Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 775
Filter
1.
Acta Neuropathol Commun ; 12(1): 90, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851733

ABSTRACT

Mitochondrial dysfunctions are key features of Alzheimer's disease (AD). The occurrence of these disturbances in the peripheral cells of AD patients and their potential correlation with disease progression are underinvestigated. We studied mitochondrial structure, function and mitophagy in fibroblasts from healthy volunteers and AD patients at the prodromal (AD-MCI) or demented (AD-D) stages. We carried out correlation studies with clinical cognitive scores, namely, (i) Mini-Mental State Examination (MMSE) and (ii) Dementia Rating-Scale Sum of Boxes (CDR-SOB), and with (iii) amyloid beta (Aß) plaque burden (PiB-PET imaging) and (iv) the accumulation of peripheral amyloid precursor protein C-terminal fragments (APP-CTFs). We revealed alterations in mitochondrial structure as well as specific mitochondrial dysfunction signatures in AD-MCI and AD-D fibroblasts and revealed that defective mitophagy and autophagy are linked to impaired lysosomal activity in AD-D fibroblasts. We reported significant correlations of a subset of these dysfunctions with cognitive decline, AD-related clinical hallmarks and peripheral APP-CTFs accumulation. This study emphasizes the potential use of peripheral cells for investigating AD pathophysiology.


Subject(s)
Alzheimer Disease , Fibroblasts , Mitochondria , Mitophagy , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Fibroblasts/pathology , Fibroblasts/metabolism , Aged , Female , Mitochondria/pathology , Mitochondria/metabolism , Male , Mitophagy/physiology , Middle Aged , Aged, 80 and over , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Cognitive Dysfunction/pathology , Cognitive Dysfunction/metabolism , Autophagy/physiology
2.
Mil Med Res ; 11(1): 32, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38812059

ABSTRACT

Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.


Subject(s)
Mitochondria , Mitophagy , Humans , Mitochondria/metabolism , Mitochondria/physiology , Mitophagy/physiology , Mitophagy/drug effects , Mitochondrial Dynamics/physiology
3.
Physiol Res ; 73(2): 253-263, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38710055

ABSTRACT

Up to now, there's a limited number of studies on the relationship between PINK1/Park2 pathway and mitophagy in NAFLD. To investigate the effect of Park2-mediated mitophagy on non-alcoholic fatty liver disease (NAFLD). Oleic acid was used for the establishment of NAFLD model. Oil red-dyed lipid drops and mitochondrial alternations were observed by transmission electron microscopy. Enzymatic kit was used to test lipid content. The levels of IL-8 and TNF-alpha were determined by ELISA. Lenti-Park2 and Park2-siRNA were designed to upregulate and downregulate Park2 expression, respectively. The changing expression of PINK and Park2 was detected by RT-qPCR and Western blot. Immunofluorescence staining was applied to measure the amount of LC3. Successful NAFLD modeling was featured by enhanced lipid accumulation, as well as the elevated total cholesterol (TC), triglyceride (TG), TNF-alpha and IL-8 levels. Mitochondria in NAFLD model were morphologically and functionally damaged. Park2 expression was upregulated by lenti-Park2 and downregulated through Park2-siRNA. The PINK1 expression showed the same trend as Park2 expression. Immunofluorescence staining demonstrated that the when Park2 was overexpressed, more LC3 protein on mitochondrial autophagosome membrane was detected, whereas Park2 knockdown impeded LC3' locating on the membrane. The transmission electron microscopy image exhibited that the extent of damage to the mitochondrial in NAFLD model was revered by enhanced Park2 expression but further exacerbated by reduced Park2 expression. Park2-mediated mitophagy could relive NAFLD and may be a novel therapeutic target for NAFLD treatment. Keywords: Non-alcoholic Fatty Liver Disease (NAFLD), Mitophagy, PINK1/Park2, Park2, PINK1.


Subject(s)
Mitophagy , Non-alcoholic Fatty Liver Disease , Protein Kinases , Ubiquitin-Protein Ligases , Animals , Humans , Male , Mice , Mitophagy/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/genetics , Protein Kinases/metabolism , Protein Kinases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
4.
Reprod Biol ; 24(2): 100889, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733657

ABSTRACT

Mitophagy, the cellular process that removes damaged mitochondria, plays a crucial role in maintaining normal cell functions. It is deeply involved in the entire process of follicle development and is associated with various ovarian diseases. This review aims to provide a comprehensive overview of mitophagy regulation, emphasizing its role at different stages of follicular development. Additionally, the study illuminates the relationship between mitophagy and ovarian diseases, including ovary aging (OA), primary ovarian insufficiency (POI), and polycystic ovary syndrome (PCOS). A detailed understanding of mitophagy could reveal valuable insights and novel strategies for managing female ovarian reproductive health.


