Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 865
Filter
1.
Theranostics ; 14(9): 3719-3738, 2024.
Article in English | MEDLINE | ID: mdl-38948070

ABSTRACT

Rationale: Autophagy dysregulation is known to be a mechanism of doxorubicin (DOX)-induced cardiotoxicity (DIC). Mitochondrial-Endoplasmic Reticulum Contacts (MERCs) are where autophagy initiates and autophagosomes form. However, the role of MERCs in autophagy dysregulation in DIC remains elusive. FUNDC1 is a tethering protein of MERCs. We aim to investigate the effect of DOX on MERCs in cardiomyocytes and explore whether it is involved in the dysregulated autophagy in DIC. Methods: We employed confocal microscopy and transmission electron microscopy to assess MERCs structure. Autophagic flux was analyzed using the mCherry-EGFP-LC3B fluorescence assay and western blotting for LC3BII. Mitophagy was studied through the mCherry-EGFP-FIS1 fluorescence assay and colocalization analysis between LC3B and mitochondria. A total dose of 18 mg/kg of doxorubicin was administrated in mice to construct a DIC model in vivo. Additionally, we used adeno-associated virus (AAV) to cardiac-specifically overexpress FUNDC1. Cardiac function and remodeling were evaluated by echocardiography and Masson's trichrome staining, respectively. Results: DOX blocked autophagic flux by inhibiting autophagosome biogenesis, which could be attributed to the downregulation of FUNDC1 and disruption of MERCs structures. FUNDC1 overexpression restored the blocked autophagosome biogenesis by maintaining MERCs structure and facilitating ATG5-ATG12/ATG16L1 complex formation without altering mitophagy. Furthermore, FUNDC1 alleviated DOX-induced oxidative stress and cardiomyocytes deaths in an autophagy-dependent manner. Notably, cardiac-specific overexpression of FUNDC1 protected DOX-treated mice against adverse cardiac remodeling and improved cardiac function. Conclusions: In summary, our study identified that FUNDC1-meditated MERCs exerted a cardioprotective effect against DIC by restoring the blocked autophagosome biogenesis. Importantly, this research reveals a novel role of FUNDC1 in enhancing macroautophagy via restoring MERCs structure and autophagosome biogenesis in the DIC model, beyond its previously known regulatory role as an mitophagy receptor.


Subject(s)
Autophagy , Cardiotoxicity , Doxorubicin , Endoplasmic Reticulum , Membrane Proteins , Mitochondrial Proteins , Myocytes, Cardiac , Animals , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Mice , Autophagy/drug effects , Cardiotoxicity/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Mitophagy/drug effects , Male , Autophagosomes/metabolism , Autophagosomes/drug effects , Mice, Inbred C57BL , Disease Models, Animal
2.
Redox Rep ; 29(1): 2377870, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39010730

ABSTRACT

OBJECTIVES: To observe the CISD2 expression among PCOS patients and to explore its profound impact on the follicular microenvironment. Moreover, we want to elucidate the intricate mechanistic contribution of CISD2 to the onset and progression of PCOS. METHODS: Oxidase NOX2, mitophagy-related proteins, and CISD2 were detected by WB. The changes in mitochondrial structure and quantity were observed by transmission electron microscopy. Mitochondrial and lysosome colocalization was used to detect the changes of mitophagy. MDA kit, GSH and GSSG Assay kit and ROS probe were used to detect oxidative stress damage. RESULTS: We found that CISD2, mitophagy and oxidase in the GCs of PCOS patients were significantly increased. Testosterone stimulation leads to the increase of oxidase, mitophagy, and CISD2 in KGN cells. CISD2 inhibition promoted the increase of mitophagy, and the activation of mitochondria-lysosome binding, while alleviating the oxidative stress. CONCLUSIONS: Inhibition of CISD2 can improve the occurrence of oxidative stress by increasing the level of mitophagy, thus affecting the occurrence and development of PCOS diseases.


