Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 790
Filtrar
1.
Redox Rep ; 29(1): 2377870, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39010730

RESUMO

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.


Assuntos
Mitofagia , Estresse Oxidativo , Síndrome do Ovário Policístico , Humanos , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Feminino , Mitofagia/efeitos dos fármacos , Mitofagia/fisiologia , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Adulto , Microambiente Celular/fisiologia
2.
Front Endocrinol (Lausanne) ; 15: 1417007, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38952389

RESUMO

Ovarian aging is a complex process characterized by a decline in oocyte quantity and quality, directly impacting fertility and overall well-being. Recent researches have identified mitochondria as pivotal players in the aging of ovaries, influencing various hallmarks and pathways governing this intricate process. In this review, we discuss the multifaceted role of mitochondria in determining ovarian fate, and outline the pivotal mechanisms through which mitochondria contribute to ovarian aging. Specifically, we emphasize the potential of targeting mitochondrial dysfunction through innovative therapeutic approaches, including antioxidants, metabolic improvement, biogenesis promotion, mitophagy enhancement, mitochondrial transfer, and traditional Chinese medicine. These strategies hold promise as effective means to mitigate age-related fertility decline and preserve ovarian health. Drawing insights from advanced researches in the field, this review provides a deeper understanding of the intricate interplay between mitochondrial function and ovarian aging, offering valuable perspectives for the development of novel therapeutic interventions aimed at preserving fertility and enhancing overall reproductive health.


Assuntos
Envelhecimento , Mitocôndrias , Ovário , Humanos , Feminino , Mitocôndrias/metabolismo , Envelhecimento/fisiologia , Envelhecimento/metabolismo , Ovário/metabolismo , Ovário/fisiologia , Animais , Antioxidantes/uso terapêutico , Oócitos/metabolismo , Oócitos/fisiologia , Mitofagia/fisiologia
3.
Acta Neuropathol Commun ; 12(1): 90, 2024 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-38851733

RESUMO

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.


Assuntos
Doença de Alzheimer , Fibroblastos , Mitocôndrias , Mitofagia , Humanos , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Fibroblastos/patologia , Fibroblastos/metabolismo , Idoso , Feminino , Mitocôndrias/patologia , Mitocôndrias/metabolismo , Masculino , Mitofagia/fisiologia , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Disfunção Cognitiva/patologia , Disfunção Cognitiva/metabolismo , Autofagia/fisiologia
4.
Mol Neurodegener ; 19(1): 49, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890703

RESUMO

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.


Assuntos
Cumarínicos , Animais , Camundongos , Cumarínicos/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Retina/metabolismo , Retina/efeitos dos fármacos , Retina/patologia , Mitofagia/efeitos dos fármacos , Mitofagia/fisiologia , Proteína Sequestossoma-1/metabolismo , Lisossomos/metabolismo , Lisossomos/efeitos dos fármacos , Humanos , Modelos Animais de Doenças , Fármacos Neuroprotetores/farmacologia , Camundongos Endogâmicos C57BL , Iodatos/toxicidade
5.
Arch Gerontol Geriatr ; 125: 105522, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38861889

RESUMO

Osteoarthritis (OA) is a prevalent chronic joint disease characterized by articular cartilage degeneration, pain, and disability. Emerging evidence indicates that mitochondrial quality control dysfunction contributes to OA pathogenesis. Mitochondria are essential organelles to generate cellular energy via oxidative phosphorylation and regulate vital processes. Impaired mitochondria can negatively impact cellular metabolism and result in the generation of harmful reactive oxygen species (ROS). Dysfunction in mitochondrial quality control mechanisms has been increasingly linked to OA onset and progression. This review summarizes current knowledge on the role of mitochondrial quality control disruption in OA, highlighting disturbed mitochondrial dynamics, impaired mitochondrial biogenesis, antioxidant defenses and mitophagy. The review also discusses potential therapeutic strategies targeting mitochondrial Quality Control in OA, offering future perspectives on advancing OA therapeutic strategies.


Assuntos
Mitocôndrias , Mitofagia , Osteoartrite , Espécies Reativas de Oxigênio , Humanos , Osteoartrite/metabolismo , Osteoartrite/terapia , Mitocôndrias/metabolismo , Mitofagia/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo/fisiologia , Dinâmica Mitocondrial/fisiologia , Antioxidantes/uso terapêutico
6.
J Alzheimers Dis ; 100(1): 279-296, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38848175

