUMÉ

To assess and validate the gene expression profile of SIRTs (SIRT1, SIRT2, SIRT3, SIRT4, SIRT5, SIRT6, and SIRT7) in relation to the pathogenesis and prognostic progression of Myelodysplastic neoplasm (MDS). Eighty bone marrow samples of patients with de novo MDS were diagnosed according to WHO 2022 and IPSS-R criteria. Ten bone marrow samples were obtained from elderly healthy volunteers and used as control samples. Gene expression levels of all SIRTs were assessed using RT-qPCR assays. Downregulation of SIRT2 (p = 0.009), SIRT3 (p = 0.048), SIRT4 (p = 0.049), SIRT5 (p = 0.046), SIRT6 (p = 0.043), and SIRT7 (p = 0.047) was identified in MDS patients compared to control individuals. Also, we identified that while SIRT2-7 genes are typically down-regulated in MDS patients compared to normal controls, there are relative expression variations among MDS patient subgroups. Specifically, SIRT4 (p = 0.029) showed increased expression in patients aged 60 or above, and both SIRT2 (p = 0.016) and SIRT3 (p = 0.036) were upregulated in patients with hemoglobin levels below 8 g/dL. SIRT2 (p = 0.045) and SIRT3 (p = 0.033) were highly expressed in patients with chromosomal abnormalities. Different SIRTs exhibited altered expression patterns concerning specific MDS clinical and prognostic characteristics. The downregulation in SIRTs genes (e.g., SIRT2 to SIRT7) expression in Brazilian MDS patients highlights their role in the disease's development. The upregulation of SIRT2 and SIRT3 in severe anemia patients suggests a potential link to manage iron overload-related complications in transfusion-dependent patients. Moreover, the association of SIRT2/SIRT3 with genomic instability and their role in MDS progression signify promising areas for future research and therapeutic targets. These findings underscore the importance of SIRT family in understanding and addressing MDS, offering novel clinical, prognostic, and therapeutic insights for patients with this condition.


Sujet(s)
Protéines mitochondriales , Syndromes myélodysplasiques , Sirtuine-3 , Sirtuines , Humains , Sirtuines/génétique , Sirtuines/métabolisme , Mâle , Femelle , Sujet âgé , Adulte d'âge moyen , Syndromes myélodysplasiques/génétique , Pronostic , Sirtuine-3/génétique , Sirtuine-3/métabolisme , Sirtuine-2/génétique , Sirtuine-2/métabolisme , Adulte , Sujet âgé de 80 ans ou plus , Sirtuine-1/génétique , Sirtuine-1/métabolisme , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes/méthodes , Études cas-témoins
12.
Theranostics ; 14(6): 2622-2636, 2024.
Article de Anglais | MEDLINE | ID: mdl-38646657

RÉSUMÉ

Rationale: In recent years, nicotinamide adenine dinucleotide (NAD+) precursors (Npre) have been widely employed to ameliorate female reproductive problems in both humans and animal models. However, whether and how Npre plays a role in the male reproductive disorder has not been fully clarified. Methods: In the present study, a busulfan-induced non-obstructive azoospermic mouse model was used, and Npre was administered for five weeks following the drug injection, with the objective of reinstating spermatogenesis and fertility. Initially, we assessed the NAD+ level, germ cell types, semen parameters and sperm fertilization capability. Subsequently, testis tissues were examined through RNA sequencing analysis, ELISA, H&E, immunofluorescence, quantitative real-time PCR, and Western blotting techniques. Results: The results indicated that Npre restored normal level of NAD+ in blood and significantly alleviated the deleterious effects of busulfan (BU) on spermatogenesis, thereby partially reestablishing fertilization capacity. Transcriptome analysis, along with recovery of testicular Fe2+, GSH, NADPH, and MDA levels, impaired by BU, and the fact that Fer-1, an inhibitor of ferroptosis, restored spermatogenesis and semen parameters close to CTRL values, supported such possibility. Interestingly, the reduction in SIRT2 protein level by the specific inhibitor AGK2 attenuated the beneficial effects of Npre on spermatogenesis and ferroptosis by affecting PGC-1α and ACLY protein levels, thus suggesting how these compounds might confer spermatogenesis protection. Conclusion: Collectively, these findings indicate that NAD+ protects spermatogenesis against ferroptosis, probably through SIRT2 dependent mechanisms. This underscores the considerable potential of Npre supplementation as a feasible strategy for preserving or restoring spermatogenesis in specific conditions of male infertility and as adjuvant therapy to preserve male fertility in cancer patients receiving sterilizing treatments.


