• 제목/요약/키워드: mitochondrial dysfunction

검색결과 253건 처리시간 0.026초

Emerging perspectives on mitochondrial dysfunction and inflammation in Alzheimer's disease

  • Yoo, Seung-Min;Park, Jisu;Kim, Seo-Hyun;Jung, Yong-Keun
    • BMB Reports
    • /
    • 제53권1호
    • /
    • pp.35-46
    • /
    • 2020
  • Despite enduring diverse insults, mitochondria maintain normal functions through mitochondrial quality control. However, the failure of mitochondrial quality control resulting from excess damage and mechanical defects causes mitochondrial dysfunction, leading to various human diseases. Recent studies have reported that mitochondrial defects are found in Alzheimer's disease (AD) and worsen AD symptoms. In AD pathogenesis, mitochondrial dysfunction-driven generation of reactive oxygen species (ROS) and their contribution to neuronal damage has been widely studied. In contrast, studies on mitochondrial dysfunction-associated inflammatory responses have been relatively scarce. Moreover, ROS produced upon failure of mitochondrial quality control may be linked to the inflammatory response and influence the progression of AD. Thus, this review will focus on inflammatory pathways that are associated with and initiated through defective mitochondria and will summarize recent progress on the role of mitochondria-mediated inflammation in AD. We will also discuss how reducing mitochondrial dysfunction-mediated inflammation could affect AD.

The Role of Mitochondrial Biogenesis Dysfunction in Diabetic Cardiomyopathy

  • Tao, Li-Chan;Wang, Ting-ting;Zheng, Lu;Hua, Fei;Li, Jian-Jun
    • Biomolecules & Therapeutics
    • /
    • 제30권5호
    • /
    • pp.399-408
    • /
    • 2022
  • Diabetic cardiomyopathy (DCM) is described as abnormalities of myocardial structure and function in diabetic patients without other well-established cardiovascular factors. Although multiple pathological mechanisms involving in this unique myocardial disorder, mitochondrial dysfunction may play an important role in its development of DCM. Recently, considerable progresses have suggested that mitochondrial biogenesis is a tightly controlled process initiating mitochondrial generation and maintaining mitochondrial function, appears to be associated with DCM. Nonetheless, an outlook on the mechanisms and clinical relevance of dysfunction in mitochondrial biogenesis among patients with DCM is not completely understood. In this review, hence, we will summarize the role of mitochondrial biogenesis dysfunction in the development of DCM, especially the molecular underlying mechanism concerning the signaling pathways beyond the stimulation and inhibition of mitochondrial biogenesis. Additionally, the evaluations and potential therapeutic strategies regarding mitochondrial biogenesis dysfunction in DCM is also presented.

The role of cell type-specific mitochondrial dysfunction in the pathogenesis of Alzheimer's disease

  • Kim, Dong Kyu;MookJung, Inhee
    • BMB Reports
    • /
    • 제52권12호
    • /
    • pp.679-688
    • /
    • 2019
  • The decrease of metabolism in the brain has been observed as the important lesions of Alzheimer's disease (AD) from the early stages of diagnosis. The cumulative evidence has reported that the failure of mitochondria, an organelle involved in diverse biological processes as well as energy production, maybe the cause or effect of the pathogenesis of AD. Both amyloid and tau pathologies have an impact upon mitochondria through physical interaction or indirect signaling pathways, resulting in the disruption of mitochondrial function and dynamics which can trigger AD. In addition, mitochondria are involved in different biological processes depending on the specific functions of each cell type in the brain. Thus, it is necessary to understand mitochondrial dysfunction as part of the pathological phenotypes of AD according to each cell type. In this review, we summarize that 1) the effects of AD pathology inducing mitochondrial dysfunction and 2) the contribution of mitochondrial dysfunction in each cell type to AD pathogenesis.