Subject(s)
Mitophagy , Ovarian Follicle , Mitophagy/physiology , Female , Ovarian Follicle/physiology , Humans , Animals , Mitochondria/physiology , Mitochondria/metabolism , Primary Ovarian Insufficiency
5.
Neurobiol Dis ; 197: 106534, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38759931

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is one of the most common neurodegenerative diseases, yet effective treatment is lacking. Moreover, the underlying pathomechanisms of ALS remain unclear, with impaired mitophagy function being increasingly recognized as a contributing factor. FUN14 domain-containing protein 1 (FUNDC1) is an autophagy receptor localized to the outer mitochondrial membrane and a mitochondrial membrane protein that mediates mitophagy and therefore considered as important factor in neurodegenerative diseases. However, its specific role in ALS is not yet clear. Therefore, this study aimed to investigate the regulatory role of FUNDC1 in ALS and determine its regulatory mechanisms. ALS transgenic mice were obtained and maintained under standard conditions. Cell lines were generated by stable transfection with hSOD1G93A or control vectors. Mice received intrathecal injections of AAV9 vectors expressing FUNDC1 or EGFP. Motor function was assessed through behavioral tests, and histological and immunostaining analyses were performed. Colocalization analysis was conducted in transfected cells, and protein expression was evaluated via western blotting. We first observed that FUNDC1 was significantly downregulated in the spinal cord tissues of SOD1G93A mice. FUNDC1 overexpression considerably improved locomotor activity and prolonged survival time in SOD1G93A mice. Mechanistically, reduced expression of FUNDC1 resulted in decreased mitophagy, as indicated by decreased recruitment through LC3 in SOD1G93A mice and cellular models. Consequently, this led to increased mitochondrial accumulation and cell apoptosis, exacerbating the ALS phenotype. Furthermore, we identified transcription factor FOXD3 as an essential upstream factor of FUNDC1, resulting in reduced transcription of FUNDC1 in ALS lesions. This study suggests a novel strategy of targeting FUNDC1-mediated mitophagy for developing therapeutic interventions to mitigate disease progression and improve outcomes for ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Mice, Transgenic , Mitochondrial Proteins , Mitophagy , Motor Neurons , Animals , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/genetics , Mitophagy/physiology , Motor Neurons/metabolism , Motor Neurons/pathology , Mice , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Spinal Cord/metabolism , Spinal Cord/pathology
6.
Exp Neurol ; 377: 114798, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670251

ABSTRACT

Mitochondrial dysfunction is closely related to brain injury and neurological dysfunction in ischemic stroke. Adenylate kinase 4 (AK4) plays a critical role in energy metabolism and mitochondrial homeostasis. However, the underlying mechanisms remain unclear. In the present study, we demonstrated an important role of AK4 in mitochondrial dysfunction in the early cerebral ischemia. Early focal cerebral ischemia induced decrease of AK4 protein expression in ischemic hemispheric brain tissue in mice. Exposure of cultured primary neuron to oxygen-glucose deprivation (OGD) also induced AK4 downregulation. Overexpression of AK4 in neuron using adeno-associated virus (AAV-AK4) in mice promoted neuronal survival reflected by decreased infarction volume and TUNEL staining. AK4 overexpression inhibited mitochondrial decline and downregulation of energy metabolism-associated proteins (p-AMPK and ATP1A3) induced by MCAO. Moreover, AK4 knock-in using lentivirus carried AK4 vector (LV-AK4) induced energy metabolism shift from glycolysis to oxidation in neuron. Using transmission electron microscope and western blot, we revealed that AK4 overexpression promoted mitophagy and mitophagy-associated proteins expression PINK1 and Parkin after MCAO. Mass spectrometry and co-immunoprecipitation revealed an interaction between AK4 and PKM2. Mechanistically, AK4 indirectly decreased PKM2 expression via enhancing its ubiquitination by increasing the interaction between PKM2 and its ubiquitin E3 ligase Parkin, and inhibits Parkin downregulation. In conclusion, our data demonstrate that AK4/ Parkin /PKM axis prevents cerebral ischemia damage via regulation of neuronal energy metabolism model and mitophagy. AK4 was a new target for intervention of early ischemic neuron injury.