Sujet(s)
Busulfan , Ferroptose , NAD , Sirtuine-2 , Spermatogenèse , Animaux , Busulfan/pharmacologie , Mâle , Spermatogenèse/effets des médicaments et des substances chimiques , Souris , NAD/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Sirtuine-2/métabolisme , Sirtuine-2/génétique , Modèles animaux de maladie humaine , Testicule/métabolisme , Testicule/effets des médicaments et des substances chimiques , Azoospermie/traitement médicamenteux , Azoospermie/métabolisme , Azoospermie/induit chimiquement
13.
Nucleic Acids Res ; 52(9): 5107-5120, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38554113

RÉSUMÉ

Sirtuin 2 (SIRT2) regulates the maintenance of genome integrity by targeting pathways of DNA damage response and homologous recombination repair. However, whether and how SIRT2 promotes base excision repair (BER) remain to be determined. Here, we found that independent of its catalytic activity SIRT2 interacted with the critical glycosylase OGG1 to promote OGG1 recruitment to its own promoter upon oxidative stress, thereby enhancing OGG1 promoter activity and increasing BER efficiency. Further studies revealed that SIRT2 was phosphorylated on S46 and S53 by ATM/ATR upon oxidative stress, and SIRT2 phosphorylation enhanced the SIRT2-OGG1 interaction and mediated the stimulatory effect of SIRT2 on OGG1 promoter activity. We also characterized 37 cancer-derived SIRT2 mutants and found that 5 exhibited the loss of the stimulatory effects on OGG1 transcription. Together, our data reveal that SIRT2 acts as a tumor suppressor by promoting OGG1 transcription and increasing BER efficiency in an ATM/ATR-dependent manner.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , DNA Glycosylases , Réparation de l'ADN , Sirtuine-2 , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/génétique , Humains , Sirtuine-2/métabolisme , Sirtuine-2/génétique , DNA Glycosylases/métabolisme , DNA Glycosylases/génétique , Phosphorylation , Régions promotrices (génétique) , Stress oxydatif , Activation de la transcription , Cellules HEK293 , Altération de l'ADN , Transcription génétique , Lignée cellulaire tumorale , Réparation par excision
14.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167129, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38513990

RÉSUMÉ

Bone cancer pain (BCP) is refractory to currently used analgesics. Recently, sirtuin 2 (SIRT2) was reported to play a vital role in neuropathic pain but its role in BCP remains unknown. It was hypothesized that spinal SIRT2 attenuates BCP by deacetylating FoxO3a and suppressing oxidative stress. The mouse model of BCP established by injecting tumor cells into the intramedullary space of the femur demonstrated that spinal SIRT2 and FoxO3a were downregulated in BCP development. Intrathecal administration of LV-SIRT2 reduced pain hypersensitivity (mechanical and thermal nociception) in BCP mice. Spinal SIRT2 overexpression upregulated FoxO3a and antioxidant genes (SOD2 and catalase) and inhibited FoxO3a acetylation, phosphorylation, and ubiquitination. Moreover, intrathecal administration of SIRT2 shRNA induced pain hypersensitivity in normal mice. Spinal SIRT2 knockdown downregulated FoxO3a and antioxidant genes and increased FoxO3a acetylation, phosphorylation, and ubiquitination. In summary, spinal SIRT2 increases FoxO3a expression in BCP mice and inhibits oxidative stress by deacetylating FoxO3a and further reducing FoxO3a phosphorylation, ubiquitination, and degradation, leading to BCP relief.