Subject(s)
Mitophagy , Oxidative Stress , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Female , Mitophagy/drug effects , Mitophagy/physiology , Mitochondria/metabolism , Mitochondria/drug effects , Adult , Cellular Microenvironment/physiology
3.
Cells ; 13(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39056765

ABSTRACT

Mitochondria play a fundamental role in the energy metabolism of eukaryotic cells. Numerous studies indicate lead (Pb) as a widely occurring environmental factor capable of disrupting oxidative metabolism by modulating the mitochondrial processes. The multitude of known molecular targets of Pb and its strong affinity for biochemical pathways involving divalent metals suggest that it may pose a health threat at any given dose. Changes in the bioenergetics of cells exposed to Pb have been repeatedly demonstrated in research, primarily showing a reduced ability to synthesize ATP. In addition, lead interferes with mitochondrial-mediated processes essential for maintaining homeostasis, such as apoptosis, mitophagy, mitochondrial dynamics, and the inflammatory response. This article describes selected aspects of mitochondrial metabolism in relation to potential mechanisms of energy metabolism disorders induced by Pb.


Subject(s)
Energy Metabolism , Lead , Mitochondria , Humans , Lead/toxicity , Lead/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Energy Metabolism/drug effects , Animals , Mitophagy/drug effects , Apoptosis/drug effects , Mitochondrial Dynamics/drug effects
4.
Mol Med Rep ; 30(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-39054966

ABSTRACT

Sepsis is a life­threatening multiple organ failure disease caused by an uncontrolled inflammatory response and can progress to acute lung injury (ALI). Heat­shock protein B8 (HSPB8) serves a cytoprotective role in multiple types of diseases; however, to the best of our knowledge, the regulatory role of HSPB8 in sepsis­induced ALI remains unclear. A549 human alveolar type II epithelial cells were treated with lipopolysaccharide (LPS) for 24 h to simulate a sepsis­induced ALI model. Cell transfection was performed to overexpress HSPB8, and cells were treated with mitochondrial division inhibitor­1 (Mdivi­1) for 2 h before LPS induction to assess the underlying mechanism. Protein expression was evaluated using western blotting and an immunofluorescence assay. Cytokines were examined using ELISA assay kits and antioxidant enzymes were examined using their detection kits. Cell apoptosis was detected using flow cytometry. The mitochondrial membrane potential was detected by JC­1 staining. HSPB8 was upregulated in A549 cells treated with LPS and HSPB8 overexpression attenuated LPS­induced inflammatory cytokine levels, oxidative stress and apoptosis in A549 cells. LPS inhibited mitophagy and reduced the mitochondrial membrane potential in A549 cells, which was partly inhibited by HSPB8 overexpression. Furthermore, Mdivi­1 decreased the inhibitory effect of HSPB8 on the inflammatory response, oxidative stress and apoptosis in LPS­treated A549 cells. In conclusion, HSPB8 overexpression attenuated the LPS­mediated inflammatory response, oxidative stress and apoptosis in A549 cells by promoting mitophagy, indicating HSPB8 as a potential therapeutic target in sepsis­induced ALI.


Subject(s)
Acute Lung Injury , Apoptosis , Cytokines , Heat-Shock Proteins , Lipopolysaccharides , Membrane Potential, Mitochondrial , Mitophagy , Molecular Chaperones , Oxidative Stress , Humans , Mitophagy/drug effects , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Lipopolysaccharides/adverse effects , A549 Cells , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Apoptosis/drug effects , Oxidative Stress/drug effects , Membrane Potential, Mitochondrial/drug effects , Cytokines/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/genetics
6.
Metallomics ; 16(7)2024 07 01.
Article in English | MEDLINE | ID: mdl-38955388

ABSTRACT

Both 8-hydroxyquinoline compounds and iridium (Ir) complexes have emerged as potential novel agents for tumor therapy. In this study, we synthesized and characterized two new Ir(III) complexes, [Ir(L1)(bppy)2] (Br-Ir) and [Ir(L2)(bppy)2] (Cl-Ir), with 5,7-dibromo-2-methyl-8-hydroxyquinoline (HL-1) or 5,7-dichloro-2-methyl-8-hydroxyquinoline as the primary ligand. Complexes Br-Ir and Cl-Ir successfully inhibited antitumor activity in Hep-G2 cells. In addition, complexes Br-Ir and Cl-Ir were localized in the mitochondrial membrane and caused mitochondrial damage, autophagy, and cellular immunity in Hep-G2 cells. We tested the proteins related to mitochondrial and mitophagy by western blot analysis, which showed that they triggered mitophagy-mediated apoptotic cell death. Remarkably, complex Br-Ir showed high in vivo antitumor activity, and the tumor growth inhibition rate was 63.0% (P < 0.05). In summary, our study on complex Br-Ir revealed promising results in in vitro and in vivo antitumor activity assays.