RESUMO

Background: Mitochondrial dysfunction exists in Alzheimer's disease (AD) brain, and damaged mitochondria need to be removed by mitophagy. Small GTPase Rab7 regulates the fusion of mitochondria and lysosome, while TBC1D5 inhibits Rab7 activation. However, it is not clear whether the regulation of Rab7 activity by TBC1D5 can improve mitophagy and inhibit AD progression. Objective: To investigate the role of TBC1D5 in mitophagy and its regulatory mechanism for Rab7, and whether activation of mitophagy can inhibit the progression of AD. Methods: Mitophagy was determined by western blot and immunofluorescence. The morphology and quantity of mitochondria were tracked by TEM. pCMV-Mito-AT1.03 was employed to detect the cellular ATP. Amyloid-ß secreted by AD cells was detected by ELISA. Co-immunoprecipitation was used to investigate the binding partner of the target protein. Golgi-cox staining was applied to observe neuronal morphology of mice. The Morris water maze test and Y-maze were performed to assess spatial learning and memory, and the open field test was measured to evaluate motor function and anxiety-like phenotype of experimental animals. Results: Mitochondrial morphology was impaired in AD models, and TBC1D5 was highly expressed. Knocking down TBC1D5 increased the expression of active Rab7, promoted the fusion of lysosome and autophagosome, thus improving mitophagy, and improved the morphology of hippocampal neurons and the impaired behavior in AD mice. Conclusions: Knocking down TBC1D5 increased Rab7 activity and promoted the fusion of autophagosome and lysosome. Our study provided insights into the mechanisms that bring new possibilities for AD therapy targeting mitophagy.


Assuntos
Doença de Alzheimer , Modelos Animais de Doenças , Proteínas Ativadoras de GTPase , Mitocôndrias , Mitofagia , Proteínas rab de Ligação ao GTP , proteínas de unión al GTP Rab7 , Animais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Mitofagia/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/genética , Camundongos , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Ativadoras de GTPase/genética , Humanos , Mitocôndrias/metabolismo , Masculino , Camundongos Transgênicos , Peptídeos beta-Amiloides/metabolismo , Neurônios/metabolismo , Neurônios/patologia
7.
CNS Neurosci Ther ; 30(6): e14800, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38887162

RESUMO

BACKGROUND: Impaired mitochondrial dynamics have been identified as a significant contributing factor to reduced neurogenesis under pathological conditions. However, the relationship among mitochondrial dynamics, neurogenesis, and spatial memory during normal development remains unclear. This study aims to elucidate the role of mitophagy in spatial memory mediated by neurogenesis during development. METHODS: Adolescent and adult male mice were used to assess spatial memory performance. Immunofluorescence staining was employed to evaluate levels of neurogenesis, and mitochondrial dynamics were assessed through western blotting and transmission electron microscopy. Pharmacological interventions further validated the causal relationship among mitophagy, neurogenesis, and behavioral performance during development. RESULTS: The study revealed differences in spatial memory between adolescent and adult mice. Diminished neurogenesis, accompanied by reduced mitophagy, was observed in the hippocampus of adult mice compared to adolescent subjects. Pharmacological induction of mitophagy in adult mice with UMI-77 resulted in enhanced neurogenesis and prolonged spatial memory retention. Conversely, inhibition of mitophagy with Mdivi-1 in adolescent mice led to reduced hippocampal neurogenesis and impaired spatial memory. CONCLUSION: The observed decline in spatial memory in adult mice is associated with decreased mitophagy, which affects neurogenesis in the dentate gyrus. This underscores the therapeutic potential of enhancing mitophagy to counteract age- or disease-related cognitive decline.


Assuntos
Hipocampo , Mitofagia , Neurogênese , Memória Espacial , Animais , Neurogênese/fisiologia , Neurogênese/efeitos dos fármacos , Mitofagia/fisiologia , Mitofagia/efeitos dos fármacos , Memória Espacial/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dinâmica Mitocondrial/fisiologia , Quinazolinonas
8.
Physiol Res ; 73(2): 253-263, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38710055

RESUMO

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.


Assuntos
Mitofagia , Hepatopatia Gordurosa não Alcoólica , Proteínas Quinases , Ubiquitina-Proteína Ligases , Animais , Humanos , Masculino , Camundongos , Mitofagia/fisiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/genética , Proteínas Quinases/metabolismo , Proteínas Quinases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética
9.
J Alzheimers Dis ; 99(4): 1375-1383, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38759019

RESUMO

Background: Currently, no evidence exists on the expression of apoptosis (CASP3), autophagy (BECN1), and mitophagy (BNIP3) genes in the CA3 area after ischemia with long-term survival. Objective: The goal of the paper was to study changes in above genes expression in CA3 area after ischemia in the period of 6-24 months. Methods: In this study, using quantitative RT-PCR, we present the expression of genes associated with neuronal death in a rat ischemic model of Alzheimer's disease. Results: First time, we demonstrated overexpression of the CASP3 gene in CA3 area after ischemia with survival ranging from 0.5 to 2 years. Overexpression of the CASP3 gene was accompanied by a decrease in the activity level of the BECN1 and BNIP3 genes over a period of 0.5 year. Then, during 1-2 years, BNIP3 gene expression increased significantly and coincided with an increase in CASP3 gene expression. However, BECN1 gene expression was variable, increased significantly at 1 and 2 years and was below control values 1.5 years post-ischemia. Conclusions: Our observations suggest that ischemia with long-term survival induces neuronal death in CA3 through activation of caspase 3 in cooperation with the pro-apoptotic gene BNIP3. This study also suggests that the BNIP3 gene regulates caspase-independent pyramidal neuronal death post-ischemia. Thus, caspase-dependent and -independent death of neuronal cells occur post-ischemia in the CA3 area. Our data suggest new role of the BNIP3 gene in the regulation of post-ischemic neuronal death in CA3. This suggests the involvement of the BNIP3 together with the CASP3 in the CA3 in neuronal death post-ischemia.


Assuntos
Doença de Alzheimer , Apoptose , Autofagia , Proteína Beclina-1 , Caspase 3 , Modelos Animais de Doenças , Proteínas de Membrana , Mitofagia , Animais , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitofagia/genética , Mitofagia/fisiologia , Autofagia/genética , Autofagia/fisiologia , Apoptose/genética , Masculino , Caspase 3/metabolismo , Caspase 3/genética , Ratos , Região CA3 Hipocampal/patologia , Região CA3 Hipocampal/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Ratos Wistar
10.
Reprod Biol ; 24(2): 100889, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38733657

RESUMO

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.


Assuntos
Mitofagia , Folículo Ovariano , Mitofagia/fisiologia , Feminino , Folículo Ovariano/fisiologia , Humanos , Animais , Mitocôndrias/fisiologia , Mitocôndrias/metabolismo , Insuficiência Ovariana Primária
11.
Neurobiol Dis ; 197: 106534, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38759931

RESUMO

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.


Assuntos
Esclerose Lateral Amiotrófica , Modelos Animais de Doenças , Camundongos Transgênicos , Proteínas Mitocondriais , Mitofagia , Neurônios Motores , Animais , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/genética , Mitofagia/fisiologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Camundongos , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Humanos , Medula Espinal/metabolismo , Medula Espinal/patologia
12.
Mil Med Res ; 11(1): 32, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38812059

RESUMO

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.


Assuntos
Mitocôndrias , Mitofagia , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Mitofagia/fisiologia , Mitofagia/efeitos dos fármacos , Dinâmica Mitocondrial/fisiologia
13.
Zhongguo Zhong Yao Za Zhi ; 49(5): 1286-1294, 2024 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-38621976

RESUMO

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.


Assuntos
Alcaloides de Berberina , Hipóxia , Mitofagia , Fenilacetatos , Humanos , Mitofagia/fisiologia , Caspase 3 , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Trifosfato de Adenosina/farmacologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais
14.
Exp Neurol ; 377: 114798, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38670251

RESUMO

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.


Assuntos
Adenilato Quinase , Isquemia Encefálica , Metabolismo Energético , Camundongos Endogâmicos C57BL , Mitofagia , Neurônios , Ubiquitina-Proteína Ligases , Animais , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Metabolismo Energético/fisiologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Masculino , Mitofagia/fisiologia , Adenilato Quinase/metabolismo , Proteínas de Ligação a Hormônio da Tireoide , Transdução de Sinais/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Células Cultivadas , Piruvato Quinase
15.
Curr Alzheimer Res ; 20(12): 827-844, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38482617

RESUMO

The prevalence of Alzheimer's disease (AD) is increasing as the elderly population, which hurts elderly people's cognition and capacity for self-care. The process of mitophagy involves the selective clearance of ageing and impaired mitochondria, which is required to preserve intracellular homeostasis and energy metabolism. Currently, it has been discovered that mitophagy abnormalities are intimately linked to the beginning and progression of AD. This article discusses the mechanism of mitophagy, abnormal mitophagy, and therapeutic effects in AD. The purpose is to offer fresh perspectives on the causes and remedies of AD.


Assuntos
Doença de Alzheimer , Mitofagia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Humanos , Mitofagia/fisiologia , Animais , Mitocôndrias/metabolismo , Mitocôndrias/patologia
16.
Int J Surg ; 110(5): 2649-2668, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38445453

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Mitofagia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Sepse , Sirtuína 1 , Animais , Sirtuína 1/metabolismo , Sirtuína 1/antagonistas & inibidores , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/etiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Sepse/metabolismo , Sepse/complicações , Mitofagia/fisiologia , Masculino , Camundongos , Proteínas de Membrana/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Transdução de Sinais/fisiologia , Endossomos/metabolismo , Modelos Animais de Doenças
17.
Eur J Clin Invest ; 54(8): e14199, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38530070

RESUMO

BACKGROUND: Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS: In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS: Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS: Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.


Assuntos
Doenças Cardiovasculares , Mitofagia , Humanos , Mitofagia/fisiologia , Aterosclerose , Insuficiência Cardíaca/fisiopatologia , Animais , Traumatismo por Reperfusão Miocárdica , Cardiomiopatias/fisiopatologia , Mitocôndrias Cardíacas/metabolismo
18.
Autophagy ; 20(6): 1447-1448, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38449384

RESUMO

Mitophagy is a cellular process that enables the selective degradation of damaged, dysfunctional, or superfluous mitochondria. During mitophagy, specific proteins recognize and tag mitochondria for degradation. These tagged mitochondria are engulfed by specialized structures called phagophores that then mature into autophagosomes/mitophagosomes. Mitophagosomes subsequently transport their mitochondrial cargo to lysosomes, where the mitochondria are broken down and recycled. While the PINK1-PRKN-dependent mitophagy pathway is well understood, mitophagy can also occur independently of this pathway. BNIP3 and BNIP3L/NIX, paralogous membrane proteins on the outer mitochondrial membrane (OMM), serve as ubiquitin-independent mitophagy receptors. Historically, BNIP3 regulation was thought to be primarily transcriptional through HIF1A (hypoxia inducible factor 1 subunit alpha). However, recent work has revealed a significant post-translational dimension, highlighting the strong role of the ubiquitin-proteasome system (UPS) in BNIP3 regulation. With these emerging concepts in mind, we aimed to develop a unified understanding of how steady-state levels of BNIP3 are established and maintained and how this regulation governs underlying cell physiology.


Assuntos
Proteínas de Membrana , Mitofagia , Proteínas Proto-Oncogênicas , Animais , Humanos , Autofagia/fisiologia , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Mitofagia/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
19.
Autophagy ; 20(6): 1359-1382, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38447939

RESUMO

Mitophagy involves the selective elimination of defective mitochondria during chemotherapeutic stress to maintain mitochondrial homeostasis and sustain cancer growth. Here, we showed that CLU (clusterin) is localized to mitochondria to induce mitophagy controlling mitochondrial damage in oral cancer cells. Moreover, overexpression and knockdown of CLU establish its mitophagy-specific role, where CLU acts as an adaptor protein that coordinately interacts with BAX and LC3 recruiting autophagic machinery around damaged mitochondria in response to cisplatin treatment. Interestingly, CLU triggers class III phosphatidylinositol 3-kinase (PtdIns3K) activity around damaged mitochondria, and inhibition of mitophagic flux causes the accumulation of excessive mitophagosomes resulting in reactive oxygen species (ROS)-dependent apoptosis during cisplatin treatment in oral cancer cells. In parallel, we determined that PPARGC1A/PGC1α (PPARG coactivator 1 alpha) activates mitochondrial biogenesis during CLU-induced mitophagy to maintain the mitochondrial pool. Intriguingly, PPARGC1A inhibition through small interfering RNA (siPPARGC1A) and pharmacological inhibitor (SR-18292) treatment counteracts CLU-dependent cytoprotection leading to mitophagy-associated cell death. Furthermore, co-treatment of SR-18292 with cisplatin synergistically suppresses tumor growth in oral cancer xenograft models. In conclusion, CLU and PPARGC1A are essential for sustained cancer cell growth by activating mitophagy and mitochondrial biogenesis, respectively, and their inhibition could provide better therapeutic benefits against oral cancer.


Assuntos
Sobrevivência Celular , Clusterina , Mitocôndrias , Mitofagia , Neoplasias Bucais , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Humanos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Clusterina/metabolismo , Clusterina/genética , Mitofagia/efeitos dos fármacos , Mitofagia/fisiologia , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Neoplasias Bucais/patologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/genética , Animais , Sobrevivência Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Biogênese de Organelas , Camundongos , Apoptose/efeitos dos fármacos , Camundongos Nus , Espécies Reativas de Oxigênio/metabolismo , Autofagia/fisiologia , Autofagia/efeitos dos fármacos
20.
Domest Anim Endocrinol ; 88: 106847, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38479188

RESUMO

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.


Assuntos
Animais Recém-Nascidos , Temperatura Baixa , Glucose , Cabras , Mitofagia , Músculo Esquelético , Animais , Cabras/fisiologia , Músculo Esquelético/metabolismo , Mitofagia/fisiologia , Glucose/metabolismo , Metabolismo dos Lipídeos , Gotículas Lipídicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...