Sujet(s)
Tumeurs osseuses , Douleur cancéreuse , Névralgie , Animaux , Souris , Antioxydants , Tumeurs osseuses/complications , Tumeurs osseuses/génétique , Douleur cancéreuse/génétique , Douleur cancéreuse/métabolisme , Sirtuine-2/génétique
15.
J Cell Mol Med ; 28(6): e18129, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38426936

RÉSUMÉ

ATP citrate lyase (ACLY), as a key enzyme in lipid metabolism, plays an important role in energy metabolism and lipid biosynthesis of a variety of tumours. Many studies have shown that ACLY is highly expressed in various tumours, and its pharmacological or gene inhibition significantly inhibits tumour growth and progression. However, the roles of ACLY in oesophageal squamous cell carcinoma (ESCC) remain unclear. Here, our data showed that ACLY inhibitor significantly attenuated cell proliferation, migration, invasion and lipid synthesis in different ESCC cell lines, whereas the proliferation, migration, invasion and lipid synthesis of ESCC cells were enhanced after ACLY overexpression. Furthermore, ACLY inhibitor dramatically suppressed tumour growth and lipid metabolism in ESCC cells xenografted tumour model, whereas ACLY overexpression displayed the opposite effect. Mechanistically, ACLY protein harboured acetylated modification and interacted with SIRT2 protein in ESCC cells. The SIRT2 inhibitor AGK2 significantly increased the acetylation level of ACLY protein and inhibited the proliferation and migration of ESCC cells, while overexpression of ACLY partially reversed the inhibitory effect of AGK2 on ESCC cells. Overall, these results suggest that targeting the SIRT2/ACLY signalling axis may be a potential therapeutic strategy for ESCC patients.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Humains , Carcinome épidermoïde de l'oesophage/génétique , ATP citrate (pro-S)-lyase , Sirtuine-2/génétique , Sirtuine-2/métabolisme , Prolifération cellulaire , Tumeurs de l'oesophage/métabolisme , Lipides , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux
16.
Immun Inflamm Dis ; 12(2): e1160, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38415949

RÉSUMÉ

INTRODUCTION: Regulatory T cells (Tregs) play an important role in inflammatory bowel diseases (IBDs) through modulating intestinal inflammation. However, the factors affecting Treg function and plasticity during IBD progression are not thoroughly disclosed. The current study aims to reveal new molecular mechanisms affecting Treg plasticity. METHODS: A mouse strain, in which tdTomato and enhanced green fluorescent protein were under the control of the Foxp3 promoter and Il17a promoter, was established and subjected to colitis induction with dextran sulfate sodium. The existence of Tregs and IL-17-expressing Tregs (i.e., Treg/T helper 17 [Th17] cells) were observed and sorted from the spleen, mesenteric lymph nodes, and lamina propria by flow cytometry, followed by measuring Sirtuin2 (Sirt2) expression using quantitative reverse transcription polymerase chain reaction and Immunoblotting. Lentivirus-induced Sirt2 silencing was applied to determine the impact of Sirt2 on Treg polarization to Treg/Th17 cells and even Th17 cells. The effect of Sirt2 on Stat3 was analyzed by flow cytometry and immunoblotting. RESULTS: Sirt2 was highly expressed in lamina propria Tregs and it moderately suppressed Foxp3 expression as well as the immunosuppressive function of Tregs. Surprisingly, lentivirus-mediated Sirt2 silencing promoted the generation of Treg/Th17 cells out of Tregs. Sirt2 silencing also enhanced the generation of Th17 cells out of Tregs under the Th17 induction condition. Furthermore, Sirt2 inhibited Th17 induction by suppressing the protein level of the signal transducer and activator of transcription 3. CONCLUSION: Sirt2 suppresses Treg function but also inhibits Treg polarization toward Treg/Th17 cells and Th17 cells. The ultimate effect of Sirt2 on colitis might depend on the balance among Tregs, Treg/Th17 cells, and Th17 cells.


Sujet(s)
Colite , , Facteur de transcription STAT-3 , Animaux , Souris , Facteur de transcription STAT-3/génétique , Lymphocytes T régulateurs , Cellules Th17 , Sirtuine-2/génétique , Colite/induit chimiquement , Colite/génétique , Modèles animaux de maladie humaine , Facteurs de transcription Forkhead/génétique
17.
Gene ; 907: 148276, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38360128

RÉSUMÉ

Cold is a common stressor that threatens colonic health by affecting internal homeostasis. From the literature, Silent information regulator 2 (SIRT2) may have important roles during cold stress, but this conjecture requires investigation. To address this knowledge gap, we investigated the effects of SIRT2 on colonic injury in chronically cold-exposure mice. In a previous study, we showed that SIRT2 regulated p65 activation after cold exposure. In the current study, mice were exposed to 4 °C for 3 h/day for 3 weeks to simulate a chronic cold exposure environment. Chronic cold exposure shortened colon length, disrupted tight junctions in colonic epithelial tissue, and disordered colonic flora. Chronic cold exposure also increased p65 acetylation levels, promoted nuclear factor (NF)-κB activation, and increased the expression of its downstream pro-inflammatory factors, while SIRT2 knockdown aggravated the consequences of tissue structure disruption and increased inflammatory factors brought about by chronic cold exposure to some extent, but could alleviate the downregulation of colonic tight junction-related proteins to some extent. We also observed direct SIRT2 regulatory effects toward p65, and in Caco-2 cells treated with lipopolysaccharide (LPS), SIRT2 knockdown increased p65 acetylation levels and pro-inflammatory factor expression, while SIRT2 overexpression reversed these phenomena. Therefore, SIRT2 deletion exacerbated chronic cold exposure-induced colonic injury and p65 activation in mice. Mechanistically, p65 modification by SIRT2 via deacetylation may affect NF-κB signaling. These findings suggest that SIRT2 is a key target of colonic health maintenance under chronic cold exposure conditions.


Sujet(s)
Côlon , Facteur de transcription NF-kappa B , Sirtuine-2 , Animaux , Humains , Souris , Cellules Caco-2 , Lipopolysaccharides/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal , Sirtuine-2/génétique , Facteur de transcription RelA/métabolisme , Côlon/traumatismes , Côlon/anatomopathologie , Basse température/effets indésirables
18.
BMC Med Genomics ; 17(1): 6, 2024 01 02.
Article de Anglais | MEDLINE | ID: mdl-38167011

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disease that can cause dementia. We aim to screen out the hub genes involved in AD based on microarray datasets. METHODS: Gene expression profiles GSE5281 and GSE28146 were retrieved from Gene Expression Omnibus database to acquire differentially expressed genes (DEGs). Gene Ontology and pathway enrichment were conducted using DAVID online tool. The STRING database and Cytoscape tools were employed to analyze protein-protein interactions and identify hub genes. The predictive value of hub genes was assessed by principal component analysis and receiver operating characteristic curves. AD mice model was constructed, and histology was then observed by hematoxylin-eosin staining. Gene expression levels were finally determined by real-time quantitative PCR. RESULTS: We obtained 197 overlapping DEGs from GSE5281 and GSE28146 datasets. After constructing protein-protein interaction network, three highly interconnected clusters were identified and 6 hub genes (RBL1, BUB1, HDAC7, KAT5, SIRT2, and ITGB1) were selected. The hub genes could be used as basis to predict AD. Histological abnormalities of brain were observed, suggesting successful AD model was constructed. Compared with the control group, the mRNA expression levels of RBL1, BUB1, HDAC7, KAT5 and SIRT2 were significantly increased, while the mRNA expression level of ITGB1 was significantly decreased in AD groups. CONCLUSION: RBL1, BUB1, HDAC7, KAT5, SIRT2 and ITGB1 are promising gene signatures for diagnosis and therapy of AD.