Subject(s)
Antineoplastic Agents , Iridium , Mitochondria , Humans , Iridium/chemistry , Iridium/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Hep G2 Cells , Mice , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Apoptosis/drug effects , Oxyquinoline/pharmacology , Oxyquinoline/chemistry , Oxyquinoline/analogs & derivatives , Mice, Inbred BALB C , Mitophagy/drug effects , Mice, Nude
8.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000117

ABSTRACT

Diabetic cardiomyopathy (DCM) is a major determinant of mortality in diabetic populations, and the potential strategies are insufficient. Canagliflozin has emerged as a potential cardioprotective agent in diabetes, yet its underlying molecular mechanisms remain unclear. We employed a high-glucose challenge (60 mM for 48 h) in vitro to rat cardiomyocytes (H9C2), with or without canagliflozin treatment (20 µM). In vivo, male C57BL/6J mice were subjected to streptozotocin and a high-fat diet to induce diabetes, followed by canagliflozin administration (10, 30 mg·kg-1·d-1) for 12 weeks. Proteomics and echocardiography were used to assess the heart. Histopathological alterations were assessed by the use of Oil Red O and Masson's trichrome staining. Additionally, mitochondrial morphology and mitophagy were analyzed through biochemical and imaging techniques. A proteomic analysis highlighted alterations in mitochondrial and autophagy-related proteins after the treatment with canagliflozin. Diabetic conditions impaired mitochondrial respiration and ATP production, alongside decreasing the related expression of the PINK1-Parkin pathway. High-glucose conditions also reduced PGC-1α-TFAM signaling, which is responsible for mitochondrial biogenesis. Canagliflozin significantly alleviated cardiac dysfunction and improved mitochondrial function both in vitro and in vivo. Specifically, canagliflozin suppressed mitochondrial oxidative stress, enhancing ATP levels and sustaining mitochondrial respiratory capacity. It activated PINK1-Parkin-dependent mitophagy and improved mitochondrial function via increased phosphorylation of adenosine monophosphate-activated protein kinase (AMPK). Notably, PINK1 knockdown negated the beneficial effects of canagliflozin on mitochondrial integrity, underscoring the critical role of PINK1 in mediating these protective effects. Canagliflozin fosters PINK1-Parkin mitophagy and mitochondrial function, highlighting its potential as an effective treatment for DCM.


Subject(s)
Canagliflozin , Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Mice, Inbred C57BL , Mitophagy , Protein Kinases , Ubiquitin-Protein Ligases , Animals , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Mitophagy/drug effects , Male , Mice , Protein Kinases/metabolism , Protein Kinases/genetics , Rats , Canagliflozin/pharmacology , Canagliflozin/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line , Signal Transduction/drug effects , Diet, High-Fat/adverse effects
9.
Alzheimers Res Ther ; 16(1): 160, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030577

ABSTRACT

BACKGROUND: Alpha-lipoic acid (ALA) has a neuroprotective effect on neurodegenerative diseases. In the clinic, ALA can improve cognitive impairments in patients with Alzheimer's disease (AD) and other dementias. Animal studies have confirmed the anti-amyloidosis effect of ALA, but its underlying mechanism remains unclear. In particular, the role of ALA in amyloid-ß precursor protein (APP) metabolism has not been fully elucidated. OBJECTIVE: To investigate whether ALA can reduce the amyloidogenic effect of APP in a transgenic mouse model of AD, and to study the mechanism underlying this effect. METHODS: ALA was infused into 2-month-old APP23/PS45 transgenic mice for 4 consecutive months and their cognitive function and AD-like pathology were then evaluated. An ALA drug concentration gradient was applied to 20E2 cells in vitro to evaluate its effect on the expression of APP proteolytic enzymes and metabolites. The mechanism by which ALA affects APP processing was studied using GI254023X, an inhibitor of A Disintegrin and Metalloproteinase 10 (ADAM10), as well as the mitochondrial toxic drug carbonyl cyanide m-chlorophenylhydrazone (CCCP). RESULTS: Administration of ALA ameliorated amyloid plaque neuropathology in the brain tissue of APP23/PS45 mice and reduced learning and memory impairment. ALA also increased the expression of ADAM10 in 20E2 cells and the non-amyloidogenic processing of APP to produce the 83 amino acid C-terminal fragment (C83). In addition to activating autophagy, ALA also significantly promoted mitophagy. BNIP3L-knockdown reduced the mat/pro ratio of ADAM10. By using CCCP, ALA was found to regulate BNIP3L-mediated mitophagy, thereby promoting the α-cleavage of APP. CONCLUSIONS: The enhanced α-secretase cleavage of APP by ADAM10 is the primary mechanism through which ALA ameliorates the cognitive deficits in APP23/PS45 transgenic mice. BNIP3L-mediated mitophagy contributes to the anti-amyloid properties of ALA by facilitating the maturation of ADAM10. This study provides novel experimental evidence for the treatment of AD with ALA.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor , Cognitive Dysfunction , Mice, Transgenic , Mitophagy , Thioctic Acid , Animals , Thioctic Acid/pharmacology , Mitophagy/drug effects , ADAM10 Protein/metabolism , Mice , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Amyloid Precursor Protein Secretases/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Disease Models, Animal , Membrane Proteins/metabolism , Membrane Proteins/genetics , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Male
10.
Cell Death Dis ; 15(7): 484, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969639