Subject(s)
Adenylate Kinase , Brain Ischemia , Energy Metabolism , Mice, Inbred C57BL , Mitophagy , Neurons , Ubiquitin-Protein Ligases , Animals , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Energy Metabolism/physiology , Mice , Neurons/metabolism , Neurons/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Male , Mitophagy/physiology , Adenylate Kinase/metabolism , Thyroid Hormone-Binding Proteins , Signal Transduction/physiology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cells, Cultured , Pyruvate Kinase
7.
Zhongguo Zhong Yao Za Zhi ; 49(5): 1286-1294, 2024 Mar.
Article in Chinese | MEDLINE | ID: mdl-38621976

ABSTRACT

This study explored the specific mechanism by which tetrahydropalmatine(THP) inhibited mitophagy through the UNC-51-like kinase 1(ULK1)/FUN14 domain containing 1(FUNDC1) pathway to reduce hypoxia/reoxygenation(H/R) injury in H9c2 cells. This study used H9c2 cells as the research object to construct a cardiomyocyte H/R injury model. First, a cell viability detection kit was used to detect cell viability, and a micro-method was used to detect lactate dehydrogenase(LDH) leakage to evaluate the protective effect of THP on H/R injury of H9c2 cells. In order to evaluate the protective effect of THP on mitochondria, the chemical fluorescence method was used to detect intracellular reactive oxygen species, intramitochondrial reactive oxygen species, mitochondrial membrane potential, and autophagosomes, and the luciferin method was used to detect intracellular adenosine 5'-triphosphate(ATP) content. Western blot was further used to detect the ratio of microtubule-associated protein 1 light chain 3(LC3) membrane type(LC3-Ⅱ) and slurry type(LC3-Ⅰ) and activated cleaved caspase-3 expression level. In addition, ULK1 expression level and its phosphorylation degree at Ser555 site, as well as the FUNDC1 expression level and its phosphorylation degree of Ser17 site were detected to explore its specific mechanism. The results showed that THP effectively reduced mitochondrial damage in H9c2 cells after H/R. THP protected mitochondria by reducing the level of reactive oxygen species in cells and mitochondria, increasing mitochondrial membrane potential, thereby increasing cellular ATP production, enhancing cellular activity, reducing cellular LDH leakage, and finally alleviating H/R damage in H9c2 cells. Further studies have found that THP could reduce the production of autophagosomes, reduce the LC3-Ⅱ/LC3-Ⅰ ratio, and lower the expression of the apoptosis-related protein, namely cleaved caspase-3, indicating that THP could reduce apoptosis by inhibiting autophagy. In-depth studies have found that THP could inhibit the activation of the ULK1/FUNDC1 pathway of mitophagy and the occurrence of mitophagy by reducing the phosphorylation degree of ULK1 at Ser555 and FUNDC1 at Ser17. The application of ULK1 agonist BL-918 reversely verified the effect of THP on reducing the phosphorylation of ULK1 and FUNDC1. In summary, THP inhibited mitophagy through the ULK1/FUNDC1 pathway to reduce H/R injury in H9c2 cells.


Subject(s)
Berberine Alkaloids , Hypoxia , Mitophagy , Phenylacetates , Humans , Mitophagy/physiology , Caspase 3 , Reactive Oxygen Species/metabolism , Apoptosis , Adenosine Triphosphate/pharmacology , Autophagy-Related Protein-1 Homolog/genetics , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mitochondrial Proteins
8.
Exp Mol Med ; 56(3): 674-685, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38443598

ABSTRACT

Mitophagy induction upon mitochondrial stress is critical for maintaining mitochondrial homeostasis and cellular function. Here, we found that Mst1/2 (Stk3/4), key regulators of the Hippo pathway, are required for the induction of mitophagy under various mitochondrial stress conditions. Knockdown of Mst1/2 or pharmacological inhibition by XMU-MP-1 treatment led to impaired mitophagy induction upon CCCP and DFP treatment. Mechanistically, Mst1/2 induces mitophagy independently of the PINK1-Parkin pathway and the canonical Hippo pathway. Moreover, our results suggest the essential involvement of BNIP3 in Mst1/2-mediated mitophagy induction upon mitochondrial stress. Notably, Mst1/2 knockdown diminishes mitophagy induction, exacerbates mitochondrial dysfunction, and reduces cellular survival upon neurotoxic stress in both SH-SY5Y cells and Drosophila models. Conversely, Mst1 and Mst2 expression enhances mitophagy induction and cell survival. In addition, AAV-mediated Mst1 expression reduced the loss of TH-positive neurons, ameliorated behavioral deficits, and improved mitochondrial function in an MPTP-induced Parkinson's disease mouse model. Our findings reveal the Mst1/2-BNIP3 regulatory axis as a novel mediator of mitophagy induction under conditions of mitochondrial stress and suggest that Mst1/2 play a pivotal role in maintaining mitochondrial function and neuronal viability in response to neurotoxic treatment.