Effects of exercise on obesity-induced mitochondrial dysfunction in skeletal muscle

  • Heo, Jun-Won;No, Mi-Hyun;Park, Dong-Ho;Kang, Ju-Hee;Seo, Dae Yun;Han, Jin;Neufer, P. Darrell;Kwak, Hyo-Bum
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제21권6호
    • /
    • pp.567-577
    • /
    • 2017
  • Obesity is known to induce inhibition of glucose uptake, reduction of lipid metabolism, and progressive loss of skeletal muscle function, which are all associated with mitochondrial dysfunction in skeletal muscle. Mitochondria are dynamic organelles that regulate cellular metabolism and bioenergetics, including ATP production via oxidative phosphorylation. Due to these critical roles of mitochondria, mitochondrial dysfunction results in various diseases such as obesity and type 2 diabetes. Obesity is associated with impairment of mitochondrial function (e.g., decrease in $O_2$ respiration and increase in oxidative stress) in skeletal muscle. The balance between mitochondrial fusion and fission is critical to maintain mitochondrial homeostasis in skeletal muscle. Obesity impairs mitochondrial dynamics, leading to an unbalance between fusion and fission by favorably shifting fission or reducing fusion proteins. Mitophagy is the catabolic process of damaged or unnecessary mitochondria. Obesity reduces mitochondrial biogenesis in skeletal muscle and increases accumulation of dysfunctional cellular organelles, suggesting that mitophagy does not work properly in obesity. Mitochondrial dysfunction and oxidative stress are reported to trigger apoptosis, and mitochondrial apoptosis is induced by obesity in skeletal muscle. It is well known that exercise is the most effective intervention to protect against obesity. Although the cellular and molecular mechanisms by which exercise protects against obesity-induced mitochondrial dysfunction in skeletal muscle are not clearly elucidated, exercise training attenuates mitochondrial dysfunction, allows mitochondria to maintain the balance between mitochondrial dynamics and mitophagy, and reduces apoptotic signaling in obese skeletal muscle.

Ginsenoside Rg1 ameliorates chronic intermittent hypoxia-induced vascular endothelial dysfunction by suppressing the formation of mitochondrial reactive oxygen species through the calpain-1 pathway

  • Fang Zhao;Meili Lu;Hongxin Wang
    • Journal of Ginseng Research
    • /
    • 제47권1호
    • /
    • pp.144-154
    • /
    • 2023
  • Background: As the major pathophysiological feature of obstructive sleep apnea (OSA), chronic intermittent hypoxia (CIH) is vital for the occurrence of cardiovascular complications. The activation of calpain-1 mediates the production of endothelial reactive oxygen species (ROS) and impairs nitric oxide (NO) bioavailability, resulting in vascular endothelial dysfunction (VED). Ginsenoside Rg1 is thought to against endothelial cell dysfunction, but the potential mechanism of CIH-induced VED remains unclear. Methods: C57BL/6 mice and human coronary artery endothelial cells (HCAECs) were exposed to CIH following knockout or overexpression of calpain-1. The effect of ginsenoside Rg1 on VED, oxidative stress, mitochondrial dysfunction, and the expression levels of calpain-1, PP2A and p-eNOS were detected both in vivo and in vitro. Results: CIH promoted VED, oxidative stress and mitochondrial dysfunction accompanied by enhanced levels of calpain-1 and PP2A and reduced levels of p-eNOS in mice and cellular levels. Ginsenoside Rg1, calpain-1 knockout, OKA, NAC and TEMPOL treatment protected against CIH-induced VED, oxidative stress and mitochondrial dysfunction, which is likely concomitant with the downregulated protein expression of calpain-1 and PP2A and the upregulation of p-eNOS in mice and cellular levels. Calpain-1 overexpression increased the expression of PP2A, reduced the level of p-eNOS, and accelerated the occurrence and development of VED, oxidative stress and mitochondrial dysfunction in HCAECs exposed to CIH. Moreover, scavengers of O2·-, H2O2, complex I or mitoKATP abolished CIH-induced impairment in endothelial-dependent relaxation. Conclusion: Ginsenoside Rg1 may alleviate CIH-induced vascular endothelial dysfunction by suppressing the formation of mitochondrial reactive oxygen species through the calpain-1 pathway.

미토콘드리아 기능 이상과 암 (Mitochondrial Dysfunction and Cancer)

  • 한유선;제갈명은;김영진
    • 생명과학회지
    • /
    • 제29권9호
    • /
    • pp.1034-1046
    • /
    • 2019
  • 미토콘드리아는 세포 에너지 공급을 위한 에너지 대사의 주요 세포 소기관으로 칼슘 조절, 활성 산소(ROS) 생성, 세포 사멸(apoptosis)을 조절하는데도 중요한 역할을 한다. 이러한 미토콘드리아에 발생한 기능 이상은 신경퇴행질환, 루게릭병, 심혈관계 질환, 정신 질환, 당뇨, 암과 같은 다양한 질병과 연관이 있다. 미토콘드리아 기능 이상 관련 질병들은 노화와 관련된 질병이 주를 차지하며, 이 논문에서는 그 중에서도 암에 초점을 맞춰 서술하고자 한다. 미토콘드리아 기능 이상은 발암을 유도하며, 많은 암 종에서 발견된다. 암종에 따라 미토콘드리아 기능 이상을 일으키는 요인들이 다르며, 이러한 변화는 치료 내성, 전이와 같은 암 악성화도 유발한다. 미토콘드리아 기능 이상의 요인으로는 미토콘드리아 수 부족, 주요 물질 제공 불능, ATP 합성 기능 이상 등이 존재하나, 암 발병과 악성화에 영향을 미치는 주요 원인으로 미토콘드리아 DNA (mtDNA)의 감소(depletion)를 들 수 있다. 미토콘드리아 기능 이상은 분자 활성 변화 혹은 발현 변화를 통해 암 악성화를 일으키나, 어떠한 변화가 암 악성화를 야기하는지 구체적으로 알려진 바가 없다. 미토콘드리아 기능 이상과 암의 상관관계는 대부분 미토콘드리아 기능 이상 세포를 이용하여 연구하는데, 그 제작 방법으로는 EtBr에 의한 화학적 방법과 shRNA, Crispr/Cas9과 같은 유전자 수선 (gene editing) 방법 등이 있다. 이러한 기법으로 제작된 미토콘드리아 기능 이상 세포주는 암을 비롯한 미토콘드리아 기능 이상에 의한 다양한 질병 연구에 이용되고 있다.

RUNX1 Upregulation Causes Mitochondrial Dysfunction via Regulating the PI3K-Akt Pathway in iPSC from Patients with Down Syndrome

  • Yanna Liu;Yuehua Zhang;Zhaorui Ren;Fanyi Zeng;Jingbin Yan
    • Molecules and Cells
    • /
    • 제46권4호
    • /
    • pp.219-230
    • /
    • 2023
  • Down syndrome (DS) is the most common autosomal aneuploidy caused by trisomy of chromosome 21. Previous studies demonstrated that DS affected mitochondrial functions, which may be associated with the abnormal development of the nervous system in patients with DS. Runt-related transcription factor 1 (RUNX1) is an encoding gene located on chromosome 21. It has been reported that RUNX1 may affect cell apoptosis via the mitochondrial pathway. The present study investigated whether RUNX1 plays a critical role in mitochondrial dysfunction in DS and explored the mechanism by which RUNX1 affects mitochondrial functions. Expression of RUNX1 was detected in induced pluripotent stem cells of patients with DS (DS-iPSCs) and normal iPSCs (N-iPSCs), and the mitochondrial functions were investigated in the current study. Subsequently, RUNX1 was overexpressed in N-iPSCs and inhibited in DS-iPSCs. The mitochondrial functions were investigated thoroughly, including reactive oxygen species levels, mitochondrial membrane potential, ATP content, and lysosomal activity. Finally, RNA-sequencing was used to explore the global expression pattern. It was observed that the expression levels of RUNX1 in DS-iPSCs were significantly higher than those in normal controls. Impaired mitochondrial functions were observed in DS-iPSCs. Of note, overexpression of RUNX1 in N-iPSCs resulted in mitochondrial dysfunction, while inhibition of RUNX1 expression could improve the mitochondrial function in DS-iPSCs. Global gene expression analysis indicated that overexpression of RUNX1 may promote the induction of apoptosis in DS-iPSCs by activating the PI3K/Akt signaling pathway. The present findings indicate that abnormal expression of RUNX1 may play a critical role in mitochondrial dysfunction in DS-iPSCs.

The relationship between muscle mitochondrial nutritional overloading and insulin resistance

  • Jeon, Jae-Han;Moon, Jun-Sung;Won, Kyu-Chang;Lee, In-Kyu
    • Journal of Yeungnam Medical Science
    • /
    • 제34권1호
    • /
    • pp.19-28
    • /
    • 2017
  • The incidence of type 2 diabetes mellitus and insulin resistance is growing rapidly. Multiple organs including the liver, skeletal muscle and adipose tissue control insulin sensitivity coordinately, but the mechanism of skeletal muscle insulin resistance has not yet been fully elucidated. However, there is a growing body of evidence that lipotoxicity induced by mitochondrial dysfunction in skeletal muscle is an important mediator of insulin resistance. However, some recent findings suggest that skeletal mitochondrial dysfunction generated by genetic manipulation is not always correlated with insulin resistance in animal models. A high fat diet can provoke insulin resistance despite a coordinate increase in skeletal muscle mitochondria, which implies that mitochondrial dysfunction is not mandatory in insulin resistance. Furthermore, incomplete fatty acid oxidation by excessive nutrition supply compared to mitochondrial demand can induce insulin resistance without preceding impairment of mitochondrial function. Taken together we suggested that skeletal muscle mitochondrial overloading, not mitochondrial dysfunction, plays a pivotal role in insulin resistance.

Mitochondrial dysfunction suppresses p53 expression via calcium-mediated nuclear factor-κB signaling in HCT116 human colorectal carcinoma cells

  • Lee, Young-Kyoung;Yi, Eui-Yeun;Park, Shi-Young;Jang, Won-Jun;Han, Yu-Seon;Jegal, Myeong-Eun;Kim, Yung-Jin
    • BMB Reports
    • /
    • 제51권6호
    • /
    • pp.296-301
    • /
    • 2018
  • Mitochondrial DNA (mtDNA) mutations are often observed in various cancer types. Although the correlation between mitochondrial dysfunction and cancer malignancy has been demonstrated by several studies, further research is required to elucidate the molecular mechanisms underlying accelerated tumor development and progression due to mitochondrial mutations. We generated an mtDNA-depleted cell line, ${\rho}^0$, via long-term ethidium bromide treatment to define the molecular mechanisms of tumor malignancy induced by mitochondrial dysfunction. Mitochondrial dysfunction in ${\rho}^0$ cells reduced drug-induced cell death and decreased the expression of pro-apoptotic proteins including p53. The p53 expression was reduced by activation of nuclear $factor-{\kappa}B$ that depended on elevated levels of free calcium in $HCT116/{\rho}^0$ cells. Overall, these data provide a novel mechanism for tumor development and drug resistance due to mitochondrial dysfunction.

An experimental approach to study the function of mitochondria in cardiomyopathy

  • Chung, Youn Wook;Kang, Seok-Min
    • BMB Reports
    • /
    • 제48권10호
    • /
    • pp.541-548
    • /
    • 2015
  • Cardiomyopathy is an inherited or acquired disease of the myocardium, which can result in severe ventricular dysfunction. Mitochondrial dysfunction is involved in the pathological process of cardiomyopathy. Many dysfunctions in cardiac mitochondria are consequences of mutations in nuclear or mitochondrial DNA followed by alterations in transcriptional regulation, mitochondrial protein function, and mitochondrial dynamics and energetics, presenting with associated multisystem mitochondrial disorders. To ensure correct diagnosis and optimal management of mitochondrial dysfunction in cardiomyopathy caused by multiple pathogenesis, multidisciplinary approaches are required, and to integrate between clinical and basic sciences, ideal translational models are needed. In this review, we will focus on experimental models to provide insights into basic mitochondrial physiology and detailed underlying mechanisms of cardiomyopathy and current mitochondria-targeted therapies for cardiomyopathy.