ABSTRACT

An increasing evidence supports that cell competition, a vital selection and quality control mechanism in multicellular organisms, is involved in tumorigenesis and development; however, the mechanistic contributions to the association between cell competition and tumor drug resistance remain ill-defined. In our study, based on a contructed lenvitinib-resistant hepatocellular carcinoma (HCC) cells display obvious competitive growth dominance over sensitive cells through reprogramming energy metabolism. Mechanistically, the hyperactivation of BCL2 interacting protein3 (BNIP3) -mediated mitophagy in lenvatinib-resistant HCC cells promotes glycolytic flux via shifting energy production from mitochondrial oxidative phosphorylation to glycolysis, by regulating AMP-activated protein kinase (AMPK) -enolase 2 (ENO2) signaling, which perpetually maintaining lenvatinib-resistant HCC cells' competitive advantage over sensitive HCC cells. Of note, BNIP3 inhibition significantly sensitized the anti-tumor efficacy of lenvatinib in HCC. Our findings emphasize a vital role for BNIP3-AMPK-ENO2 signaling in maintaining the competitive outcome of lenvitinib-resistant HCC cells via regulating energy metabolism reprogramming; meanwhile, this work recognizes BNIP3 as a promising target to overcome HCC drug resistance.


Subject(s)
Carcinoma, Hepatocellular , Drug Resistance, Neoplasm , Energy Metabolism , Liver Neoplasms , Membrane Proteins , Mitophagy , Phenylurea Compounds , Quinolines , Humans , Quinolines/pharmacology , Mitophagy/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Membrane Proteins/metabolism , Energy Metabolism/drug effects , Phenylurea Compounds/pharmacology , Drug Resistance, Neoplasm/drug effects , Animals , Cell Line, Tumor , Proto-Oncogene Proteins/metabolism , Mice , Mice, Nude , Cell Proliferation/drug effects , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Mice, Inbred BALB C , Metabolic Reprogramming
11.
Int Immunopharmacol ; 138: 112652, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38986301

ABSTRACT

Tendinopathy is one of the most prevalent sports injury diseases in orthopedics. However, there is no effective treatment or medicine. Recently, the discovery of tendon stem cells (TSCs) provides a new perspective to find new therapeutic methods for Tendinopathy. Studies have shown that oxidative stress will inevitably cause TSCs injury during tendinopathy, but the mechanism has not been fully elucidated. Here, we report the oxidative damage of TSCs induced by H2O2 via ferroptosis, as well, treatment with H2O2 raised the proportion of mitochondria engulfed by autophagosomes in TSCs. The suppression of mitophagy by Mdivi-1 significantly attenuates the H2O2-induced ferroptosis in TSCs. Mechanically, H2O2 actives the cGAS-STING pathway, which can regulate the level of mitophagy. Interfering with cGAS could impair mitophagy and the classical ferroptotic events. In the rat model of tendinopathy, interference of cGAS could relieve tendon injury by inhibiting ferroptosis. Overall, these results provided novel implications to reveal the molecular mechanism of tendinopathy, by which pointed to cGAS as a potential therapeutic target for the treatment of tendinopathy.