Subject(s)
Mitophagy , Neuroblastoma , Protein Serine-Threonine Kinases , Serine-Threonine Kinase 3 , Animals , Humans , Mice , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitophagy/genetics , Mitophagy/physiology , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Serine-Threonine Kinase 3/genetics , Serine-Threonine Kinase 3/metabolism , Drosophila/genetics
9.
Int J Surg ; 110(5): 2649-2668, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38445453

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a leading cause of mortality in patients with sepsis due to proinflammatory endothelial changes and endothelial permeability defects. Mitochondrial dysfunction is recognized as a critical mediator in the pathogenesis of sepsis-induced ALI. Although mitophagy regulation of mitochondrial quality is well recognized, little is known about its role in lung ECs during sepsis-induced ALI. Sirtuin 1 (SIRT1) is a histone protein deacetylase involved in inflammation, mitophagy, and cellular senescence. Here, the authors show a type of late endosome-dependent mitophagy that inhibits NLRP3 and STING activation through SIRT1 signaling during sepsis-induced ALI. METHODS: C57BL/6J male mice with or without administration of the SIRT1 inhibitor EX527 in the CLP model and lung ECs in vitro were developed to identify mitophagy mechanisms that underlie the cross-talk between SIRT1 signaling and sepsis-induced ALI. RESULTS: SIRT1 deficient mice exhibited exacerbated sepsis-induced ALI. Knockdown of SIRT1 interfered with mitophagy through late endosome Rab7, leading to the accumulation of damaged mitochondria and inducing excessive mitochondrial reactive oxygen species (mtROS) generation and cytosolic release of mitochondrial DNA (mtDNA), which triggered NLRP3 inflammasome and the cytosolic nucleotide sensing pathways (STING) over-activation. Pharmacological inhibition of STING and NLRP3 i n vivo or genetic knockdown in vitro reversed SIRT1 deficiency mediated endothelial permeability defects and endothelial inflammation in sepsis-induced ALI. Moreover, activation of SIRT1 with SRT1720 in vivo or overexpression of SIRT1 in vitro protected against sepsis-induced ALI. CONCLUSION: These findings suggest that SIRT1 signaling is essential for restricting STING and NLRP3 hyperactivation by promoting endosomal-mediated mitophagy in lung ECs, providing potential therapeutic targets for treating sepsis-induced ALI.


Subject(s)
Acute Lung Injury , Membrane Proteins , Mice, Inbred C57BL , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Sirtuin 1/antagonists & inhibitors , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sepsis/metabolism , Sepsis/complications , Mitophagy/physiology , Male , Mice , Membrane Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Signal Transduction/physiology , Endosomes/metabolism , Disease Models, Animal
10.
Autophagy ; 20(5): 983-984, 2024 May.
Article in English | MEDLINE | ID: mdl-38456640

ABSTRACT

The selective clearance of unwanted, damaged or dangerous components by macroautophagy/autophagy is critical for maintaining cellular homeostasis in organisms from yeast to humans. In recent years, significant progress has been made in understanding how phagophores selectively sequester specific cargo. Nevertheless, a fundamental question remains: Can distinct selective autophagy programs simultaneously operate within the same cell? A recent study from the Baehrecke lab has unveiled a developmentally programmed Pink1-dependent reticulophagy process in the Drosophila intestine. Furthermore, this study demonstrated that autophagy differentially clears mitochondria and ER in the same cell under the regulation of Pink1 through different E3 ubiquitin ligases, highlighting the need for further exploration in understanding the complexity of autophagic substrate selection and crosstalk between diverse autophagy programs.


Subject(s)
Mitophagy , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Mitophagy/physiology , Animals , Humans , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Autophagy/physiology , Protein Serine-Threonine Kinases/metabolism , Mitochondria/metabolism , Drosophila melanogaster/metabolism
11.
Domest Anim Endocrinol ; 88: 106847, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38479188

ABSTRACT

Cold exposure is a common stressor for newborn goats. Skeletal muscle plays an important role in maintaining whole-body homeostasis of glucose and lipid metabolism. However, the molecular mechanisms underlying regulation of skeletal muscle of newborn goats by cold exposure remains unclear. In this study, we found a significant increase (P < 0.01) in serum glucagon levels after 24 h of cold exposure (COLD, 6°C), while glucose and insulin concentrations were significantly decreased (P < 0.01) compared to room temperature (RT, 25°C). Additionally, we found that cold exposure reduced glycogen content (P < 0.01) in skeletal muscle. Pathway enrichment analysis revealed that cold exposure activated skeletal muscle glucose metabolism pathways (including insulin resistance and the insulin signaling pathway) and mitophagy-related pathways. Cold exposure up-regulated the expression of genes involved in fatty acid and triglyceride synthesis, promoting skeletal muscle lipid deposition. Notably, cold exposure induced mitophagy in skeletal muscle.


Subject(s)
Animals, Newborn , Cold Temperature , Glucose , Goats , Mitophagy , Muscle, Skeletal , Animals , Goats/physiology , Muscle, Skeletal/metabolism , Mitophagy/physiology , Glucose/metabolism , Lipid Metabolism , Lipid Droplets/metabolism
12.
Biol Res ; 57(1): 10, 2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38494498

ABSTRACT

BACKGROUND: The senescence of renal tubular epithelial cells (RTECs) is crucial in the progression of diabetic kidney disease (DKD). Accumulating evidence suggests a close association between insufficient mitophagy and RTEC senescence. Yeast mitochondrial escape 1-like 1 (YME1L), an inner mitochondrial membrane metalloprotease, maintains mitochondrial integrity. Its functions in DKD remain unclear. Here, we investigated whether YME1L can prevent the progression of DKD by regulating mitophagy and cellular senescence. METHODS: We analyzed YME1L expression in renal tubules of DKD patients and mice, explored transcriptomic changes associated with YME1L overexpression in RTECs, and assessed its impact on RTEC senescence and renal dysfunction using an HFD/STZ-induced DKD mouse model. Tubule-specific overexpression of YME1L was achieved through the use of recombinant adeno-associated virus 2/9 (rAAV 2/9). We conducted both in vivo and in vitro experiments to evaluate the effects of YME1L overexpression on mitophagy and mitochondrial function. Furthermore, we performed LC-MS/MS analysis to identify potential protein interactions involving YME1L and elucidate the underlying mechanisms. RESULTS: Our findings revealed a significant decrease in YME1L expression in the renal tubules of DKD patients and mice. However, tubule-specific overexpression of YME1L significantly alleviated RTEC senescence and renal dysfunction in the HFD/STZ-induced DKD mouse model. Moreover, YME1L overexpression exhibited positive effects on enhancing mitophagy and improving mitochondrial function both in vivo and in vitro. Mechanistically, our LC-MS/MS analysis uncovered a crucial mitophagy receptor, BCL2-like 13 (BCL2L13), as an interacting partner of YME1L. Furthermore, YME1L was found to promote the phosphorylation of BCL2L13, highlighting its role in regulating mitophagy. CONCLUSIONS: This study provides compelling evidence that YME1L plays a critical role in protecting RTECs from cellular senescence and impeding the progression of DKD. Overexpression of YME1L demonstrated significant therapeutic potential by ameliorating both RTEC senescence and renal dysfunction in the DKD mice. Moreover, our findings indicate that YME1L enhances mitophagy and improves mitochondrial function, potentially through its interaction with BCL2L13 and subsequent phosphorylation. These novel insights into the protective mechanisms of YME1L offer a promising strategy for developing therapies targeting DKD.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Humans , Mice , Animals , Mitophagy/physiology , Saccharomyces cerevisiae , Chromatography, Liquid , Tandem Mass Spectrometry , Epithelial Cells/metabolism , Disease Models, Animal , Cellular Senescence , Diabetes Mellitus/metabolism , Metalloendopeptidases/metabolism , Metalloendopeptidases/pharmacology
13.
Cells ; 13(5)2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38474413

ABSTRACT

Cardiomyocytes rely on proper mitochondrial homeostasis to maintain contractility and achieve optimal cardiac performance. Mitochondrial homeostasis is controlled by mitochondrial fission, fusion, and mitochondrial autophagy (mitophagy). Mitophagy plays a particularly important role in promoting the degradation of dysfunctional mitochondria in terminally differentiated cells. However, the precise mechanisms by which this is achieved in cardiomyocytes remain opaque. Our study identifies GRAF1 as an important mediator in PINK1-Parkin pathway-dependent mitophagy. Depletion of GRAF1 (Arhgap26) in cardiomyocytes results in actin remodeling defects, suboptimal mitochondria clustering, and clearance. Mechanistically, GRAF1 promotes Parkin-LC3 complex formation and directs autophagosomes to damaged mitochondria. Herein, we found that these functions are regulated, at least in part, by the direct binding of GRAF1 to phosphoinositides (PI(3)P, PI(4)P, and PI(5)P) on autophagosomes. In addition, PINK1-dependent phosphorylation of Parkin promotes Parkin-GRAF1-LC3 complex formation, and PINK1-dependent phosphorylation of GRAF1 (on S668 and S671) facilitates the clustering and clearance of mitochondria. Herein, we developed new phosphor-specific antibodies to these sites and showed that these post-translational modifications are differentially modified in human hypertrophic cardiomyopathy and dilated cardiomyopathy. Furthermore, our metabolic studies using serum collected from isoproterenol-treated WT and GRAF1CKO mice revealed defects in mitophagy-dependent cardiomyocyte fuel flexibility that have widespread impacts on systemic metabolism. In summary, our study reveals that GRAF1 co-regulates actin and membrane dynamics to promote cardiomyocyte mitophagy and that dysregulation of GRAF1 post-translational modifications may underlie cardiac disease pathogenesis.


Subject(s)
GTPase-Activating Proteins , Mitophagy , Myocytes, Cardiac , Phosphatidylinositol Phosphates , Ubiquitin-Protein Ligases , Animals , Humans , Mice , Actins , GTPase-Activating Proteins/metabolism , Mitophagy/physiology , Myocytes, Cardiac/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
14.
Exp Mol Med ; 56(3): 747-759, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38531963

ABSTRACT

Intervertebral disc degeneration (IDD) is an important pathological basis for degenerative spinal diseases and is involved in mitophagy dysfunction. However, the molecular mechanisms underlying mitophagy regulation in IDD remain unclear. This study aimed to clarify the role of DJ-1 in regulating mitophagy during IDD pathogenesis. Here, we showed that the mitochondrial localization of DJ-1 in nucleus pulposus cells (NPCs) first increased and then decreased in response to oxidative stress. Subsequently, loss- and gain-of-function experiments revealed that overexpression of DJ-1 in NPCs inhibited oxidative stress-induced mitochondrial dysfunction and mitochondria-dependent apoptosis, whereas knockdown of DJ-1 had the opposite effect. Mechanistically, mitochondrial translocation of DJ-1 promoted the recruitment of hexokinase 2 (HK2) to damaged mitochondria by activating Akt and subsequently Parkin-dependent mitophagy to inhibit oxidative stress-induced apoptosis in NPCs. However, silencing Parkin, reducing mitochondrial recruitment of HK2, or inhibiting Akt activation suppressed DJ-1-mediated mitophagy. Furthermore, overexpression of DJ-1 ameliorated IDD in rats through HK2-mediated mitophagy. Taken together, these findings indicate that DJ-1 promotes HK2-mediated mitophagy under oxidative stress conditions to inhibit mitochondria-dependent apoptosis in NPCs and could be a therapeutic target for IDD.


Subject(s)
Intervertebral Disc Degeneration , Mitophagy , Protein Deglycase DJ-1 , Animals , Rats , Apoptosis , Hexokinase/genetics , Hexokinase/pharmacology , Hexokinase/therapeutic use , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Mitophagy/genetics , Mitophagy/physiology , Proto-Oncogene Proteins c-akt , Ubiquitin-Protein Ligases/genetics , Protein Deglycase DJ-1/metabolism
15.
Mol Cell ; 84(6): 1090-1100.e6, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38340717

ABSTRACT

To maintain mitochondrial homeostasis, damaged or excessive mitochondria are culled in coordination with the physiological state of the cell. The integrated stress response (ISR) is a signaling network that recognizes diverse cellular stresses, including mitochondrial dysfunction. Because the four ISR branches converge to common outputs, it is unclear whether mitochondrial stress detected by this network can regulate mitophagy, the autophagic degradation of mitochondria. Using a whole-genome screen, we show that the heme-regulated inhibitor (HRI) branch of the ISR selectively induces mitophagy. Activation of the HRI branch results in mitochondrial localization of phosphorylated eukaryotic initiation factor 2, which we show is sufficient to induce mitophagy. The HRI mitophagy pathway operates in parallel with the mitophagy pathway controlled by the Parkinson's disease related genes PINK1 and PARKIN and is mechanistically distinct. Therefore, HRI repurposes machinery that is normally used for translational initiation to trigger mitophagy in response to mitochondrial damage.


Subject(s)
Mitophagy , Protein Kinases , Mitophagy/physiology , Protein Kinases/genetics , Protein Kinases/metabolism , Autophagy/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Protein Processing, Post-Translational , Signal Transduction
16.
Autophagy ; 20(5): 985-993, 2024 May.
Article in English | MEDLINE | ID: mdl-38361280

ABSTRACT

Mitophagy is the process of selective autophagy that removes superfluous and dysfunctional mitochondria. Mitophagy was first characterized in mammalian cells and is now recognized to follow several pathways including basal forms in specific organs. Mitophagy pathways are regulated by multiple, often interconnected factors. The present review aims to streamline this complexity and evaluate common elements that may define the evolutionary origin of mitophagy. Key issues surrounding mitophagy signaling at the mitochondrial surface may fundamentally derive from mitochondrial membrane dynamics. Elements of such membrane dynamics likely originated during the endosymbiosis of the alphaproteobacterial ancestor of our mitochondria but underwent an evolutionary leap forward in basal metazoa that determined the currently known variations in mitophagy signaling.Abbreviations: AGPAT, 1-acylglycerol-3-phosphate O-acyltransferase; ATG, autophagy related; BCL2L13, BCL2 like 13; BNIP3, BCL2 interacting protein 3; BNIP3L, BCL2 interacting protein 3 like; CALCOCO, calcium binding and coiled-coil domain; CL, cardiolipin; ER, endoplasmic reticulum; ERMES, ER-mitochondria encounter structure; FBXL4, F-box and leucine rich repeat protein 4; FUNDC1, FUN14 domain containing 1; GABARAPL1, GABA type A receptor associated protein like 1; HIF, hypoxia inducible factor; IMM, inner mitochondrial membrane; LBPA/BMP, lysobisphosphatidic acid; LIR, LC3-interacting region; LPA, lysophosphatidic acid; MAM, mitochondria-associated membranes; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; MCL, monolysocardiolipin; ML, maximum likelihood; NBR1, NBR1 autophagy cargo receptor; OMM, outer mitochondrial membrane; PA, phosphatidic acid; PACS2, phosphofurin acidic cluster sorting protein 2; PC/PLC, phosphatidylcholine; PE, phosphatidylethanolamine; PHB2, prohibitin 2; PINK1, PTEN induced kinase 1; PtdIns, phosphatidylinositol; SAR, Stramenopiles, Apicomplexa and Rhizaria; TAX1BP1, Tax1 binding protein 1; ULK1, unc-51 like autophagy activating kinase 1; VDAC/porin, voltage dependent anion channel.


Subject(s)
Mitochondria , Mitophagy , Mitophagy/physiology , Mitochondria/metabolism , Humans , Animals , Prohibitins , Mitochondrial Membranes/metabolism , Signal Transduction
17.
J Obstet Gynaecol Res ; 50(5): 775-792, 2024 May.
Article in English | MEDLINE | ID: mdl-38417972

ABSTRACT

AIM: Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by menstrual irregularities, androgen excess, and polycystic ovarian morphology, but its pathogenesis remains largely unknown. This review focuses on how androgen excess influences the molecular basis of energy metabolism, mitochondrial function, and mitophagy in granulosa cells and oocytes, summarizes our current understanding of the pathogenesis of PCOS, and discuss perspectives on future research directions. METHODS: A search of PubMed and Google Scholar databases were used to identify relevant studies for this narrative literature review. RESULTS: Female offspring born of pregnant animals exposed to androgens recapitulates the PCOS phenotype. Abnormal mitochondrial morphology, altered expression of genes related to glycolysis, mitochondrial biogenesis, fission/fusion dynamics, and mitophagy have been identified in PCOS patients and androgenic animal models. Androgen excess causes uncoupling of the electron transport chain and depletion of the cellular adenosine 5'-triphosphate pool, indicating further impairment of mitochondrial function. A shift toward mitochondrial fission restores mitochondrial quality control mechanisms. However, prolonged mitochondrial fission disrupts autophagy/mitophagy induction due to loss of compensatory reserve for mitochondrial biogenesis. Disruption of compensatory mechanisms that mediate the quality control switch from mitophagy to apoptosis may cause a disease phenotype. Furthermore, genetic predisposition, altered expression of genes related to glycolysis and oxidative phosphorylation, or a combination of these factors may also contribute to the development of PCOS. CONCLUSION: In conclusion, fetuses exposed to a hyperandrogenemic intrauterine environment may cause the PCOS phenotype possibly through disruption of the compensatory regulation of the mitophagy-apoptosis axis.


Subject(s)
Apoptosis , Mitophagy , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/metabolism , Female , Humans , Mitophagy/physiology , Apoptosis/physiology , Animals , Mitochondria/metabolism
18.
Zhongguo Zhong Yao Za Zhi ; 49(1): 46-54, 2024 Jan.
Article in Chinese | MEDLINE | ID: mdl-38403337

ABSTRACT

Diabetes mellitus(DM) is a chronic endocrine disease characterized by hyperglycemia caused by carbohydrate or lipid metabolism disorders or insulin dysfunction. Hyperglycemia and long-term metabolic disorders in DM can damage tissues and organs throughout the body, leading to serious complications. Mitochondrial autophagy(mitophagy) is an important mitochondrial quality control process in cells and a special autophagy phenomenon, in which damaged or redundant mitochondria can be selectively removed by autophagic lysosome, which is crucial to maintain cell stability and survival under stress. Studies have confirmed that changes in autophagy play a role in the development and control of DM and its complications. Mitophagy has become a research hotspot in recent years and it is closely associated with the pathogenesis of a variety of diseases. Substantial evidence suggests that mitophagy plays a crucial role in regulating the metabolic homeostasis in the case of DM and its complications. Because the destructive great vessel complications and microvascular complications cause increased mortality, blindness, renal failure, and declined quality of life of DM patients, it is urgent to develop targeted therapies to intervene in DM and its complications. Traditional Chinese medicine(TCM), with a multi-component, multi-target, and multi-level action manner, can prevent the development of drug resistance and have significant therapeutic effects in the prevention and treatment of DM and its complications. Therefore, exploring the mechanisms of TCM in regulating mito-phagy may become a new method for treating DM and its complications. With focus on the roles and mechanisms of mitophagy in DM and its complications, this paper summarizes and prospects the research on the treatment of DM and its complications with TCM via re-gulating mitophagy, aiming to provide new ideas for the clinical practice.


Subject(s)
Diabetes Mellitus , Hyperglycemia , Humans , Mitophagy/physiology , Medicine, Chinese Traditional , Quality of Life , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics
19.
Int J Biol Macromol ; 262(Pt 1): 129950, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38320636

ABSTRACT

Intervertebral disc degeneration (IVDD) contributes largely to low back pain. Recent studies have highlighted the exacerbating role of diabetes mellitus (DM) in IVDD, mainly due to the influence of hyperglycemia (HG) or the accumulation of advanced glycation end products (AGEs). Vascular endothelial growth factor A (VEGFA) newly assumed a distinct impact in nonvascular tissues through mitophagy regulation. However, the combined actions of HG and AGEs on IVDD and the involved role of VEGFA remain unclear. We confirmed the potential relation between VEGFA and DM through bioinformatics and biological specimen detection. Then we observed that AGEs induced nucleus pulposus (NP) cell degeneration by upregulating cellular reactive oxygen species (ROS), and HG further aggravated ROS level through breaking AGEs-induced protective mitophagy. Furthermore, this adverse effect could be strengthened by VEGFA knockdown. Importantly, we identified that the regulation of VEGFA and mitophagy were vital mechanisms in AGEs-HG-induced NP cell degeneration through Parkin/Akt/mTOR and AMPK/mTOR pathway. Additionally, VEGFA overexpression through local injection with lentivirus carrying VEGFA plasmids significantly alleviated NP degeneration and IVDD in STZ-induced diabetes and puncture rat models. In conclusion, the findings first confirmed that VEGFA protects against AGEs-HG-induced IVDD, which may represent a therapeutic strategy for DM-related IVDD.


Subject(s)
Intervertebral Disc Degeneration , Nucleus Pulposus , Rats , Animals , Down-Regulation , Nucleus Pulposus/metabolism , Mitophagy/physiology , Reactive Oxygen Species/metabolism , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , TOR Serine-Threonine Kinases/metabolism , Glucose/metabolism , Apoptosis
20.
Crit Rev Immunol ; 44(2): 25-33, 2024.
Article in English | MEDLINE | ID: mdl-38305334

ABSTRACT

Platelet hyperactivity represents a deleterious physiological phenomenon in diabetes mellitus (DM). This study aimed to explore the role of FUN14 domain containing 1 (FUNDC1) in platelet activation within the context of DM and to uncover relevant mechanisms, with a focus on mitophagy. A mouse model of DM was established by high-fat feeding and streptozotocin injection. Platelets isolated from whole blood were exposed to carbonyl cyanide-4-(trifluo-romethoxy)phenylhydrazone (FCCP) to induce mitophagy. The relative mRNA expression of FUNDC1 was detected by quantitative real-time PCR (qRT-PCR). Western blotting was employed to measure the protein levels of FUNDC1, the ratio of LC3-II toLC3-I, and cleaved caspase-3. Immunofluorescence and flow cytometry were performed to assess LC3-positive mitochondria and platelet activation factor CD62P, respectively. Additionally, serum levels of ß-thrombo-globulin (ß-TG) and platelet factor 4 (PF4)were measured by enzyme-linked immunosorbent assay. FUNDC1 expression was elevated in DM mice, and its silencing decreased the body weight and fasting blood glucose. Inhibition of FUNDC1 also significantly attenuated FCCP-induced platelet mitophagy, as evidenced by the down-regulation of the LC3-II/LC3-I ratio, up-regulation of Tomm20, and diminished presence of LC3-positive mitochondria. Moreover, platelet activation was noted in DM mice; this activation was mitigated upon FUNDC1 silencing, which was confirmed by the down-regulation of cleaved caspase-3 and CD62P as well as reductions in ß-TG and PF4 serum levels. Silencing of FUNDC1 inhibited platelet hyperactivity in DM by impeding mitophagy. As such, FUNDC1-midiated mitophagy may be a promising target for the treatment of DM and its associated cardiovascular complications related cardiovascular events.


Subject(s)
Diabetes Mellitus , Membrane Proteins , Mitochondrial Proteins , Mitophagy , Animals , Mice , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone , Caspase 3 , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Platelet Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...