Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.745
Filter
1.
Toxicol Appl Pharmacol ; 489: 117019, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950736

ABSTRACT

Maternal hypoxia is strongly linked to insulin resistance (IR) in adult offspring, and altered insulin signaling for muscle glucose uptake is thought to play a central role. However, whether the SIRT3/GSK-3ß/GLUT4 axis is involved in maternal hypoxia-induced skeletal muscle IR in old male rat offspring has not been investigated. Maternal hypoxia was established from Days 5 to 21 of pregnancy by continuous infusion of nitrogen and air. The biochemical parameters and levels of key insulin signaling molecules of old male rat offspring were determined through a series of experiments. Compared to the control (Ctrl) old male rat offspring group, the hypoxic (HY) group exhibited elevated fasting blood glucose (FBG) (∼30%), fasting blood insulin (FBI) (∼35%), total triglycerides (TGs), and low-density lipoprotein cholesterol (LDL-C), as well as results showing impairment in the glucose tolerance test (GTT) and insulin tolerance test (ITT). In addition, hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) revealed impaired cellular structures and mitochondria in the longitudinal sections of skeletal muscle from HY group mice, which might be associated with decreased SIRT3 expression. Furthermore, the expression of insulin signaling molecules, such as GSK-3ß and GLUT4, was also altered. In conclusion, the present results indicate that the SIRT3/GSK-3ß/GLUT4 axis might be involved in maternal hypoxia-induced skeletal muscle IR in old male rat offspring.


Subject(s)
Glucose Transporter Type 4 , Glycogen Synthase Kinase 3 beta , Hypoxia , Insulin Resistance , Muscle, Skeletal , Sirtuin 3 , Animals , Male , Glycogen Synthase Kinase 3 beta/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Female , Glucose Transporter Type 4/metabolism , Pregnancy , Sirtuin 3/metabolism , Rats , Hypoxia/metabolism , Signal Transduction , Prenatal Exposure Delayed Effects/metabolism , Rats, Sprague-Dawley , Insulin/blood , Insulin/metabolism , Blood Glucose/metabolism , Sirtuins
2.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000044

ABSTRACT

Kidney diseases, including chronic kidney disease (CKD), diabetic nephropathy, and acute kidney injury (AKI), represent a significant global health burden. The kidneys are metabolically very active organs demanding a large amount of ATP. They are composed of highly specialized cell types in the glomerulus and subsequent tubular compartments which fine-tune metabolism to meet their numerous and diverse functions. Defective renal cell metabolism, including altered fatty acid oxidation or glycolysis, has been linked to both AKI and CKD. Mitochondria play a vital role in renal metabolism, and emerging research has identified mitochondrial sirtuins (SIRT3, SIRT4 and SIRT5) as key regulators of renal cell metabolic adaptation, especially SIRT3. Sirtuins belong to an evolutionarily conserved family of mainly NAD+-dependent deacetylases, deacylases, and ADP-ribosyl transferases. Their dependence on NAD+, used as a co-substrate, directly links their enzymatic activity to the metabolic status of the cell. In the kidney, SIRT3 has been described to play crucial roles in the regulation of mitochondrial function, and the antioxidative and antifibrotic response. SIRT3 has been found to be constantly downregulated in renal diseases. Genetic or pharmacologic upregulation of SIRT3 has also been associated with beneficial renal outcomes. Importantly, experimental pieces of evidence suggest that SIRT3 may act as an important energy sensor in renal cells by regulating the activity of key enzymes involved in metabolic adaptation. Activation of SIRT3 may thus represent an interesting strategy to ameliorate renal cell energetics. In this review, we discuss the roles of SIRT3 in lipid and glucose metabolism and in mediating a metabolic switch in a physiological and pathological context. Moreover, we highlight the emerging significance of other mitochondrial sirtuins, SIRT4 and SIRT5, in renal metabolism. Understanding the role of mitochondrial sirtuins in kidney diseases may also open new avenues for innovative and efficient therapeutic interventions and ultimately improve the management of renal injuries.


Subject(s)
Kidney Diseases , Kidney , Mitochondria , Sirtuin 3 , Sirtuins , Humans , Sirtuins/metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Mitochondria/metabolism , Animals , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics
3.
Redox Biol ; 74: 103224, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38865904

ABSTRACT

BACKGROUND: Silicosis, characterized by interstitial lung inflammation and fibrosis, poses a significant health threat. ATII cells play a crucial role in alveolar epithelial repair and structural integrity maintenance. Inhibiting ATII cell senescence has shown promise in silicosis treatment. However, the mechanism behind silica-induced senescence remains elusive. METHODS: The study employed male C57BL/6 N mice and A549 human alveolar epithelial cells to investigate silicosis and its potential treatment. Silicosis was induced in mice via intratracheal instillation of crystalline silica particles, with honokiol administered intraperitoneally for 14 days. Silica-induced senescence in A549 cells was confirmed, and SIRT3 knockout and overexpression cell lines were generated. Various analyses were conducted, including immunoblotting, qRT-PCR, histology, and transmission electron microscopy. Statistical significance was determined using one-way ANOVA with Tukey's post-hoc test. RESULTS: This study elucidates how silica induces ATII cell senescence, emphasizing mtDNA damage. Notably, honokiol (HKL) emerges as a promising anti-senescence and anti-fibrosis agent, acting through sirt3. honokiol effectively attenuated senescence in ATII cells, dependent on sirt3 expression, while mitigating mtDNA damage. Sirt3, a class III histone deacetylase, regulates senescence and mitochondrial stress. HKL activates sirt3, protecting against pulmonary fibrosis and mitochondrial damage. Additionally, HKL downregulated cGAS expression in senescent ATII cells induced by silica, suggesting sirt3's role as an upstream regulator of the cGAS/STING signaling pathway. Moreover, honokiol treatment inhibited the activation of the NF-κB signaling pathway, associated with reduced oxidative stress and mtDNA damage. Notably, HKL enhanced the activity of SOD2, crucial for mitochondrial function, through sirt3-mediated deacetylation. Additionally, HKL promoted the deacetylation activity of sirt3, further safeguarding mtDNA integrity. CONCLUSIONS: This study uncovers a natural compound, HKL, with significant anti-fibrotic properties through activating sirt3, shedding light on silicosis pathogenesis and treatment avenues.