Subject(s)
Ferroptosis , Hydrogen Peroxide , Membrane Proteins , Mitophagy , Nucleotidyltransferases , Oxidative Stress , Signal Transduction , Stem Cells , Tendons , Mitophagy/drug effects , Animals , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Stem Cells/metabolism , Tendons/pathology , Tendons/metabolism , Rats , Hydrogen Peroxide/metabolism , Humans , Male , Rats, Sprague-Dawley , Tendinopathy/metabolism , Tendinopathy/pathology , Cells, Cultured
12.
Mol Biol Rep ; 51(1): 870, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080104

ABSTRACT

BACKGROUND: Pediatric postoperative cognitive dysfunction (POCD) is a prevalent complication following anesthesia and surgery. Hypoxia and propofol are the primary risk factors contributing to pediatric POCD. Our previous in vivo animal research has demonstrated that cognitive dysfunction in immature Sprague-Dawley (SD) rats, induced by hypoxia combined with propofol (HCWP), is closely associated with hippocampal neuron ferroptosis. METHODS AND RESULTS: In vivo transcriptome sequencing and KEGG functional analysis revealed significant enrichment of the mitophagy pathway. To further elucidate the relationship between mitophagy and ferroptosis, HT22 cells were selected to construct an in vitro HCWP model. Our findings indicate that HCWP activates excessive mitophagy in HT22 cells, leading to decreased mitochondrial membrane potential (ΔΨm), reactive oxygen species (ROS) burst, mitochondrial fragmentation, and the induction of ferroptosis. To explore this causal relationship further, we employed Mdivi-1, a mitophagy inhibitor. Notably, low-dose Mdivi-1 (10 µM) effectively suppressed excessive mitophagy in HT22 cells, improved mitochondrial function and morphology, and mitigated markers associated with ferroptosis. The mechanism by which Mdivi-1 alleviates HCWP-induced ferroptosis in HT22 cells is likely due to its inhibition of excessive mitophagy, thereby promoting mitochondrial homeostasis. CONCLUSIONS: Our study suggests that mitophagy may be an upstream event in HCWP-induced ferroptosis in HT22 cells. Consequently, targeted regulation of mitophagy by Mdivi-1 may represent a promising approach to prevent cognitive dysfunction following HCWP exposure.


Subject(s)
Ferroptosis , Membrane Potential, Mitochondrial , Mitophagy , Propofol , Quinazolinones , Reactive Oxygen Species , Mitophagy/drug effects , Propofol/pharmacology , Ferroptosis/drug effects , Animals , Quinazolinones/pharmacology , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Rats , Mitochondria/drug effects , Mitochondria/metabolism , Cell Line , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Mice , Hypoxia/metabolism , Hypoxia/complications , Neurons/metabolism , Neurons/drug effects , Rats, Sprague-Dawley , Cell Hypoxia/drug effects , Postoperative Cognitive Complications/metabolism
13.
FASEB J ; 38(15): e23838, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39082250

ABSTRACT

EXPRESSION OF CONCERN: I. Ramesh, J. C. Campos, P. Lee, Y. Song, G. Hernandez, J. Sin, K. C. Tucker, H. Saadaeijahromi, M. Gurney, J. C. B. Ferreira, and A. M. Andres, "Mitophagy Protects Against Statin-Mediated Skeletal Muscle Toxicity," The FASEB Journal 33, no. 11 (2019): 11857-11869, https://doi.org/10.1096/fj.201900807RR. This Expression of Concern is for the above article, published online on August 23, 2019, in Wiley Online Library (wileyonlinelibrary.com) and has been published by agreement between the journal Editor-in-Chief, Loren E. Wold; the Federation of American Societies for Experimental Biology; and Wiley Periodicals LLC. The Expression of Concern has been published due to concerns raised by a third party regarding a duplication between the COX-IV panel of Figure 3C and the COX-IV panel of Figure 5D. The authors have been informed about the concerns, but due to the time elapsed since publication, they could not provide the original raw data. Consequently, the journal team could not verify the validity of these figures describing different experimental conditions and could not exclude that these image duplications affect the overall conclusions of the article. Therefore, the journal has decided to issue an Expression of Concern to inform and alert the readers.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Mitophagy , Muscle, Skeletal , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Animals , Mitophagy/drug effects , Mice , Humans
14.
Biomed Pharmacother ; 177: 117144, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39004063

ABSTRACT

Alzheimer's disease (AD) is a prevalent neurodegenerative disorder and the leading cause of age-related cognitive decline. Recent studies have established a close relationship between mitophagy and the pathogenesis of AD. Various phytochemicals have shown promising therapeutic effects in mitigating the onset and progression of AD. This review offers a comprehensive overview of the typical features of mitophagy and the underlying mechanisms leading to its occurrence in AD, highlighting its significance in the disease's pathogenesis and progression. Additionally, we examine the therapeutic mechanisms of synthetic drugs that induce mitophagy in AD. Finally, we summarize recent advances in research on phytochemicals that regulate mitophagy in the treatment of AD, potentially guiding the development of new anti-AD drugs.