Sujet(s)
Maladie d'Alzheimer , Maladies neurodégénératives , Animaux , Souris , Sirtuine-2/génétique , Analyse de profil d'expression de gènes , Maladie d'Alzheimer/diagnostic , Maladie d'Alzheimer/génétique , Cartes d'interactions protéiques/génétique , Biologie informatique , ARN messager , Réseaux de régulation génique
19.
Front Biosci (Landmark Ed) ; 29(1): 27, 2024 01 18.
Article de Anglais | MEDLINE | ID: mdl-38287804

RÉSUMÉ

BACKGROUND: The pentose phosphate pathway (PPP) is a critical metabolic pathway that generates NADPH and ribose-5-phosphate for nucleotide biosynthesis and redox homeostasis. In this study, we investigated a potential regulatory role for Krüppel-like factor 8 (KLF8) in the control of PPP in lung adenocarcinoma (LUAD) cells. METHODS: Based on a comprehensive set of experimental approaches, including cell culture, molecular techniques, and functional assays, we revealed a novel mechanism by which KLF8 promotes the activation of glucose-6-phosphate dehydrogenase (G6PD), a component enzyme in the PPP. RESULTS: Our findings demonstrate that KLF8 inhibits the acetylation of G6PD, leading to its increased enzymatic activity. Additionally, we observed that KLF8 activates the transcription of SIRT2, which has been implicated in regulating G6PD acetylation. These results highlight the interplay between KLF8, G6PD, and protein acetylation in the regulation of PPP in LUAD. CONCLUSIONS: Understanding the intricate molecular mechanisms underlying the metabolic reprogramming driven by KLF8 in lung cancer provides valuable insights into potential therapeutic strategies targeting the PPP. This study emphasizes the significance of KLF8 as a key modulator of metabolic pathways and indicates the potential of targeting the KLF8-G6PD axis for lung cancer treatment.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Humains , Voie des pentoses phosphates/physiologie , Sirtuine-2/génétique , Sirtuine-2/métabolisme , Adénocarcinome pulmonaire/génétique , Tumeurs du poumon/génétique , Poumon/métabolisme , Facteurs de transcription Krüppel-like/génétique , Facteurs de transcription Krüppel-like/métabolisme
20.
Neuro Oncol ; 26(1): 55-67, 2024 01 05.
Article de Anglais | MEDLINE | ID: mdl-37625115

RÉSUMÉ

BACKGROUND: Functional inactivation of ATRX characterizes large subgroups of malignant gliomas in adults and children. ATRX deficiency in glioma induces widespread chromatin remodeling, driving transcriptional shifts and oncogenic phenotypes. Effective strategies to therapeutically target these broad epigenomic sequelae remain undeveloped. METHODS: We utilized integrated multiomics and the Broad Institute Connectivity Map (CMAP) to identify drug candidates that could potentially revert ATRX-deficient transcriptional changes. We then employed disease-relevant experimental models to evaluate functional phenotypes, coupling these studies with epigenomic profiling to elucidate molecular mechanism(s). RESULTS: CMAP analysis and transcriptional/epigenomic profiling implicated the Class III HDAC Sirtuin2 (SIRT2) as a central mediator of ATRX-deficient cellular phenotypes and a driver of unfavorable prognosis in ATRX-deficient glioma. SIRT2 inhibitors reverted Atrx-deficient transcriptional signatures in murine neuroepithelial progenitor cells (mNPCs), impaired cell migration in Atrx/ATRX-deficient mNPCs and human glioma stem cells (GSCs), and increased expression of senescence markers in glioma models. Moreover, SIRT2 inhibition impaired growth and increased senescence in ATRX-deficient GSCs in vivo. These effects were accompanied by genome-wide shifts in enhancer-associated H3K27ac and H4K16ac marks, with the latter in particular demonstrating compelling transcriptional links to SIRT2-dependent phenotypic reversals. Motif analysis of these data identified the transcription factor KLF16 as a mediator of phenotype reversal in Atrx-deficient cells upon SIRT2 inhibition. CONCLUSIONS: Our findings indicate that SIRT2 inhibition selectively targets ATRX-deficient gliomas for senescence through global chromatin remodeling, while demonstrating more broadly a viable approach to combat complex epigenetic rewiring in cancer.


Sujet(s)
Chromatine , Gliome , Adulte , Enfant , Humains , Animaux , Souris , Sirtuine-2/génétique , Sirtuine-2/métabolisme , Gliome/anatomopathologie , Protéine nucléaire liée à l'X/génétique , Facteurs de transcription Krüppel-like/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...