Subject(s)
Alveolar Epithelial Cells , Biphenyl Compounds , Cellular Senescence , Lignans , Signal Transduction , Silicosis , Sirtuin 3 , Animals , Silicosis/metabolism , Silicosis/drug therapy , Silicosis/pathology , Silicosis/etiology , Sirtuin 3/metabolism , Sirtuin 3/genetics , Cellular Senescence/drug effects , Mice , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Biphenyl Compounds/pharmacology , Humans , Lignans/pharmacology , Signal Transduction/drug effects , Male , A549 Cells , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Disease Models, Animal , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , DNA Damage/drug effects , Allyl Compounds , Phenols
4.
Chem Biol Interact ; 398: 111110, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38876248

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disorder that is linked to metabolic syndrome, mitochondrial dysfunction and impaired autophagy. Polydatin (PD), a natural polyphenol from Polygonum cuspidatum, exhibits various pharmacological effects and protects against NAFLD. The aim of this study was to reveal the molecular mechanisms and therapeutic potential of PD for NAFLD, with a focus on the role of mitochondrial autophagy mediated by sirtuin 3 (SIRT3), fork-head box O3 (FOXO3) and BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), and by PTEN-induced putative kinase 1 (PINK1) and parkin (PRKN). We combined network pharmacology analysis, animal models and cell culture experiments to show that PD could regulate the mitochondrial autophagy pathway by modulating several key genes related to mitochondrial function, and ameliorate the liver function, histopathology and mitochondrial biogenesis of NAFLD mice and hepatocytes by activating the SIRT3-FOXO3-BNIP3 axis and the PINK1-PRKN-dependent mechanism of mitochondrial autophagy. We also identified the core targets of PD, including SIRT3, FOXO3A, CASP3, PARKIN, EGFR, STAT3, MMP9 and PINK, and confirmed that silencing SIRT3 could significantly attenuate the beneficial effect of PD. This study provided novel theoretical and experimental support for PD as a promising candidate for NAFLD treatment, and also suggested new avenues and methods for investigating the role of mitochondrial autophagy in the pathogenesis and intervention of NAFLD.


Subject(s)
Forkhead Box Protein O3 , Glucosides , Mice, Inbred C57BL , Mitochondria , Non-alcoholic Fatty Liver Disease , Protein Kinases , Sirtuin 3 , Stilbenes , Ubiquitin-Protein Ligases , Animals , Forkhead Box Protein O3/metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Glucosides/pharmacology , Glucosides/therapeutic use , Glucosides/chemistry , Stilbenes/pharmacology , Stilbenes/therapeutic use , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Kinases/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Humans , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Autophagy/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Membrane Proteins
5.
Food Funct ; 15(12): 6692-6704, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38828499

ABSTRACT

Recently, the investigation of neuroprotective peptides has gained attention in addressing memory impairment and cognitive decline. Although the potential neuroprotective peptide Serine-Phenylalanine-Glycine-Aspartic acid-Isoleucine (SFGDI) has been identified from sea cucumber, the molecular mechanisms remain unclear. This study was conducted to explore the neuroprotection of SFGDI against 3-TYP-induced oxidative stress in BV2 cells. The results showed a retention rate of 76.70% during in vitro simulated gastrointestinal digestion and an absorption rate of 10.41% in a rat-everted gut sac model for SFGDI. Two hours following the administration of SFGDI via gavage in mice, a notable fluorescence was observed in the brain, indicating a potential neuroprotection of SFGDI through its interactions with nerve cells. By utilizing a model of oxidative stress injury induced by 3-TYP in BV2 cells, it was determined that pretreatment with SFGDI (50-200 µg mL-1) resulted in a dose-dependent reduction in the acetylated SOD level, leading to enhanced SOD activity and reduced levels of ROS and MDA. In addition, this pretreatment triggered an increase in unsaturated lipid levels, which helped maintain the intracellular lipid metabolism balance and preserve the mitochondrial function and glycolysis levels to regulate energy metabolism. The results of this study indicate that SFGDI demonstrates neuroprotective properties through its modulation of the Sirt3/SOD/ROS pathway, regulation of lipid metabolism, and enhancement of energy metabolism in BV2 cells. These findings suggest potential novel therapeutic approaches for addressing Sirt3-related memory deficits and neurodegenerative disorders.


Subject(s)
Energy Metabolism , Neuroprotective Agents , Oxidative Stress , Signal Transduction , Animals , Male , Mice , Rats , Cell Line , Energy Metabolism/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sirtuin 3/metabolism , Sirtuin 3/genetics , Superoxide Dismutase/metabolism
6.
Aging (Albany NY) ; 16(11): 9558-9568, 2024 05 31.
Article in English | MEDLINE | ID: mdl-38829778

ABSTRACT

Osteoarthritis (OA) is one of the most important causes of global disability, and dysfunction of chondrocytes is an important risk factor. The treatment of OA is still a challenge. Orexin-A is a hypothalamic peptide, and its effects in OA are unknown. In this study, we found that exposure to interleukin-1ß (IL-1ß) reduced the expression of orexin-2R, the receptor of orexin-A in TC-28a2 chondrocytes. Importantly, the senescence-associated ß-galactosidase (SA-ß-gal) staining assay demonstrated that orexin-A treatment ameliorates IL-1ß-induced cellular senescence. Importantly, the presence of IL-1ß significantly reduced the telomerase activity of TC-28a2 chondrocytes, which was rescued by orexin-A. We also found that orexin-A prevented IL-1ß-induced increase in the levels of Acetyl-p53 and the expression of p21. It is shown that orexin-A mitigates IL-1ß-induced reduction of sirtuin 3 (SIRT3). Silencing of SIRT3 abolished the protective effects of orexin-A against IL-1ß-induced cellular senescence. These results imply that orexin-A might serve as a promising therapeutic agent for OA.


Subject(s)
Cellular Senescence , Chondrocytes , Interleukin-1beta , Orexins , Cellular Senescence/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Orexins/pharmacology , Orexins/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Osteoarthritis/metabolism , Osteoarthritis/drug therapy , Humans , Sirtuin 3/metabolism , Sirtuin 3/genetics , Animals , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Orexin Receptors/metabolism , Orexin Receptors/genetics , Cell Line
7.
Acta Physiol (Oxf) ; 240(8): e14184, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38822624

ABSTRACT

AIM: Sepsis-induced myocardial injury (SIMI) may be associated with insufficient mitophagy in cardiomyocytes, but the exact mechanism involved remains unknown. Sirtuin 3 (Sirt3) is mainly found in the mitochondrial matrix and is involved in repairing mitochondrial function through means such as the activation of autophagy. Previously, we demonstrated that the annexin-A1 small peptide (ANXA1sp) can promote Sirt3 expression in mitochondria. In this study, we hypothesized that the activation of Sirt3 by ANXA1sp induces mitophagy, thereby providing a protective effect against SIMI in mice. METHODS: A mouse model of SIMI was established via cecal ligation and puncture. Intraperitoneal injections of ANXA1sp, 3TYP, and 3MA were administered prior to modeling. After successful modeling, IL-6, TNF-α, CK-MB, and CTn-I levels were measured; cardiac function was assessed using echocardiography; myocardial mitochondrial membrane potential, ROS, and ATP production were determined; myocardial mitochondrial ultrastructure was observed using transmission electron microscopy; and the expression levels of Sirt3 and autophagy-related proteins were detected using western blotting. RESULTS: ANXA1sp significantly reduced serum IL-6, TNF-α, CK-MB, and CTn-I levels; decreased myocardial ROS production; increased mitochondrial membrane potential and ATP synthesis; and improved myocardial mitochondrial ultrastructure in septic mice. Furthermore, ANXA1sp promoted Sirt3 expression and activated the AMPK-mTOR pathway to induce myocardial mitophagy. These protective effects of ANXA1sp were reversed upon treatment with the Sirt3 blocker, 3-TYP. CONCLUSION: ANXA1sp can reverse SIMI, and the underlying mechanism may be related to the activation of the AMPK-mTOR pathway following upregulation of Sirt3 by ANXA1sp, which, in turn, induces autophagy.


Subject(s)
Annexin A1 , Mitophagy , Sepsis , Sirtuin 3 , Animals , Sepsis/complications , Sepsis/metabolism , Mitophagy/drug effects , Sirtuin 3/metabolism , Sirtuin 3/genetics , Mice , Annexin A1/metabolism , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Disease Models, Animal , Autophagy , Peptides
8.
Clin Nutr ; 43(7): 1816-1831, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38870662

ABSTRACT

BACKGROUND: Optical atrophy 1 (OPA1), a protein accountable for mitochondrial fusion, facilitates the restoration of mitochondrial structure and function following cerebral ischemia/reperfusion (I/R) injury. The OPA1-conferred mitochondrial protection involves its expression and activity, which can be improved by SIRT3 in non-cerebral ischemia. Nevertheless, it remains obscure whether SIRT3 enhances the expression and activity of OPA1 after cerebral I/R injury. METHODS: Mature male Sprague Dawley rats were intracranially injected with adeno-associated viral-Sirtuin-3(AAV-SIRT3) and AAV-sh_OPA1, followed by a 90-min temporary blockage of the middle cerebral artery and subsequent restoration of blood flow. Cultured cortical neurons of rats were transfected with LV-SIRT3 or LV-sh_OPA1 before a 2-h oxygen-glucose deprivation and reoxygenation. The rats and neurons were subsequently treated with a selective OPA1 activity inhibitor (MYLS22). The interaction between SIRT3 and OPA1 was assessed by molecular dynamics simulation technology and co-immunoprecipitation. The expression, function, and specific protective mechanism of SIRT3 were examined by various analyses. RESULTS: SIRT3 interacted with OPA1 in the rat cerebral cortex before and after cerebral I/R. After cerebral I/R damage, SIRT3 upregulation increased the OPA1 expression, which enhanced deacetylation and OPA1 activity, thus alleviating cerebral infarct volume, neuronal apoptosis, oxidative pressure, and impairment in mitochondrial energy production; SIRT3 upregulation also improved neuromotor performance, repaired mitochondrial ultrastructure and membrane composition, and promoted the mitochondrial biogenesis. These neuroprotective effects were partly reversed by OPA1 expression interference and OPA1 activity inhibitor MYLS22. CONCLUSION: In rats, SIRT3 enhances the expression and activity of OPA1, facilitating the repair of mitochondrial structure and functional recovery following cerebral I/R injury. These findings highlight that regulating SIRT3 may be a promising therapeutic strategy for ischemic stroke.


Subject(s)
GTP Phosphohydrolases , Ischemic Stroke , Mitochondria , Rats, Sprague-Dawley , Sirtuin 3 , Animals , Male , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Sirtuin 3/metabolism , Sirtuin 3/genetics , Rats , Mitochondria/metabolism , Ischemic Stroke/metabolism , Neurons/metabolism , Reperfusion Injury/metabolism , Disease Models, Animal , Recovery of Function , Sirtuins
9.
Biomed Pharmacother ; 176: 116919, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38876053

ABSTRACT

Albumin has a variety of biological functions, such as immunomodulatory and antioxidant activity, which depends largely on its thiol activity. However, in clinical trials, the treatment of albumin by injection of commercial human serum albumin (HSA) did not achieve the desired results. Here, we constructed reduced modified albumin (SH-Alb) for in vivo and in vitro experiments to investigate the reasons why HSA did not achieve the expected effects. SH-Alb was found to delay the progression of liver fibrosis in mice by alleviating liver inflammation and oxidative stress. Although R-Alb also has some of the above roles, the effect of SH-Alb is more remarkable. Mechanism studies have shown that SH-Alb reduces the release of pro-inflammatory and pro-fibrotic cytokine through the mitogen-activated protein kinase (MAPK) signaling pathway. In addition, SH-Alb deacetylates SOD2, a key enzyme of mitochondrial reactive oxygen species (ROS) production, by promoting the expression of SIRT3, thereby reducing the accumulation of ROS. Finally, macrophages altered by R-Alb or SH-Alb can inhibit the activation of hepatic stellate cells and endothelial cells, further delaying the progression of liver fibrosis. These results indicate that SH-Alb can remodel the phenotype of macrophages, thereby affecting the intrahepatic microenvironment and delaying the process of liver fibrosis. It provides a good foundation for the application of albumin in clinical treatment.


Subject(s)
Liver Cirrhosis , Macrophages , Sirtuin 3 , Superoxide Dismutase , Animals , Humans , Male , Mice , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/pathology , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Macrophages/metabolism , Macrophages/drug effects , Mice, Inbred C57BL , Oxidative Stress/drug effects , Phenotype , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction , Sirtuin 3/metabolism , Superoxide Dismutase/metabolism
10.
Eur J Neurosci ; 60(1): 3677-3693, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38711280

ABSTRACT

Gastrodin, an anti-inflammatory herbal agent, is known to suppress microglia activation. Here, we investigated whether it would exert a similar effect in reactive astrocytes and whether it might act through the renin-angiotensin system (RAS) and sirtuin 3 (SIRT3). Angiotensinogen (ATO), angiotensin-converting enzyme (ACE), angiotensin II type 1 (AT1) and type 2 (AT2) receptor and SIRT3 expression was detected in TNC-1 astrocytes treated with BV-2 microglia conditioned medium (CM) with or without gastrodin and lipopolysaccharide (LPS) pre-treatment by RT-PCR, immunofluorescence and western blotting analysis. Expression of C3 (A1 astrocyte marker), S100A10 (A2 astrocyte marker), proinflammatory cytokines and neurotrophic factors was then evaluated. The results showed a significant increase of ATO, ACE, AT1, SIRT3, C3, proinflammatory cytokines and neurotrophic factors expression in TNC-1 astrocytes incubated in CM + LPS when compared with cells incubated in the CM, but AT2 and S100A10 expression was reduced. TNC-1 astrocytes responded vigorously to BV-2 CM treated with gastrodin + LPS as compared with the control. This was evident by the decreased expression of the abovementioned protein markers, except for AT2 and S100A10. Interestingly, SIRT3, IGF-1 and BDNF expression was enhanced, suggesting that gastrodin inhibited the expression of RAS and proinflammatory mediators but promoted the expression of neurotrophic factors. And gastrodin regulated the phenotypic changes of astrocytes through AT1. Additionally, azilsartan (a specific inhibitor of AT1) inhibited the expression of C3 and S100A10, which remained unaffected in gastrodin and azilsartan combination treatment. These findings provide evidence that gastrodin may have a therapeutic effect via regulating RAS-SIRT3.


Subject(s)
Astrocytes , Benzyl Alcohols , Glucosides , Microglia , Renin-Angiotensin System , Sirtuin 3 , Glucosides/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Microglia/drug effects , Microglia/metabolism , Animals , Benzyl Alcohols/pharmacology , Mice , Sirtuin 3/metabolism , Renin-Angiotensin System/drug effects , Lipopolysaccharides/pharmacology , Inflammation Mediators/metabolism , Cytokines/metabolism , Cell Line
11.
Arterioscler Thromb Vasc Biol ; 44(7): 1570-1583, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38813697

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) represents an important phenotype in heart failure with preserved ejection fraction (HFpEF). However, management of PH-HFpEF is challenging because mechanisms involved in the regulation of PH-HFpEF remain unclear. METHODS: We used a mass spectrometry-based comparative plasma proteomics approach as a sensitive and comprehensive hypothesis-generating discovery technique to profile proteins in patients with PH-HFpEF and control subjects. We then validated and investigated the role of one of the identified proteins using in vitro cell cultures, in vivo animal models, and independent cohort of human samples. RESULTS: Plasma proteomics identified high protein abundance levels of B2M (ß2-microglobulin) in patients with PH-HFpEF. Interestingly, both circulating and skeletal muscle levels of B2M were increased in mice with skeletal muscle SIRT3 (sirtuin-3) deficiency or high-fat diet-induced PH-HFpEF. Plasma and muscle biopsies from a validation cohort of PH-HFpEF patients were found to have increased B2M levels, which positively correlated with disease severity, especially pulmonary capillary wedge pressure and right atrial pressure at rest. Not only did the administration of exogenous B2M promote migration/proliferation in pulmonary arterial vascular endothelial cells but it also increased PCNA (proliferating cell nuclear antigen) expression and cell proliferation in pulmonary arterial vascular smooth muscle cells. Finally, B2m deletion improved glucose intolerance, reduced pulmonary vascular remodeling, lowered PH, and attenuated RV hypertrophy in mice with high-fat diet-induced PH-HFpEF. CONCLUSIONS: Patients with PH-HFpEF display higher circulating and skeletal muscle expression levels of B2M, the magnitude of which correlates with disease severity. Our findings also reveal a previously unknown pathogenic role of B2M in the regulation of pulmonary vascular proliferative remodeling and PH-HFpEF. These data suggest that circulating and skeletal muscle B2M can be promising targets for the management of PH-HFpEF.


Subject(s)
Disease Models, Animal , Heart Failure , Hypertension, Pulmonary , Proteomics , Stroke Volume , beta 2-Microglobulin , Adult , Aged , Animals , Humans , Male , Mice , Middle Aged , beta 2-Microglobulin/genetics , beta 2-Microglobulin/blood , beta 2-Microglobulin/metabolism , Biomarkers/blood , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Heart Failure/physiopathology , Heart Failure/metabolism , Heart Failure/blood , Heart Failure/genetics , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Proteomics/methods , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism , Vascular Remodeling , Ventricular Function, Left
12.
Mol Cell Endocrinol ; 591: 112268, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38735622

ABSTRACT

Menopause causes important bodily and metabolic changes, which favor the increased occurrence of cardiovascular diseases, obesity, diabetes, and osteoporosis. Resveratrol exerts proven effects on body metabolism, improving glucose and lipid homeostasis and reducing inflammation and oxidative stress in various organs and tissues. Accordingly, this study evaluates the effects of resveratrol supplementation on the expression of markers associated with thermogenesis in brown adipose tissue, and on the body, metabolic and hormonal parameters of female mice submitted to bilateral oophorectomy. Eighteen female mice were randomized into three groups: G1: control (CONTROL), G2: oophorectomy (OOF), and G3: oophorectomy + resveratrol (OOF + RSV); the animals were kept under treatment for twelve weeks, being fed a standard diet and treated with resveratrol via gavage. Body, biochemical, hormonal, and histological parameters were measured; in addition to the expression of markers associated with thermogenesis in brown adipose tissue. The results showed that animals supplemented with resveratrol showed reduced body weight and visceral adiposity, in addition to glucose, total cholesterol, and triglyceride levels; decreased serum FSH levels and increased estrogen levels were observed compared to the OOF group and mRNA expression of PRDM16, UCP1, and SIRT3 in brown adipose tissue. The findings of this study suggest the important role of resveratrol in terms of improving body, metabolic, and hormonal parameters, as well as modulating markers associated with thermogenesis in brown adipose tissue of female mice submitted to oophorectomy.


Subject(s)
Adipose Tissue, Brown , Dietary Supplements , Ovariectomy , Resveratrol , Thermogenesis , Uncoupling Protein 1 , Animals , Resveratrol/pharmacology , Resveratrol/administration & dosage , Female , Thermogenesis/drug effects , Thermogenesis/genetics , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Administration, Oral , Gene Expression Regulation/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism , Body Weight/drug effects , Hormones/blood
13.
Int J Biol Macromol ; 270(Pt 1): 132026, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704074

ABSTRACT

Multiple phenolic substances have been shown to promote SIRT3 expression, however, few studies have focused on the effects of these phenolics on SIRT3 enzyme activity. This study constructed a variety of reaction systems to elucidate the mechanisms by which different polyphenols affect SIRT3 enzyme activity. The results showed that acP53317-320 was the most suitable substrate among the five acetylated peptide substrates (Kcat/Km = 74.85 ± 1.86 M-1•s-1). All the phenolic compounds involved in the experiment inhibited the enzymatic activity of SIRT3, and the lowest IC50 among them was quercetin (0.12 ± 0.01 mM) and the highest was piceatannol (1.29 ± 0.08 mM). Their inhibition types were mainly competitive and mixed. In addition, piceatannol was found to be a natural SIRT3 agonist by enzyme kinetic analysis and validation of deacetylation efficiency. This study will provide a useful reference for polyphenol modulation of SIRT3 dosage, as well as the development and application of polyphenol-based SIRT3 activators and agonists.


Subject(s)
Polyphenols , Sirtuin 3 , Sirtuin 3/metabolism , Polyphenols/pharmacology , Polyphenols/chemistry , Acetylation , Humans , Kinetics , Stilbenes/pharmacology , Stilbenes/chemistry
14.
Signal Transduct Target Ther ; 9(1): 133, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38744811

ABSTRACT

Sirtuin 3 (SIRT3) is well known as a conserved nicotinamide adenine dinucleotide+ (NAD+)-dependent deacetylase located in the mitochondria that may regulate oxidative stress, catabolism and ATP production. Accumulating evidence has recently revealed that SIRT3 plays its critical roles in cardiac fibrosis, myocardial fibrosis and even heart failure (HF), through its deacetylation modifications. Accordingly, discovery of SIRT3 activators and elucidating their underlying mechanisms of HF should be urgently needed. Herein, we identified a new small-molecule activator of SIRT3 (named 2-APQC) by the structure-based drug designing strategy. 2-APQC was shown to alleviate isoproterenol (ISO)-induced cardiac hypertrophy and myocardial fibrosis in vitro and in vivo rat models. Importantly, in SIRT3 knockout mice, 2-APQC could not relieve HF, suggesting that 2-APQC is dependent on SIRT3 for its protective role. Mechanically, 2-APQC was found to inhibit the mammalian target of rapamycin (mTOR)-p70 ribosomal protein S6 kinase (p70S6K), c-jun N-terminal kinase (JNK) and transforming growth factor-ß (TGF-ß)/ small mother against decapentaplegic 3 (Smad3) pathways to improve ISO-induced cardiac hypertrophy and myocardial fibrosis. Based upon RNA-seq analyses, we demonstrated that SIRT3-pyrroline-5-carboxylate reductase 1 (PYCR1) axis was closely assoiated with HF. By activating PYCR1, 2-APQC was shown to enhance mitochondrial proline metabolism, inhibited reactive oxygen species (ROS)-p38 mitogen activated protein kinase (p38MAPK) pathway and thereby protecting against ISO-induced mitochondrialoxidative damage. Moreover, activation of SIRT3 by 2-APQC could facilitate AMP-activated protein kinase (AMPK)-Parkin axis to inhibit ISO-induced necrosis. Together, our results demonstrate that 2-APQC is a targeted SIRT3 activator that alleviates myocardial hypertrophy and fibrosis by regulating mitochondrial homeostasis, which may provide a new clue on exploiting a promising drug candidate for the future HF therapeutics.


Subject(s)
Cardiomegaly , Fibrosis , Sirtuin 3 , Animals , Humans , Male , Mice , Rats , Cardiomegaly/genetics , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Fibrosis/genetics , Homeostasis/drug effects , Isoproterenol , Mice, Knockout , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/pathology , Mitochondria/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/pathology , Myocardium/metabolism , Sirtuin 3/drug effects , Sirtuin 3/metabolism
15.
Sci Adv ; 10(20): eadj5942, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758779

ABSTRACT

Acetyl-CoA synthetase short-chain family member 1 (ACSS1) uses acetate to generate mitochondrial acetyl-CoA and is regulated by deacetylation by sirtuin 3. We generated an ACSS1-acetylation (Ac) mimic mouse, where lysine-635 was mutated to glutamine (K635Q). Male Acss1K635Q/K635Q mice were smaller with higher metabolic rate and blood acetate and decreased liver/serum ATP and lactate levels. After a 48-hour fast, Acss1K635Q/K635Q mice presented hypothermia and liver aberrations, including enlargement, discoloration, lipid droplet accumulation, and microsteatosis, consistent with nonalcoholic fatty liver disease (NAFLD). RNA sequencing analysis suggested dysregulation of fatty acid metabolism, cellular senescence, and hepatic steatosis networks, consistent with NAFLD. Fasted Acss1K635Q/K635Q mouse livers showed increased fatty acid synthase (FASN) and stearoyl-CoA desaturase 1 (SCD1), both associated with NAFLD, and increased carbohydrate response element-binding protein binding to Fasn and Scd1 enhancer regions. Last, liver lipidomics showed elevated ceramide, lysophosphatidylethanolamine, and lysophosphatidylcholine, all associated with NAFLD. Thus, we propose that ACSS1-K635-Ac dysregulation leads to aberrant lipid metabolism, cellular senescence, and NAFLD.


Subject(s)
Cellular Senescence , Mitochondria , Non-alcoholic Fatty Liver Disease , Stearoyl-CoA Desaturase , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Mice , Cellular Senescence/genetics , Acetylation , Mitochondria/metabolism , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Male , Acetate-CoA Ligase/metabolism , Acetate-CoA Ligase/genetics , Gene Knock-In Techniques , Liver/metabolism , Liver/pathology , Lipid Metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Disease Models, Animal , Coenzyme A Ligases , Fatty Acid Synthase, Type I
16.
J Mol Cell Cardiol ; 192: 36-47, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734062

ABSTRACT

AIMS: Ferroptosis is a form of iron-regulated cell death implicated in ischemic heart disease. Our previous study revealed that Sirtuin 3 (SIRT3) is associated with ferroptosis and cardiac fibrosis. In this study, we tested whether the knockout of SIRT3 in cardiomyocytes (SIRT3cKO) promotes mitochondrial ferroptosis and whether the blockade of ferroptosis would ameliorate mitochondrial dysfunction. METHODS AND RESULTS: Mitochondrial and cytosolic fractions were isolated from the ventricles of mice. Cytosolic and mitochondrial ferroptosis were analyzed by comparison to SIRT3loxp mice. An echocardiography study showed that SIRT3cKO mice developed heart failure as evidenced by a reduction of EF% and FS% compared to SIRT3loxp mice. Comparison of mitochondrial and cytosolic fractions of SIRT3cKO and SIRT3loxp mice revealed that, upon loss of SIRT3, mitochondrial, but not cytosolic, total lysine acetylation was significantly increased. Similarly, acetylated p53 was significantly upregulated only in the mitochondria. These data demonstrate that SIRT3 is the primary mitochondrial deacetylase. Most importantly, loss of SIRT3 resulted in significant reductions of frataxin, aconitase, and glutathione peroxidase 4 (GPX4) in the mitochondria. This was accompanied by a significant increase in levels of mitochondrial 4-hydroxynonenal. Treatment of SIRT3cKO mice with the ferroptosis inhibitor ferrostatin-1 (Fer-1) for 14 days significantly improved preexisting heart failure. Mechanistically, Fer-1 treatment significantly increased GPX4 and aconitase expression/activity, increased mitochondrial iron­sulfur clusters, and improved mitochondrial membrane potential and Complex IV activity. CONCLUSIONS: Inhibition of ferroptosis ameliorated cardiac dysfunction by specifically targeting mitochondrial aconitase and iron­sulfur clusters. Blockade of mitochondrial ferroptosis may be a novel therapeutic target for mitochondrial cardiomyopathies.


Subject(s)
Aconitate Hydratase , Ferroptosis , Mice, Knockout , Myocytes, Cardiac , Phenylenediamines , Sirtuin 3 , Animals , Sirtuin 3/metabolism , Sirtuin 3/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Aconitate Hydratase/metabolism , Ferroptosis/drug effects , Mice , Acetylation , Phenylenediamines/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron/metabolism , Frataxin , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Iron-Binding Proteins/metabolism , Iron-Binding Proteins/genetics , Heart Failure/metabolism , Heart Failure/genetics , Cytosol/metabolism , Cyclohexylamines
17.
Biomed Pharmacother ; 175: 116689, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703508

ABSTRACT

Ischemic heart disease invariably leads to devastating damage to human health. Nicotinamide ribose (NR), as one of the precursors of NAD+ synthesis, has been discovered to exert a protective role in various neurological and cardiovascular disorders. Our findings demonstrated that pretreatment with 200 mg/kg NR for 3 h significantly reduced myocardial infarct area, decreased levels of CK-MB and LDH in serum, and improved cardiac function in the rats during myocardial ischemia-reperfusion (I/R) injury. Meanwhile, 0.5 mM NR also effectively increased the viability and decreased the LDH release of H9c2 cells during OGD/R. We had provided evidence that NR pretreatment could decrease mitochondrial reactive oxygen species (mtROS) production and MDA content, and enhance SOD activity, thereby mitigating mitochondrial damage and inhibiting apoptosis during myocardial I/R injury. Further investigations revealed that NR increased NAD+ content and upregulated SIRT3 protein expression in myocardium. Through using of SIRT3 small interfering RNA and the SIRT3 deacetylase activity inhibitor 3-TYP, we had confirmed that the cardioprotective effect of NR on cardiomyocytes was largely dependent on the inhibition of mitochondrial oxidative stress via SIRT3-SOD2 axis. Overall, our study suggested that exogenous supplementation with NR mitigated mitochondrial damage and inhibited apoptosis during myocardial I/R injury by reducing mitochondrial oxidative stress via SIRT3-SOD2-mtROS pathway.


Subject(s)
Apoptosis , Myocardial Reperfusion Injury , Niacinamide , Oxidative Stress , Pyridinium Compounds , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 3 , Superoxide Dismutase , Animals , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Sirtuin 3/metabolism , Signal Transduction/drug effects , Male , Niacinamide/pharmacology , Niacinamide/analogs & derivatives , Superoxide Dismutase/metabolism , Rats , Apoptosis/drug effects , Oxidative Stress/drug effects , Pyridinium Compounds/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Cell Line , Cardiotonic Agents/pharmacology , Sirtuins
18.
Redox Biol ; 73: 103179, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733909

ABSTRACT

Increasing evidences demonstrate that environmental stressors are important inducers of acute kidney injury (AKI). This study aimed to investigate the impact of exposure to Cd, an environmental stressor, on renal cell ferroptosis. Transcriptomics analyses showed that arachidonic acid (ARA) metabolic pathway was disrupted in Cd-exposed mouse kidneys. Targeted metabolomics showed that renal oxidized ARA metabolites were increased in Cd-exposed mice. Renal 4-HNE, MDA, and ACSL4, were upregulated in Cd-exposed mouse kidneys. Consistent with animal experiments, the in vitro experiments showed that mitochondrial oxidized lipids were elevated in Cd-exposed HK-2 cells. Ultrastructure showed mitochondrial membrane rupture in Cd-exposed mouse kidneys. Mitochondrial cristae were accordingly reduced in Cd-exposed mouse kidneys. Mitochondrial SIRT3, an NAD+-dependent deacetylase that regulates mitochondrial protein stability, was reduced in Cd-exposed mouse kidneys. Subsequently, mitochondrial GPX4 acetylation was elevated and mitochondrial GPX4 protein was reduced in Cd-exposed mouse kidneys. Interestingly, Cd-induced mitochondrial GPX4 acetylation and renal cell ferroptosis were exacerbated in Sirt3-/- mice. Conversely, Cd-induced mitochondrial oxidized lipids were attenuated in nicotinamide mononucleotide (NMN)-pretreated HK-2 cells. Moreover, Cd-evoked mitochondrial GPX4 acetylation and renal cell ferroptosis were alleviated in NMN-pretreated mouse kidneys. These results suggest that mitochondrial GPX4 acetylation, probably caused by SIRT3 downregulation, is involved in Cd-evoked renal cell ferroptosis.


Subject(s)
Cadmium , Ferroptosis , Mitochondria , Phospholipid Hydroperoxide Glutathione Peroxidase , Sirtuin 3 , Animals , Ferroptosis/drug effects , Mice , Cadmium/toxicity , Cadmium/adverse effects , Sirtuin 3/metabolism , Sirtuin 3/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Acetylation , Humans , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Cell Line , Male , Mice, Knockout , Coenzyme A Ligases
19.
Chem Res Toxicol ; 37(6): 1062-1069, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38815162

ABSTRACT

Multiple myeloma is a hematological cancer that can be treated but remains incurable. With the advancement of science and technology, more drugs have been developed for myeloma chemotherapy that greatly improve the quality of life of patients. However, relapse remains a serious problem puzzling patients and doctors. Thus, developing more highly active and specific inhibitors is urgent for myeloma-targeted therapy. In this study, we identified the SIRT3 inhibitor 3-TYP (3-(1H-1,2,3-triazol-4-yl) pyridine) after screening a histone modification compound library, which showed high cytotoxicity and induced DNA damage in myeloma cells. Furthermore, the inhibitory effect of 3-TYP in our xenograft tumor studies also confirmed that compound 3-TYP could inhibit primary myeloma growth by reducing c-Myc protein stability by decreasing c-Myc Ser62 phosphorylation levels. Taken together, the results of our study identified 3-TYP as a novel c-Myc inhibitor, which could be a potential chemotherapeutic agent to target multiple myeloma.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Multiple Myeloma , Proto-Oncogene Proteins c-myc , Sirtuin 3 , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Humans , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Sirtuin 3/antagonists & inhibitors , Sirtuin 3/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Mice , Pyridines/pharmacology , Pyridines/chemistry , Triazoles/pharmacology , Triazoles/chemistry , Cell Line, Tumor , Molecular Structure , Drug Screening Assays, Antitumor , Protein Stability/drug effects , Mice, Nude
20.
J Pharm Biomed Anal ; 246: 116206, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38733762

ABSTRACT

Ischemic stroke, accounting for 80 % of all strokes, is a major cause of morbidity and mortality worldwide. However, effective and safe pharmacotherapy options for ischemic injury are limited. This study investigated the therapeutic effects of wogonoside, a compound derived from Radix Scutellariae, on ischemia/reperfusion (I/R) injury. The results showed that wogonoside treatment had significant therapeutic effects in rats with middle cerebral artery occlusion. It effectively reduced mortality rates, neurological deficits, cerebral infarct size, and brain water content. In an in vitro model using PC12 cells, wogonoside activated the Nrf2/Sirt3 signaling pathway. This activation contributed to the attenuation of oxidative damage and inflammation. Metabolomics analysis revealed increased levels of γ-aminobutyric acid (GABA) and glutathione in response to wogonoside treatment, suggesting their potential as therapeutic biomarkers for ischemic stroke. Additionally, wogonoside restored perturbed energy metabolism, including the tricarboxylic acid cycle. Wogonoside has the potential to ameliorate cerebral ischemic injury by targeting GABA-related amino acid metabolism, energy metabolism, and glutathione metabolism, maintaining redox homeostasis, and attenuating oxidative stress. These findings provide valuable insights into the protective mechanisms of wogonoside in cerebral I/R injury and highlight the promising therapeutic approach of wogonoside in the treatment of ischemic stroke.


Subject(s)
Ischemic Stroke , Metabolomics , NF-E2-Related Factor 2 , Oxidative Stress , Rats, Sprague-Dawley , Reperfusion Injury , Signal Transduction , Sirtuin 3 , Tandem Mass Spectrometry , Animals , Rats , NF-E2-Related Factor 2/metabolism , Metabolomics/methods , Signal Transduction/drug effects , PC12 Cells , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Male , Tandem Mass Spectrometry/methods , Oxidative Stress/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Sirtuin 3/metabolism , Flavanones/pharmacology , Flavanones/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Chromatography, High Pressure Liquid/methods , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Glucosides/pharmacology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Glutathione/metabolism , Disease Models, Animal , Sirtuins
SELECTION OF CITATIONS
SEARCH DETAIL
...