Subject(s)
Alzheimer Disease , Mitophagy , Phytochemicals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , Mitophagy/drug effects , Phytochemicals/therapeutic use , Phytochemicals/pharmacology , Animals , Mitochondria/drug effects , Mitochondria/metabolism
15.
Biomed Pharmacother ; 177: 117092, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38976956

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE) is a disease characterized by neuroinflammation and cognitive dysfunction caused by systemic infection. Inflammation-induced microglial activation is closely associated with neuroinflammation in SAE. It is widely understood that melatonin has strong anti-inflammatory and immunomodulatory properties beneficial for sepsis-related brain damage. However, the mechanism of melatonin action in SAE has not been fully elucidated. METHODS: The SAE cell model and SAE mouse model were induced by lipopolysaccharide (LPS). Behavioral tests were performed to analyze cognitive function. Microglial markers and M1/M2 markers were measured by immunofluorescence. Mitophagy was assessed by western blot, mt-Keima and transmission electron microscopy experiments. Immunoprecipitation and co-immunoprecipitation assays investigated the interactions between AMP-activated protein kinase α2 (AMPKα2) and PTEN-induced putative kinase 1 (PINK1). RESULTS: Melatonin suppresses LPS-induced microglia M1 polarization by enhancing mitophagy, thereby attenuating LPS-induced neuroinflammation and behavioral deficits. However, inhibition or knockdown of AMPKα2 can inhibit the enhancement of melatonin on mitophagy, then weaken its promotion of microglia polarization towards M2 phenotype, and eliminate its protective effect on brain function. Furthermore, melatonin enhances mitophagy through activating AMPKα2, promotes PINK1 Ser495 site phosphorylation, and ultimately regulates microglial polarization from M1 to M2. CONCLUSIONS: Our findings demonstrate that melatonin facilitates microglia polarization towards M2 phenotype to alleviate LPS-induced neuroinflammation, primarily through AMPKα2-mediated enhancement of mitophagy.


Subject(s)
AMP-Activated Protein Kinases , Lipopolysaccharides , Melatonin , Microglia , Mitophagy , Sepsis-Associated Encephalopathy , Melatonin/pharmacology , Animals , Sepsis-Associated Encephalopathy/metabolism , Sepsis-Associated Encephalopathy/drug therapy , AMP-Activated Protein Kinases/metabolism , Microglia/drug effects , Microglia/metabolism , Mitophagy/drug effects , Mice , Male , Mice, Inbred C57BL , Protein Kinases/metabolism , Disease Models, Animal , Sepsis/complications , Sepsis/metabolism , Sepsis/drug therapy , Cell Line , Cell Polarity/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism
16.
Int Immunopharmacol ; 138: 112527, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38950457

ABSTRACT

BACKGROUND: Sepsis-associated acute kidney injury (SA-AKI) represents a frequent complication of in critically ill patients. The objective of this study is to illuminate the potential protective activity of Micheliolide (MCL) and its behind mechanism against SA-AKI. METHODS: The protective potential of MCL on SA-AKI was investigated in lipopolysaccharide (LPS) treated HK2 cells and SA-AKI mice model. The mitochondrial damage was determined by detection of reactive oxygen species and membrane potential. The Nrf2 silencing was achieved by transfection of Nrf2-shRNA in HK2 cells, and Nrf2 inhibitor, ML385 was employed in SA-AKI mice. The mechanism of MCL against SA-AKI was evaluated through detecting hallmarks related to inflammation, mitophagy and Nrf2 pathway via western blotting, immunohistochemistry, and enzyme linked immunosorbent assay. RESULTS: MCL enhanced viability, suppressed apoptosis, decreased inflammatory cytokine levels and improved mitochondrial damage in LPS-treated HK2 cells, and ameliorated renal injury in SA-AKI mice. Moreover, MCL could reduce the activation of NLRP3 inflammasome via enhancing mitophagy. Additionally, Nrf2 deficiency reduced the suppression effect of MCL on NLRP3 inflammasome activation and blocked the facilitation effect of MCL on mitophagy in LPS-treated HK2 cells, the consistent is true for ML385 treatment in SA-AKI mice. CONCLUSIONS: MCL might target Nrf2 and further reduce the NLRP3 inflammasome activation via enhancing mitophagy, which alleviated SA-AKI.


Subject(s)
Acute Kidney Injury , Mitophagy , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Kinases , Sesquiterpenes, Guaiane , Ubiquitin-Protein Ligases , Animals , Humans , Male , Mice , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/chemically induced , Cell Line , Disease Models, Animal , Inflammasomes/metabolism , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , Mitophagy/drug effects , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Kinases/metabolism , Sepsis/drug therapy , Sepsis/complications , Sesquiterpenes, Guaiane/pharmacology , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
17.
Sci Rep ; 14(1): 13655, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38871974

ABSTRACT

Barth syndrome (BTHS) is a lethal rare genetic disorder, which results in cardiac dysfunction, severe skeletal muscle weakness, immune issues and growth delay. Mutations in the TAFAZZIN gene, which is responsible for the remodeling of the phospholipid cardiolipin (CL), lead to abnormalities in mitochondrial membrane, including alteration of mature CL acyl composition and the presence of monolysocardiolipin (MLCL). The dramatic increase in the MLCL/CL ratio is the hallmark of patients with BTHS, which is associated with mitochondrial bioenergetics dysfunction and altered membrane ultrastructure. There are currently no specific therapies for BTHS. Here, we showed that cardiac mitochondria isolated from TAFAZZIN knockdown (TazKD) mice presented abnormal ultrastructural membrane morphology, accumulation of vacuoles, pro-fission conditions and defective mitophagy. Interestingly, we found that in vivo treatment of TazKD mice with a CL-targeted small peptide (named SS-31) was able to restore mitochondrial morphology in tafazzin-deficient heart by affecting specific proteins involved in dynamic process and mitophagy. This agrees with our previous data showing an improvement in mitochondrial respiratory efficiency associated with increased supercomplex organization in TazKD mice under the same pharmacological treatment. Taken together our findings confirm the beneficial effect of SS-31 in the amelioration of tafazzin-deficient dysfunctional mitochondria in a BTHS animal model.


Subject(s)
Acyltransferases , Barth Syndrome , Cardiolipins , Disease Models, Animal , Mitochondria, Heart , Mitophagy , Animals , Barth Syndrome/metabolism , Barth Syndrome/genetics , Barth Syndrome/pathology , Barth Syndrome/drug therapy , Mitophagy/drug effects , Mice , Acyltransferases/metabolism , Acyltransferases/genetics , Cardiolipins/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Lysophospholipids/metabolism , Mice, Knockout , Oligopeptides
18.
Chem Res Toxicol ; 37(6): 1053-1061, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38847154

ABSTRACT

Poisoning caused by the mushroom Amanita phalloides, due to the toxin α-amanitin, accounts for approximately 90% of food poisoning deaths in China with no specific antidotes. To investigate the role of salidroside (Sal) in α-amanitin (α-AMA)-induced mitophagy, mouse liver cells AML-12 were exposed to α-AMA in the presence of Sal or not. Intracellular reactive oxygen species (ROS) levels were measured using a ROS detection kit, mitochondrial activity was evaluated using a mitochondrial red fluorescent probe kit or JC-1 dye, and protein expression levels of PINK1, Parkin, LC3 II, P62, Bax, Bcl-2, Caspase 3, Cleaved-Caspase 3, PARP I, and Cleaved-PARP I were detected through Western blot. Results demonstrated that α-AMA led to increased intracellular ROS levels, cell apoptosis, and decreased mitochondrial membrane potential. Notably, expression levels of mitophagy-related proteins PINK1, Parkin, and LC3 increased significantly while the P62 protein expression decreased remarkably. Furthermore, Sal reversed the α-AMA-induced decrease in cell viability and mitochondrial membrane potential and increase in intracellular ROS level. In addition, Sal promoted expression levels of PINK1, Parkin, and LC3 II while suppressing the Bax/Bcl-2 ratio, Cleaved-Caspase 3, and Cleaved-PARP I as well as P62. The results above proved that salidroside alleviates α-AMA-induced mouse liver cells damage via promoting PINK1/Parkin-mediated mitophagy and reducing cell apoptosis.


Subject(s)
Apoptosis , Glucosides , Mitochondria , Mitophagy , Phenols , Protein Kinases , Reactive Oxygen Species , Ubiquitin-Protein Ligases , Animals , Apoptosis/drug effects , Ubiquitin-Protein Ligases/metabolism , Phenols/pharmacology , Phenols/chemistry , Glucosides/pharmacology , Glucosides/chemistry , Mice , Protein Kinases/metabolism , Mitophagy/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Cell Line , Cell Survival/drug effects
19.
Mol Neurodegener ; 19(1): 49, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890703

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in elderly people in the developed world, and the number of people affected is expected to almost double by 2040. The retina presents one of the highest metabolic demands in our bodies that is partially or fully fulfilled by mitochondria in the neuroretina and retinal pigment epithelium (RPE), respectively. Together with its post-mitotic status and constant photooxidative damage from incoming light, the retina requires a tightly-regulated housekeeping system that involves autophagy. The natural polyphenol Urolithin A (UA) has shown neuroprotective benefits in several models of aging and age-associated disorders, mostly attributed to its ability to induce mitophagy and mitochondrial biogenesis. Sodium iodate (SI) administration recapitulates the late stages of AMD, including geographic atrophy and photoreceptor cell death. METHODS: A combination of in vitro, ex vivo and in vivo models were used to test the neuroprotective potential of UA in the SI model. Functional assays (OCT, ERGs), cellular analysis (flow cytometry, qPCR) and fine confocal microscopy (immunohistochemistry, tandem selective autophagy reporters) helped address this question. RESULTS: UA alleviated neurodegeneration and preserved visual function in SI-treated mice. Simultaneously, we observed severe proteostasis defects upon SI damage induction, including autophagosome accumulation, that were resolved in animals that received UA. Treatment with UA restored autophagic flux and triggered PINK1/Parkin-dependent mitophagy, as previously reported in the literature. Autophagy blockage caused by SI was caused by severe lysosomal membrane permeabilization. While UA did not induce lysosomal biogenesis, it did restore upcycling of permeabilized lysosomes through lysophagy. Knockdown of the lysophagy adaptor SQSTM1/p62 abrogated viability rescue by UA in SI-treated cells, exacerbated lysosomal defects and inhibited lysophagy. CONCLUSIONS: Collectively, these data highlight a novel putative application of UA in the treatment of AMD whereby it bypasses lysosomal defects by promoting p62-dependent lysophagy to sustain proteostasis.


Subject(s)
Coumarins , Animals , Mice , Coumarins/pharmacology , Autophagy/drug effects , Autophagy/physiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Retina/metabolism , Retina/drug effects , Retina/pathology , Mitophagy/drug effects , Mitophagy/physiology , Sequestosome-1 Protein/metabolism , Lysosomes/metabolism , Lysosomes/drug effects , Humans , Disease Models, Animal , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Iodates/toxicity
20.
FASEB J ; 38(12): e23723, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38865198

ABSTRACT

Hypoxia-induced inflammation and apoptosis are important pathophysiological features of heat stroke-induced acute kidney injury (HS-AKI). Hypoxia-inducible factor (HIF) is a key protein that regulates cell adaptation to hypoxia. HIF-prolyl hydroxylase inhibitor (HIF-PHI) stabilizes HIF to increase cell adaptation to hypoxia. Herein, we reported that HIF-PHI pretreatment significantly improved renal function, enhanced thermotolerance, and increased the survival rate of mice in the context of HS. Moreover, HIF-PHI could alleviate HS-induced mitochondrial damage, inflammation, and apoptosis in renal tubular epithelial cells (RTECs) by enhancing mitophagy in vitro and in vivo. By contrast, mitophagy inhibitors Mdivi-1, 3-MA, and Baf-A1 reversed the renoprotective effects of HIF-PHI. Mechanistically, HIF-PHI protects RTECs from inflammation and apoptosis by enhancing Bcl-2 adenovirus E18 19-kDa-interacting protein 3 (BNIP3)-mediated mitophagy, while genetic ablation of BNIP3 attenuated HIF-PHI-induced mitophagy and abolished HIF-PHI-mediated renal protection. Thus, our results indicated that HIF-PHI protects renal function by upregulating BNIP3-mediated mitophagy to improve HS-induced inflammation and apoptosis of RTECs, suggesting HIF-PHI as a promising therapeutic agent to treat HS-AKI.


Subject(s)
Acute Kidney Injury , Heat Stroke , Membrane Proteins , Mitophagy , Prolyl-Hydroxylase Inhibitors , Animals , Male , Mice , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/etiology , Apoptosis/drug effects , Heat Stroke/complications , Heat Stroke/drug therapy , Heat Stroke/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitophagy/drug effects , Prolyl-Hydroxylase Inhibitors/pharmacology , Prolyl-Hydroxylase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL