Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.241
Filter
1.
Gen Physiol Biophys ; 43(5): 435-443, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140682

ABSTRACT

MicroRNAs (miRNAs) are essential modulators of gene expression and are associated with various pathological processes, including spinal cord injury (SCI). This investigation aimed to elucidate miR-10a activity in SCI and its potential interaction with sirtuin 1 (SIRT1). The SCI rat model was established to assess hind limb movement, measure levels of miR-10a, SIRT1, neuronal survival, and inflammatory factors. An in-vitro SCI cell model was also developed to evaluate cell viability and inflammatory factor levels. The interaction between miR10a and SIRT1 was verified. Upregulated miR-10a and downregulated SIRT1 expression were found in the tissues of SCI rats. miR-10a knockdown in SCI rats enhanced the recovery of motor function, increased neuronal survival, and reduced the levels of inflammatory cytokines. Luciferase reporter assays confirmed that miR-10a targeted SIRT1 directly. In PC12 cells, downregulation of miR-10a increased SIRT1 expression, enhanced cell viability, and reduced inflammatory factor levels after LPS stimulation. Conversely, SIRT1 knockdown inhibited the protective effects of downregulated miR-10a on cell viability and inflammatory responses. The results suggest that miR-10a downregulation protects against SCI by upregulating SIRT1 expression, improving functional recovery, and reducing inflammation. Targeting the miR-10a/SIRT1 axis is a promising strategy for SCI treatment.


Subject(s)
COVID-19 , MicroRNAs , Sirtuin 1 , Animals , Rats , Sirtuin 1/metabolism , Sirtuin 1/genetics , PC12 Cells , MicroRNAs/metabolism , MicroRNAs/genetics , COVID-19/metabolism , Male , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/genetics , Rats, Sprague-Dawley , Cell Survival
2.
Clin Exp Pharmacol Physiol ; 51(9): e13912, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103220

ABSTRACT

Sevoflurane (Sev) is a commonly used inhalation anaesthetic that has been shown to cause hippocampus dysfunction through multiple underlying molecular processes, including mitochondrial malfunction, oxidative stress and inflammation. Dihydromyricetin (DHM) is a 2,3-dihydroflavonoid with various biological properties, such as anti-inflammation and anti-oxidative stress. The purpose of this study was to investigate the effect of DHM on Sev-induced neuronal dysfunction. HT22 cells were incubated with 10, 20 and 30 µM of DHM for 24 h, and then stimulated with 4% Sev for 6 h. The effects and mechanism of DHM on inflammation, oxidative stress and mitochondrial dysfunction were explored in Sev-induced HT22 cells by Cell Counting Kit-8, flow cytometry, enzyme-linked immunosorbent assay, reverse transcription-quantitative polymerase chain reaction, colorimetric detections, detection of the level of reactive oxygen species (ROS), mitochondrial ROS and mitochondrial membrane potential (MMP), immunofluorescence and western blotting. Our results showed that DHM increased Sev-induced cell viability of HT22 cells. Pretreatment with DHM attenuated apoptosis, inflammation, oxidative stress and mitochondrial dysfunction in Sev-elicited HT22 cells by remedying the abnormality of the indicators involved in these progresses, including apoptosis rate, the cleaved-caspase 3 expression, as well as the level of tumour necrosis factor α, interleukin (IL)-1ß, IL-6, malondialdehyde, superoxide dismutase, catalase, ROS, mitochondrial ROS and MMP. Mechanically, pretreatment with DHM restored the Sev-induced the expression of SIRT1/FOXO3a pathway in HT22 cells. Blocking of SIRT1 counteracted the mitigatory effect of DHM on apoptosis, inflammation, oxidative stress and mitochondrial dysfunction in Sev-elicited HT22 cells. Collectively, pretreatment with DHM improved inflammation, oxidative stress and mitochondrial dysfunction via SIRT1/FOXO3a pathway in Sev-induced HT22 cells.


Subject(s)
Apoptosis , Flavonols , Hippocampus , Mitochondria , Oxidative Stress , Sevoflurane , Flavonols/pharmacology , Animals , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/cytology , Hippocampus/pathology , Cell Line , Sevoflurane/pharmacology , Oxidative Stress/drug effects , Apoptosis/drug effects , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Sirtuin 1/metabolism , Neuroprotective Agents/pharmacology
3.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125657

ABSTRACT

Well-controlled type 1 diabetes (T1DM) is characterized by inflammation and endothelial dysfunction, thus constituting a suitable model of subclinical cardiovascular disease (CVD). miR-199b-5p overexpression in murine CVD has shown proatherosclerotic effects. We hypothesized that miR-199b-5p would be overexpressed in subclinical CVD yet downregulated following metformin therapy. Inflammatory and vascular markers were measured in 29 individuals with T1DM and 20 matched healthy controls (HCs). miR-199b-5p expression in CFU-Hill's colonies was analyzed from each study group, and correlations with inflammatory/vascular health indices were evaluated. Significant upregulation of miR-199b-5p was observed in T1DM, which was significantly downregulated by metformin. miR-199b-5p correlated positively with vascular endothelial growth factor-D and c-reactive protein (CRP: nonsignificant). ROC analysis determined miR-199b-5p to define subclinical CVD by discriminating between HCs and T1DM individuals. ROC analyses of HbA1c and CRP showed that the upregulation of miR-199b-5p in T1DM individuals defined subclinical CVD at HbA1c > 44.25 mmol and CRP > 4.35 × 106 pg/mL. Ingenuity pathway analysis predicted miR-199b-5p to inhibit the target genes SIRT1, ETS1, and JAG1. Metformin was predicted to downregulate miR-199b-5p via NFATC2 and STAT3 and reverse its downstream effects. This study validated the antiangiogenic properties of miR-199b-5p and substantiated miR-199b-5p overexpression as a biomarker of subclinical CVD. The downregulation of miR-199b-5p by metformin confirmed its cardio-protective effect.


Subject(s)
Cardiovascular Diseases , Metformin , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Humans , Male , Female , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Adult , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Inflammation/genetics , Inflammation/drug therapy , Inflammation/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Middle Aged , Gene Expression Regulation/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Biomarkers , Case-Control Studies
4.
Int J Biol Sci ; 20(10): 4077-4097, 2024.
Article in English | MEDLINE | ID: mdl-39113710

ABSTRACT

Triptolide (TP), known for its effectiveness in treating various rheumatoid diseases, is also associated with significant hepatotoxicity risks. This study explored Catalpol (CAT), an iridoid glycoside with antioxidative and anti-inflammatory effects, as a potential defense against TP-induced liver damage. In vivo and in vitro models of liver injury were established using TP in combination with different concentrations of CAT. Metabolomics analyses were conducted to assess energy metabolism in mouse livers. Additionally, a Seahorse XF Analyzer was employed to measure glycolysis rate, mitochondrial respiratory functionality, and real-time ATP generation rate in AML12 cells. The study also examined the expression of proteins related to glycogenolysis and gluconeogenesis. Using both in vitro SIRT1 knockout/overexpression and in vivo liver-specific SIRT1 knockout models, we confirmed SIRT1 as a mechanism of action for CAT. Our findings revealed that CAT could alleviate TP-induced liver injury by activating SIRT1, which inhibited lysine acetylation of hypoxia-inducible factor-1α (HIF-1α), thereby restoring the balance between glycolysis and oxidative phosphorylation. This action improved mitochondrial dysfunction and reduced glucose metabolism disorder and oxidative stress caused by TP. Taken together, these insights unveil a hitherto undocumented mechanism by which CAT ameliorates TP-induced liver injury, positioning it as a potential therapeutic agent for managing TP-induced hepatotoxicity.


Subject(s)
Diterpenes , Epoxy Compounds , Glucose , Hypoxia-Inducible Factor 1, alpha Subunit , Iridoid Glucosides , Liver , Oxidative Stress , Phenanthrenes , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Diterpenes/pharmacology , Diterpenes/therapeutic use , Oxidative Stress/drug effects , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Phenanthrenes/pharmacology , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/metabolism , Liver/drug effects , Glucose/metabolism , Male , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Mice, Inbred C57BL
5.
Nat Commun ; 15(1): 7000, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143095

ABSTRACT

Mutations in the nuclear envelope (NE) protein lamin A/C (encoded by LMNA), cause a severe form of dilated cardiomyopathy (DCM) with early-onset life-threatening arrhythmias. However, molecular mechanisms underlying increased arrhythmogenesis in LMNA-related DCM (LMNA-DCM) remain largely unknown. Here we show that a frameshift mutation in LMNA causes abnormal Ca2+ handling, arrhythmias and disformed NE in LMNA-DCM patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). Mechanistically, lamin A interacts with sirtuin 1 (SIRT1) where mutant lamin A/C accelerates degradation of SIRT1, leading to mitochondrial dysfunction and oxidative stress. Elevated reactive oxygen species (ROS) then activates the Ca2+/calmodulin-dependent protein kinase II (CaMKII)-ryanodine receptor 2 (RYR2) pathway and aggravates the accumulation of SUN1 in mutant iPSC-CMs, contributing to arrhythmias and NE deformation, respectively. Taken together, the lamin A/C deficiency-mediated ROS disorder is revealed as central to LMNA-DCM development. Manipulation of impaired SIRT1 activity and excessive oxidative stress is a potential future therapeutic strategy for LMNA-DCM.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Lamin Type A , Myocytes, Cardiac , Oxidative Stress , Reactive Oxygen Species , Sirtuin 1 , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Lamin Type A/metabolism , Lamin Type A/genetics , Induced Pluripotent Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Humans , Sirtuin 1/metabolism , Sirtuin 1/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Frameshift Mutation , Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Nuclear Envelope/metabolism , Mitochondria/metabolism , Male , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
6.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126109

ABSTRACT

Sodium-glucose cotransporter-2 inhibitors (SGLT2i) have a variety of cardiovascular and renoprotective effects and have been developed as novel agents for the treatment of heart failure. However, the beneficial mechanisms of SGLT2i on cardiac tissue need to be investigated further. In this study, we established a mouse model of acute myocardial infarction (AMI) using coronary artery constriction surgery and investigated the role of dapagliflozin (DAPA) in protecting cardiomyocytes from hypoxic injury induced by AMI. In vitro experiments were done using hypoxic cultured H9c2 ventricular cells to verify this potential mechanism. Expression of the SIRT family and related genes and proteins was verified by qPCR, Western blotting and immunofluorescence staining, and the intrinsic potential mechanism of cardiomyocyte death due to AMI and hypoxia was comprehensively investigated by RNA sequencing. The RNA sequencing results of cardiomyocytes from AMI mice showed that the SIRT family may be mainly involved in the mechanisms of hypoxia-induced cardiomyocyte death. In vitro hypoxia-induced ventricular cells showed the role of dapagliflozin in conferring resistance to hypoxic injury in cardiomyocytes. It showed that SIRT1/3/6 were downregulated in H9c2 cells in a hypoxic environment, and the addition of dapagliflozin significantly increased the gene and protein expression of SIRT1, 3 and 6. We then verified the underlying mechanisms induced by dapagliflozin in hypoxic cardiomyocytes using RNA-seq, and found that dapagliflozin upregulated the hypoxia-induced gene downregulation, which includes ESRRA, EPAS1, AGTRAP, etc., that associated with SIRTs-related and apoptosis-related signaling to prevent H9c2 cell death. This study provides laboratory data for SGLT2i dapagliflozin treatment of AMI and confirms that dapagliflozin can be used to treat hypoxia-induced cellular necrosis in cardiomyocytes, in which SIRT1 and SIRT3 may play an important role. This opens up further opportunities for SGLT2i in the treatment of heart disease.


Subject(s)
Benzhydryl Compounds , Glucosides , Myocardial Infarction , Myocytes, Cardiac , Signal Transduction , Sirtuin 1 , Sodium-Glucose Transporter 2 Inhibitors , Glucosides/pharmacology , Glucosides/therapeutic use , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Benzhydryl Compounds/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Sirtuin 1/metabolism , Sirtuin 1/genetics , Signal Transduction/drug effects , Male , Sirtuin 3/metabolism , Sirtuin 3/genetics , Sirtuins/metabolism , Sirtuins/genetics , Cell Line , Mice, Inbred C57BL , Disease Models, Animal , Cell Hypoxia/drug effects , Rats , Apoptosis/drug effects
7.
PLoS One ; 19(8): e0309301, 2024.
Article in English | MEDLINE | ID: mdl-39172877

ABSTRACT

Tendinopathy is a common age-related disease which causes significant morbidity for both human athletes and performance horses. In the latter, the superficial digital flexor tendon is an excellent model for human tendinopathies because it is a functional homologue of the human Achilles tendon and a primary site of injuries with strong similarities to the human disease. Corticosteroids have been previously used clinically to treat tendinopathic inflammation, but they upregulate the p53-p21 axis with concomitant reductions in cell proliferation and collagen synthesis in human tenocytes. This phenotype is consistent with the induction of cellular senescence in vitro and in vivo and probably represents an important clinical barrier to their effective use. Because of the many differences in senescence mechanisms between species, this study aimed to evaluate these mechanisms after corticosteroid treatment in equine tenocytes. Exposure to clinically reflective levels of dexamethasone for 48 hours drove equine tenocytes into steroid induced senescence (SIS). This was characterised by permanent growth arrest and upregulation of p53, the cyclin dependent kinase inhibitors p21waf and p16ink4a as well as the matrix degrading enzymes MMP1, MMP2 and MMP13. SIS also induced a distinctive equine senescence associated secretory phenotype (eSASP) characterised by enhanced secretion of IL-8 and MCP-1. Preincubation with resveratrol or the potent SIRT1 activator SRT1720 prevented SIS in equine tenocytes, while treatment with the non-SIRT1 activating resveratrol analogue V29 was equally protective against SIS, consistent with a novel, as yet uncharacterised SIRT1-indendent mechanism which has relevance for the development of future preventative and therapeutic strategies.


Subject(s)
Cellular Senescence , Dexamethasone , Sirtuin 1 , Tenocytes , Animals , Horses , Sirtuin 1/metabolism , Cellular Senescence/drug effects , Tenocytes/drug effects , Tenocytes/metabolism , Dexamethasone/pharmacology , Resveratrol/pharmacology , Cell Proliferation/drug effects , Tumor Suppressor Protein p53/metabolism , Tendinopathy/metabolism , Tendinopathy/pathology , Tendinopathy/drug therapy , Cells, Cultured , Tendons/drug effects , Tendons/cytology , Tendons/metabolism
8.
J Biochem Mol Toxicol ; 38(9): e23817, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39177155

ABSTRACT

Heavy metal contamination is an alarming concern on a global scale, as drinking tainted water significantly increases human susceptibility to heavy metals. In a realistic scenario, humans are often exposed to a combination of harmful chemicals rather than a single toxicant. Phloretin (PHL), biochanin-A (BCA), and coenzyme Q10 (CoQ10) are bioactive compounds owning plentiful pharmacological properties. Henceforth, the current research explored the putative energizing effects of selected nutraceuticals in combined chromium (Cr) and arsenic (As) intoxicated Swiss albino mice. Potassium dichromate (75 ppm) and sodium meta-arsenite (100 ppm) were given in the drinking water to induce hepatotoxicity, conjugated with PHL and BCA (50 mg/kg each), and CoQ10 (10 mg/kg) intraperitoneally for 2 weeks. After the statistical evaluation, it was observed that the hepato-somatic index, metal load, and antioxidant activity (lipid peroxidation and protein carbonyl content) increased along with the concomitant decrease in the antioxidants (catalase, glutathione-S-transferase, superoxide dismutase, reduced glutathione, and total thiol) in the Cr and As intoxicated mice. Additionally, light microscopy observations, DNA breakages, decreased silent information regulator 1 (SIRT1), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), heme oxygenase (HO-1), and NAD(P)H quinone dehydrogenase 1 (NQO1) gene expressions, together with stimulated apoptotic cell death manifested by the increased expressions of caspase 8 and caspase 3, thus, proved consistency with the aforementioned outcomes. Importantly, the treatment with nutraceuticals not only restored the antioxidant activity but also favorably altered the expressions of SIRT1, Nrf2, HO-1, and NQO1 signaling and apoptosis markers. These findings highlight the crucial role of the PHL, BCA, and CoQ10 combination in reducing Cr and As-induced hepatotoxicity in mice. By averting the triggered apoptosis in conjunction with oxidative stress, this combination increases the SIRT1, Nrf2, HO-1, and NQO1 signaling, thereby reassuringly maintaining the cellular equilibrium.


Subject(s)
Apoptosis , Chromium , Genistein , Liver , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Oxidative Stress , Phloretin , Signal Transduction , Sirtuin 1 , Ubiquinone , Animals , Sirtuin 1/metabolism , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Genistein/pharmacology , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Chromium/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Phloretin/pharmacology , Signal Transduction/drug effects , Male , Arsenic/toxicity , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins
9.
Transl Vis Sci Technol ; 13(8): 37, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39177995

ABSTRACT

Purpose: Adeno-associated virus (AAV) demonstrates promise in delivering therapeutic genes to retinal ganglion cells (RGCs). Delivery of neuroprotective genes is constrained by packaging size and/or cell selectivity. This study compares the ability of the RGC-selective gamma-synuclein (SNCG) promoter and the smaller RGC-selective neurofilament heavy chain (NEFH) promoter, as well as portions of the RGC-selective atonal bHLH transcription factor 7 (ATOH7) enhancer, to drive gene expression in RGCs. Methods: AAV2 constructs with green fluorescent protein (GFP) or human sirtuin 1 (hSIRT1) driven by cytomegalovirus (CMV) enhancer and NEFH promoter (AAV2-eCMV-NEFH) or distal active sequences of the ATOH7 enhancer (DiATOH7) with the SNCG promoter (AAV2-DiATOH7-SNCG) were intravitreally injected into C57BL/6J mice. RGCs were immunolabeled with Brn3a antibodies and counted. AAV constructs with the utmost transduction efficiency were used to test the therapeutic efficacy of the hSIRT1 gene in 12-week-old C57BL/6J mice subjected to microbead (MB)-induced intraocular pressure (IOP) elevation. Visual function was measured using optokinetic responses (OKRs). Results: The eGFP transduction efficiency of AAV2-eCMV-NEFH was similar to that of AAV2-eCMV-SNCG and AAV2-DiATOH7-SNCG. When combined with the SNCG promoter, a larger ATOH7 enhancer was less efficient than the shorter DiATOH7 enhancer. Similarly, the hSIRT1 efficiency of AAV2-eCMV-NEFH was similar to that of AAV2-eCMV-SNCG. The latter two vectors were equally efficient in increasing RGC survival and improving visual function in the mouse model of MB-induced IOP elevation. Conclusions: SNCG and NEFH promoters represent two equally efficient and comparable RGC selective promoter sequences; however, the NEFH promoter offers a smaller packaging size. Translational Relevance: Smaller enhancer-promoter combinations can be used to deliver larger genes in human cells and for treatment in optic neuropathies including glaucoma.


Subject(s)
Dependovirus , Disease Models, Animal , Glaucoma , Mice, Inbred C57BL , Promoter Regions, Genetic , Retinal Ganglion Cells , Sirtuin 1 , gamma-Synuclein , Animals , Sirtuin 1/genetics , Sirtuin 1/metabolism , Mice , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Humans , Glaucoma/genetics , Glaucoma/therapy , Glaucoma/metabolism , Dependovirus/genetics , gamma-Synuclein/genetics , gamma-Synuclein/metabolism , Intravitreal Injections , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Intraocular Pressure/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation
10.
Rev Assoc Med Bras (1992) ; 70(8): e20240314, 2024.
Article in English | MEDLINE | ID: mdl-39166679

ABSTRACT

OBJECTIVE: Placenta accreta spectrum (PAS) is defined as the attachment of the placenta to the uterine wall in varying degrees. However, the studies have explored that the underlying molecular mechanisms of the PAS are very limited. Sirtuins 1 (SIRT1) is associated with placental development by controlling trophoblast cell invasion and remodeling of spiral arteries. We aimed to determine the expression level of SIRT1 in placentas, and maternal and umbilical cord serum of patients with PAS. METHODS: In total, 30 individuals in control, 20 patients in the placenta previa group, and 30 patients in the PAS group were included in this study. The expression levels of SIRT1 in the placentas were determined by Western blot and immunohistochemistry. Serum levels of SIRT1 in maternal and umbilical cord blood were determined by ELISA. RESULTS: SIRT1 was significantly lower in placentas of the PAS. However, maternal and umbilical cord serum samples were not significantly different between groups. CONCLUSION: SIRT1 may play an important role in the pathogenesis of the PAS.


Subject(s)
Fetal Blood , Placenta Accreta , Placenta , Sirtuin 1 , Humans , Female , Pregnancy , Sirtuin 1/blood , Sirtuin 1/analysis , Adult , Placenta/metabolism , Placenta Accreta/blood , Placenta Accreta/pathology , Fetal Blood/metabolism , Case-Control Studies , Immunohistochemistry , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Umbilical Cord/metabolism , Umbilical Cord/pathology , Placenta Previa/blood
11.
Cell Rep Med ; 5(8): 101684, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39128469

ABSTRACT

Sirtuin 1 (SIRT1) is a histone deacetylase and plays diverse functions in various physiological events, from development to lifespan regulation. Here, in Parkinson's disease (PD) model mice, we demonstrated that SIRT1 ameliorates parkinsonism, while SIRT1 knockdown further aggravates PD phenotypes. Mechanistically, SIRT1 interacts with and deacetylates pyruvate kinase M2 (PKM2) at K135 and K206, thus leading to reduced PKM2 enzyme activity and lactate production, which eventually results in decreased glial activation in the brain. Administration of lactate in the brain recapitulates PD-like phenotypes. Furthermore, increased expression of PKM2 worsens PD symptoms, and, on the contrary, inhibition of PKM2 by shikonin or PKM2-IN-1 alleviates parkinsonism in mice. Collectively, our data indicate that excessive lactate in the brain might be involved in the progression of PD. By improving lactate homeostasis, SIRT1, together with PKM2, are likely drug targets for developing agents for the treatment of neurodegeneration in PD.


Subject(s)
Brain , Homeostasis , Lactic Acid , Pyruvate Kinase , Sirtuin 1 , Sirtuin 1/metabolism , Sirtuin 1/genetics , Animals , Brain/metabolism , Brain/pathology , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Mice , Lactic Acid/metabolism , Humans , Acetylation/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/genetics , Disease Models, Animal , Male , Mice, Inbred C57BL , Thyroid Hormone-Binding Proteins , Thyroid Hormones/metabolism , Naphthoquinones/pharmacology
12.
Food Res Int ; 192: 114778, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39147466

ABSTRACT

Fermentation can transform bioactive compounds in food and improve their biological activity. This study aims to explore the transformation of polyphenols in mulberry juice and the improvement of its anti-aging effect. The results demonstrated that Lactobacillus plantarum SC-5 transformed anthocyanin in mulberry juice into more phenolic acids, especially improved 2-hydroxy-3-(4-hydroxyphenyl) propanoic acid from 4.16 ± 0.06 to 10.07 ± 0.03. In the D-gal-induced mouse model, fermented mulberry juice significantly raised the abundance of Bifidobacteriaceae (303.7 %) and Lactobacillaceae (237.2 %) and Short-chain fatty acids (SCFAs) in intestine, further reducing the level of oxidative stress (12.3 %). Meanwhile, the expression of Sirtuin 1 (SIRT1) and Brain-derived neurotrophic factor (BDNF) increased, which protected the integrity of hippocampal tissue. Morris water maze results approved that fermented mulberry juice improved cognitive ability in aging mice (30.3 %). This study provides theoretical support for the view that fermentation is an effective means of developing functional foods.


Subject(s)
Fermentation , Hydroxybenzoates , Lactobacillus plantarum , Morus , Polyphenols , Animals , Morus/chemistry , Polyphenols/pharmacology , Lactobacillus plantarum/metabolism , Hydroxybenzoates/pharmacology , Mice , Male , Fruit and Vegetable Juices , Aging/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Gastrointestinal Microbiome/drug effects , Anthocyanins/pharmacology , Oxidative Stress/drug effects , Fatty Acids, Volatile/metabolism , Sirtuin 1
14.
Redox Biol ; 75: 103300, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39142179

ABSTRACT

Glyoxalase I (GLO1) is the primary enzyme for detoxification of the reactive dicarbonyl methylglyoxal (MG). Loss of GLO1 promotes accumulation of MG resulting in a recapitulation of diabetic phenotypes. We previously demonstrated attenuated GLO1 protein in skeletal muscle from individuals with type 2 diabetes (T2D). However, whether GLO1 attenuation occurs prior to T2D and the mechanisms regulating GLO1 abundance in skeletal muscle are unknown. GLO1 expression and activity were determined in skeletal muscle tissue biopsies from 15 lean healthy individuals (LH, BMI: 22.4 ± 0.7) and 5 individuals with obesity (OB, BMI: 32.4 ± 1.3). GLO1 protein was attenuated by 26 ± 0.3 % in OB compared to LH skeletal muscle (p = 0.019). Similar reductions for GLO1 activity were observed (p = 0.102). NRF2 and Keap1 expression were equivocal between groups despite a 2-fold elevation in GLO1 transcripts in OB skeletal muscle (p = 0.008). GLO1 knock-down (KD) in human immortalized myotubes promoted downregulation of muscle contraction and organization proteins indicating the importance of GLO1 expression for skeletal muscle function. SIRT1 KD had no effect on GLO1 protein or activity whereas, SIRT2 KD attenuated GLO1 protein by 28 ± 0.29 % (p < 0.0001) and GLO1 activity by 42 ± 0.12 % (p = 0.0150). KD of NAMPT also resulted in attenuation of GLO1 protein (28 ± 0.069 %, p = 0.003), activity (67 ± 0.09 %, p = 0.011) and transcripts (50 ± 0.13 %, p = 0.049). Neither the provision of the NAD+ precursors NR nor NMN were able to prevent this attenuation in GLO1 protein. However, NR did augment GLO1 specific activity (p = 0.022 vs NAMPT KD). These perturbations did not alter GLO1 acetylation status. SIRT1, SIRT2 and NAMPT protein levels were all equivocal in skeletal muscle tissue biopsies from individuals with obesity and lean individuals. These data implicate NAD+-dependent regulation of GLO1 in skeletal muscle independent of altered GLO1 acetylation and provide rationale for exploring NR supplementation to rescue attenuated GLO1 abundance and activity in conditions such as obesity.


Subject(s)
Cytokines , Lactoylglutathione Lyase , Muscle, Skeletal , Nicotinamide Phosphoribosyltransferase , Obesity , Sirtuin 2 , Humans , Muscle, Skeletal/metabolism , Lactoylglutathione Lyase/metabolism , Lactoylglutathione Lyase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Sirtuin 2/metabolism , Sirtuin 2/genetics , Cytokines/metabolism , Male , Obesity/metabolism , Obesity/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Female , Adult , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Gene Expression Regulation , Middle Aged , Sirtuin 1/metabolism , Sirtuin 1/genetics
15.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(8): 852-860, 2024 Aug 15.
Article in Chinese | MEDLINE | ID: mdl-39148391

ABSTRACT

OBJECTIVES: To investigate the effect of reactive oxygen species (ROS)/silent information regulator 1 (SIRT1) on hyperoxia-induced mitochondrial injury in BEAS-2B cells. METHODS: The experiment was divided into three parts. In the first part, cells were divided into H0, H6, H12, H24, and H48 groups. In the second part, cells were divided into control group, H48 group, H48 hyperoxia+SIRT1 inhibitor group (H48+EX 527 group), and H48 hyperoxia+SIRT1 agonist group (H48+SRT1720 group). In the third part, cells were divided into control group, 48-hour hyperoxia+N-acetylcysteine group (H48+NAC group), and H48 group. The ROS kit was used to measure the level of ROS. Western blot and immunofluorescent staining were used to measure the expression levels of SIRT1 and mitochondria-related proteins. Transmission electron microscopy was used to observe the morphology of mitochondria. RESULTS: Compared with the H0 group, the H6, H12, H24, and H48 groups had a significantly increased fluorescence intensity of ROS (P<0.05), the H48 group had significant reductions in the expression levels of SIRT1 protein and mitochondria-related proteins (P<0.05), and the H24 and H48 groups had a significant reduction in the fluorescence intensity of mitochondria-related proteins (P<0.05). Compared with the H48 group, the H48+SRT1720 group had significant increases in the expression levels of mitochondria-related proteins and the mitochondrial aspect ratio (P<0.05), and the H48+EX 527 group had a significant reduction in the mitochondrial area (P<0.05). Compared with the H48 group, the H48+NAC group had a significantly decreased fluorescence intensity of ROS (P<0.05) and significantly increased levels of SIRT1 protein, mitochondria-related proteins, mitochondrial area, and mitochondrial aspect ratio (P<0.05). CONCLUSIONS: The ROS/SIRT1 axis is involved in hyperoxia-induced mitochondrial injury in BEAS-2B cells.


Subject(s)
Bronchi , Epithelial Cells , Hyperoxia , Reactive Oxygen Species , Sirtuin 1 , Sirtuin 1/metabolism , Sirtuin 1/physiology , Sirtuin 1/genetics , Humans , Reactive Oxygen Species/metabolism , Hyperoxia/complications , Hyperoxia/metabolism , Epithelial Cells/metabolism , Bronchi/metabolism , Mitochondria/metabolism , Cells, Cultured , Cell Line
16.
Sci Rep ; 14(1): 19171, 2024 08 19.
Article in English | MEDLINE | ID: mdl-39160173

ABSTRACT

The current study was assigned to determine the putative preventive role of vinpocetine (VIN) in cervical hyperkeratosis (CHK) in female rats. Estradiol Benzoate (EB) was utilized in a dose f (60 µg/100 g, i.m) three times/week for 4 weeks to induce cervical hyperkeratosis. VIN was administered alone in a dose of (10 mg/kg/day, orally) for 4 weeks and in the presence of EB. Levels of malondialdehyde (MDA), total nitrites (NOx), reduced glutathione (GSH), interleukin-18 (IL-18), IL-1ß, tumor necrosis factor-alpha (TNF-α) were measured in cervical tissue. The expression of NLRP3/GSDMD/Caspase-1, and SIRT1/Nrf2 was determined using ELISA. Cervical histopathological examination was also done. EB significantly raised MDA, NOx, TNF-α, IL-18, IL-1ß, and GSDMD and up-regulated NLRP3/Caspase-1 proteins. However, GSH, SIRT1, and Nrf2 levels were reduced in cervical tissue. VIN significantly alleviates all biochemical and histopathological abnormalities. VIN considerably mitigates EB-induced cervical hyperkeratosis via NLRP3-induced pyroptosis and SIRT1/Nrf2 signaling pathway.


Subject(s)
Cervix Uteri , Estradiol , Inflammasomes , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Sirtuin 1 , Vinca Alkaloids , Animals , Female , Sirtuin 1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-E2-Related Factor 2/metabolism , Rats , Estradiol/pharmacology , Estradiol/analogs & derivatives , Inflammasomes/metabolism , Inflammasomes/drug effects , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use , Cervix Uteri/pathology , Cervix Uteri/metabolism , Cervix Uteri/drug effects , Signal Transduction/drug effects
17.
Mol Med ; 30(1): 123, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138434

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease associated with high morbidity and mortality worldwide. Oxidative injury and mitochondrial dysfunction in the airway epithelium are major events in COPD progression. METHODS AND RESULTS: The therapeutic effects of Progesterone (P4) were investigated in vivo and in vitro in this study. In vivo, in a cigarette smoke (CS) exposure-induced COPD mouse model, P4 treatment significantly ameliorated CS exposure-induced physiological and pathological characteristics, including inflammatory cell infiltration and oxidative injury, in a dose-dependent manner. The c-MYC/SIRT1/PGC-1α pathway is involved in the protective function of P4 against CS-induced COPD. In vitro, P4 co-treatment significantly ameliorated H2O2-induced oxidative injury and mitochondrial dysfunctions by promoting cell proliferation, increasing mitochondrial membrane potential, decreasing ROS levels and apoptosis, and increasing ATP content. Moreover, P4 co-treatment partially attenuated H2O2-caused inhibition in Nrf1, Tfam, Mfn1, PGR-B, c-MYC, SIRT1, and PGC-1α levels. In BEAS-2B and ASM cells, the c-MYC/SIRT1 axis regulated P4's protective effects against H2O2-induced oxidative injury and mitochondrial dysfunctions. CONCLUSION: P4 activates the c-MYC/SIRT1 axis, ameliorating CS-induced COPD and protecting both airway epithelial cells and smooth muscle cells against H2O2-induced oxidative damage. PGC-1α and downstream mitochondrial signaling pathways might be involved.


Subject(s)
Disease Models, Animal , Hydrogen Peroxide , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Progesterone , Pulmonary Disease, Chronic Obstructive , Sirtuin 1 , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Progesterone/pharmacology , Mice , Sirtuin 1/metabolism , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Hydrogen Peroxide/metabolism , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Line , Cigarette Smoking/adverse effects , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Smoke/adverse effects , Membrane Potential, Mitochondrial/drug effects , Male , Cell Proliferation/drug effects
18.
Sci Rep ; 14(1): 17867, 2024 08 01.
Article in English | MEDLINE | ID: mdl-39090182

ABSTRACT

Diabetic nephropathy (DN) is a prototypical chronic energy metabolism imbalance disease. The AMPK/Sirt1/PGC-1α signaling pathway plays a pivotal role in regulating energy metabolism throughout the body. Gut microbiota ferment indigestible carbohydrates to produce a variety of metabolites, particularly short-chain fatty acids (SCFAs), which exert positive effects on energy metabolism. However, the potential for SCFAs to ameliorate DN-associated renal injury via the AMPK/Sirt1/PGC-1α pathway remains a matter of debate. In this study, we investigated the effects of sodium butyrate (NaB), a SCFA, on energy metabolism in mice with spontaneous DN at two different doses. Body weight, blood glucose and lipid levels, urinary protein excretion, liver and kidney function, interleukin-6 (IL-6) levels, and the expressions of AMPK, phosphorylated AMPK (p-AMPK), mitofusin 2 (MFN2), optic atrophy 1 (OPA1), and glucagon-like peptide-1 receptor (GLP-1R) were monitored in mice. Additionally, butyrate levels, gut microbiota composition, and diversity in colonic stool were also assessed. Our findings demonstrate that exogenous NaB supplementation can improve hyperglycemia and albuminuria, reduce renal tissue inflammation, inhibit extracellular matrix accumulation and glomerular hypertrophy, and could alter the gut microbiota composition in DN. Furthermore, NaB was found to upregulate the expressions of MFN2, OPA1, p-AMPK, and GLP-1R in DN renal tissue. These results suggest that NaB could improve the composition of gut microbiota in DN, activate the AMPK/Sirt1/PGC-1α signaling pathway, and enhance mitochondrial function to regulate energy metabolism throughout the body. Collectively, our findings indicate that NaB may be a novel therapeutic agent for the treatment of DN.


Subject(s)
AMP-Activated Protein Kinases , Butyric Acid , Diabetic Nephropathies , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Mice , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Butyric Acid/pharmacology , Male , Gastrointestinal Microbiome/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Energy Metabolism/drug effects , Mice, Inbred C57BL
19.
Exp Eye Res ; 246: 110019, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39117137

ABSTRACT

Cataracts are the world's number one blinding eye disease. Cataracts can only be effectively treated surgically, although there is a chance of surgical complications. One of the pathogenic processes of cataracts is oxidative stress, which closely correlated with pyroptosis. SIRT1 is essential for the regulation of pyroptosis. Nevertheless, the role of SIRT1 in formation of cataracts is unclear. In this work, we developed an in vitro model of shortwave blue light (SWBL)-induced scotomization in human lens epithelial cells (HLECs) and an in vivo model of SWBL-induced cataracts in rats. The study aimed to understand how the SIRT1/NF-κB/NLRP3 pathway functions. Additionally, the evaluation included cell death and the release of lactate dehydrogenase (LDH), a cytotoxicity marker, from injured cells. First, we discovered that SWBL exposure resulted in lens clouding in Sprague- Dawley (SD) rats and that the degree of clouding was positively linked to the duration of irradiation. Second, we discovered that SIRT1 exhibited antioxidant properties and was connected to the NF-κB/NLRP3 pathway. SWBL irradiation inhibited SIRT1 expression, exacerbated oxidative stress, and promoted nuclear translocation of NF-κB and the activation of the NLRP3 inflammasome, which caused LEC pyroptosis and ultimately led to cataract formation. Transient transfection to increase the expression of SIRT1 decreased the protein expression levels of NF-κB, NLRP3, caspase-1, and GSDMD, inhibited HLEC pyroptosis, and reduced the release of LDH, providing a potential method for cataract prevention and treatment.


Subject(s)
Cataract , Epithelial Cells , Lens, Crystalline , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Sirtuin 1 , Animals , Humans , Rats , Blotting, Western , Blue Light/adverse effects , Cataract/metabolism , Cataract/pathology , Cataract/etiology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Lens, Crystalline/radiation effects , Lens, Crystalline/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Pyroptosis/physiology , Pyroptosis/radiation effects , Rats, Sprague-Dawley , Signal Transduction/physiology , Sirtuin 1/metabolism
20.
Discov Med ; 36(186): 1398-1407, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054711

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disease, often leading to neuroinflammation and neuronal damage. Activation of the Nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome is closely associated with post-SAH neuroinflammation, while activation of Nicotinamide Adenine Dinucleotide (NAD)-dependent deacetylase sirtuin-1 (SIRT1) has neuroprotective effects. This study aimed to investigate the impact of injectable Collagen Binding Domain-Brain Derived Neurotrophic Factor (CBD-BDNF) on neuroinflammation and neuronal damage following SAH. METHODS: After establishing the SAH model, experimental animals were divided into three groups: sham surgery group (Sham), SAH group, and SAH+neuroregenerative scaffold (CBD-BDNF treatment) group. Behavioral performance was evaluated using neurofunctional deficit, beam balance, and Y-maze tests. Expression of inflammatory factors and essential proteins was quantitatively analyzed using Enzyme-Linked Immunosorbent Assay (ELISA) kits and immunoblotting. Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) staining was used to assess cell apoptosis. To further investigate the mechanism of action of CBD-BDNF on SIRT1, the model animals were treated with EX527 (SIRT1 inhibitor) for comparative studies. RESULTS: Neurological deficit tests, CBD-BDNF improves functional outcomes after SAH. Compared to the SAH group, the SAH+neuroregenerative scaffold group showed significantly increased expression of SIRT1 protein and significantly decreased expression of NLRP3, Apoptosis-associated speck-like protein containing a CARD (ASC), and c-caspase-1. The inflammatory cytokines Interleukin-1 beta (IL-1ß), IL-6, and IL-18 levels also significantly decreased in the SAH+neuroregenerative scaffold group. Additionally, animals in the SAH+neuroregenerative scaffold group showed better neurofunctional recovery in neurofunctional deficit and beam balance tests. The number of apoptotic cells significantly decreased in the SAH+neuroregenerative scaffold group compared to the SAH group. However, when SIRT1 was inhibited with EX527, the aforementioned neuroprotective effects were reversed, indicating the involvement of CBD-BDNF through SIRT1 activation. CONCLUSION: This study demonstrates that injectable CBD-BDNF can significantly alleviate neuroinflammation and neuronal damage resulting from SAH by blocking NLRP3 inflammasome activation and promoting SIRT1 expression. These findings provide a new therapeutic strategy for neuroprotection after SAH and reveal the mechanism of action of CBD-BDNF as a potential therapeutic agent. Future research will further explore the long-term efficacy and safety of CBD-BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor , Sirtuin 1 , Subarachnoid Hemorrhage , Sirtuin 1/metabolism , Sirtuin 1/antagonists & inhibitors , Animals , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/complications , Brain-Derived Neurotrophic Factor/metabolism , Male , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Disease Models, Animal , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Rats , Apoptosis/drug effects , Collagen/metabolism , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL