Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.741
Filter
1.
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
2.
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
3.
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
4.
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 , Mice, Inbred C57BL , Phenotype , Reactive Oxygen Species , Sirtuin 3 , Superoxide Dismutase , Animals , Sirtuin 3/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Mice , Macrophages/metabolism , Macrophages/drug effects , Male , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Humans , Oxidative Stress/drug effects , Signal Transduction , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/pathology , Liver/drug effects , Liver/metabolism , RAW 264.7 Cells
5.
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
6.
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 , Reactive Oxygen Species , Sirtuin 3 , Superoxide Dismutase , Animals , Oxidative Stress/drug effects , Sirtuin 3/metabolism , Sirtuin 3/genetics , Neuroprotective Agents/pharmacology , Mice , Energy Metabolism/drug effects , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Male , Cell Line , Rats, Sprague-Dawley , Signal Transduction/drug effects
7.
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
8.
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
9.
Nat Commun ; 15(1): 4383, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782909

ABSTRACT

Macrophages (Mφ) autophagy is a pivotal contributor to inflammation-related diseases. However, the mechanistic details of its direct role in acute kidney injury (AKI) were unclear. Here, we show that Mφ promote AKI progression via crosstalk with tubular epithelial cells (TECs), and autophagy of Mφ was activated and then inhibited in cisplatin-induced AKI mice. Mφ-specific depletion of ATG7 (Atg7Δmye) aggravated kidney injury in AKI mice, which was associated with tubulointerstitial inflammation. Moreover, Mφ-derived exosomes from Atg7Δmye mice impaired TEC mitochondria in vitro, which may be attributable to miR-195a-5p enrichment in exosomes and its interaction with SIRT3 in TECs. Consistently, either miR-195a-5p inhibition or SIRT3 overexpression improved mitochondrial bioenergetics and renal function in vivo. Finally, adoptive transfer of Mφ from AKI mice to Mφ-depleted mice promotes the kidney injury response to cisplatin, which is alleviated when Mφ autophagy is activated with trehalose. We conclude that exosomal miR-195a-5p mediate the communication between autophagy-deficient Mφ and TECs, leading to impaired mitochondrial biogenetic in TECs and subsequent exacerbation of kidney injury in AKI mice via miR-195a-5p-SIRT3 axis.


Subject(s)
Acute Kidney Injury , Autophagy , Cisplatin , Macrophages , MicroRNAs , Mitochondria , Sirtuin 3 , Animals , Humans , Male , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Autophagy/drug effects , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Cisplatin/pharmacology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Exosomes/metabolism , Kidney/pathology , Kidney/metabolism , Kidney Tubules/pathology , Kidney Tubules/metabolism , Macrophages/metabolism , Macrophages/drug effects , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Sirtuin 3/metabolism , Sirtuin 3/genetics , Trehalose/pharmacology
10.
Cell Death Dis ; 15(5): 364, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802337

ABSTRACT

Mitochondrial dysfunction and oxidative stress are important mechanisms for secondary injury after traumatic brain injury (TBI), which result in progressive pathophysiological exacerbation. Although the Fibronectin type III domain-containing 5 (FNDC5) was reported to repress oxidative stress by retaining mitochondrial biogenesis and dynamics, its possible role in the secondary injury after TBI remain obscure. In present study, we observed that the level of serum irisin (the cleavage product of FNDC5) significantly correlated with the neurological outcomes of TBI patients. Knockout of FNDC5 increased the lesion volume and exacerbated apoptosis and neurological deficits after TBI in mice, while FNDC5 overexpression yielded a neuroprotective effect. Moreover, FNDC5 deficiency disrupted mitochondrial dynamics and function. Activation of Sirtuin 3 (SIRT3) alleviated FNDC5 deficiency-induced disruption of mitochondrial dynamics and bioenergetics. In neuron-specific SIRT3 knockout mice, FNDC5 failed to attenuate TBI-induced mitochondrial damage and brain injuries. Mechanically, FNDC5 deficiency led to reduced SIRT3 expression via enhanced ubiquitin degradation of transcription factor Nuclear factor erythroid 2-related factor 2 (NRF2), which contributed to the hyperacetylation and inactivation of key regulatory proteins of mitochondrial dynamics and function, including OPA1 and SOD2. Finally, engineered RVG29-conjugated nanoparticles were generated to selectively and efficiently deliver irisin to the brain of mice, which yielded a satisfactory curative effect against TBI. In conclusion, FNDC5/irisin exerts a protective role against acute brain injury by promoting SIRT3-dependent mitochondrial quality control and thus represents a potential target for neuroprotection after TBI.


Subject(s)
Apoptosis , Brain Injuries, Traumatic , Fibronectins , Mice, Knockout , Mitochondria , Neurons , Oxidative Stress , Sirtuin 3 , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/genetics , Sirtuin 3/metabolism , Sirtuin 3/genetics , Fibronectins/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Mice , Humans , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Mitochondrial Dynamics
11.
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
13.
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
14.
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
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.
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
17.
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
18.
Plant Foods Hum Nutr ; 79(2): 417-424, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710924

ABSTRACT

Hepatocellular carcinoma (HCC) is an alarming epidemiological clinical problem worldwide. Pharmacological approaches currently available do not provide adequate responses due to poor effectiveness, high toxicity, and serious side effects. Our previous studies have shown that the wild edible plant Crithmum maritimum L. inhibits the growth of liver cancer cells and promotes liver cell differentiation by reducing lactic acid fermentation (Warburg effect). Here, we aimed to further characterise the effects of C. maritimum on lipid metabolism and markers of cellular metabolic health, such as AMP-activated protein kinase (AMPK), Sirtuin 1 (SIRT1), and Sirtuin 3 (SIRT3), as well as the insulin signalling pathway. To better mimic the biological spectrum of HCC, we employed four HCC cell lines with different degrees of tumorigenicity and lactic acid fermentation/Warburg phenotype. Lipid accumulation was assessed by Oil Red O (ORO) staining, while gene expression was measured by real-time quantitative PCR (RT-qPCR). The activation of AMPK and insulin signalling pathways was determined by Western blotting. Results indicate that C. maritimum prevents lipid accumulation, downregulates lipid and cholesterol biosynthesis, and modulates markers of metabolic health, such as AMPK, SIRT1 and SIRT3. This modulation is different amongst HCC cell lines, revealing an important functional versatility of C. maritimum. Taken together, our findings corroborate the importance of C. maritimum as a valuable nutraceutical, reinforcing its role for the improvement of metabolic health.


Subject(s)
AMP-Activated Protein Kinases , Carcinoma, Hepatocellular , Lipid Metabolism , Liver Neoplasms , Plant Extracts , Sirtuin 1 , Humans , Plant Extracts/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Lipid Metabolism/drug effects , Sirtuin 1/metabolism , Sirtuin 1/genetics , Cell Line, Tumor , AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Signal Transduction/drug effects , Homeostasis/drug effects , Insulin/metabolism , Phenotype , Cholesterol/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...