• 제목/요약/키워드: cellular dysfunction

검색결과 186건 처리시간 0.024초

Etoposide Induces Mitochondrial Dysfunction and Cellular Senescence in Primary Cultured Rat Astrocytes

  • Bang, Minji;Kim, Do Gyeong;Gonzales, Edson Luck;Kwon, Kyoung Ja;Shin, Chan Young
    • Biomolecules & Therapeutics
    • /
    • 제27권6호
    • /
    • pp.530-539
    • /
    • 2019
  • Brain aging is an inevitable process characterized by structural and functional changes and is a major risk factor for neurodegenerative diseases. Most brain aging studies are focused on neurons and less on astrocytes which are the most abundant cells in the brain known to be in charge of various functions including the maintenance of brain physical formation, ion homeostasis, and secretion of various extracellular matrix proteins. Altered mitochondrial dynamics, defective mitophagy or mitochondrial damages are causative factors of mitochondrial dysfunction, which is linked to age-related disorders. Etoposide is an anti-cancer reagent which can induce DNA stress and cellular senescence of cancer cell lines. In this study, we investigated whether etoposide induces senescence and functional alterations in cultured rat astrocytes. Senescence-associated ${\beta}$-galactosidase (SA-${\beta}$-gal) activity was used as a cellular senescence marker. The results indicated that etoposide-treated astrocytes showed cellular senescence phenotypes including increased SA-${\beta}$-gal-positive cells number, increased nuclear size and increased senescence-associated secretory phenotypes (SASP) such as IL-6. We also observed a decreased expression of cell cycle markers, including PhosphoHistone H3/Histone H3 and CDK2, and dysregulation of cellular functions based on wound-healing, neuronal protection, and phagocytosis assays. Finally, mitochondrial dysfunction was noted through the determination of mitochondrial membrane potential using tetramethylrhodamine methyl ester (TMRM) and the measurement of mitochondrial oxygen consumption rate (OCR). These data suggest that etoposide can induce cellular senescence and mitochondrial dysfunction in astrocytes which may have implications in brain aging and neurodegenerative conditions.

세포교정영양요법(OCNT)를 이용한 중증근무력증 개선사례 (A Case Study on Improvement of Myasthenia Gravis Using Ortho-Cellular Nutrition Therapy (OCNT))

  • 지은실
    • 셀메드
    • /
    • 제14권5호
    • /
    • pp.78.1-78.5
    • /
    • 2024
  • Objective: Improvement of myasthenia gravis through Ortho-Cellular Nutrition Therapy (OCNT). Methods: A 52-year-old patient with myasthenia gravis was prescribed 21 different OCNT treatments, including Cyaplex. Results: Following the administration of OCNT, symptoms such as eyelid ptosis, masticatory dysfunction, diplopia, and fatigue showed improvement. Conclusion: OCNT can assist in alleviating and mitigating symptoms for patients suffering from thymoma and myasthenia gravis.

Impaired Autophagic Flux in Glucose-Deprived Cells: An Outcome of Lysosomal Acidification Failure Exacerbated by Mitophagy Dysfunction

  • Eun Seong Hwang;Seon Beom Song
    • Molecules and Cells
    • /
    • 제46권11호
    • /
    • pp.655-663
    • /
    • 2023
  • Autophagy dysfunction is associated with human diseases and conditions including neurodegenerative diseases, metabolic issues, and chronic infections. Additionally, the decline in autophagic activity contributes to tissue and organ dysfunction and aging-related diseases. Several factors, such as down-regulation of autophagy components and activators, oxidative damage, microinflammation, and impaired autophagy flux, are linked to autophagy decline. An autophagy flux impairment (AFI) has been implicated in neurological disorders and in certain other pathological conditions. Here, to enhance our understanding of AFI, we conducted a comprehensive literature review of findings derived from two well-studied cellular stress models: glucose deprivation and replicative senescence. Glucose deprivation is a condition in which cells heavily rely on oxidative phosphorylation for ATP generation. Autophagy is activated, but its flux is hindered at the autolysis step, primarily due to an impairment of lysosomal acidity. Cells undergoing replicative senescence also experience AFI, which is also known to be caused by lysosomal acidity failure. Both glucose deprivation and replicative senescence elevate levels of reactive oxygen species (ROS), affecting lysosomal acidification. Mitochondrial alterations play a crucial role in elevating ROS generation and reducing lysosomal acidity, highlighting their association with autophagy dysfunction and disease conditions. This paper delves into the underlying molecular and cellular pathways of AFI in glucose-deprived cells, providing insights into potential strategies for managing AFI that is driven by lysosomal acidity failure. Furthermore, the investigation on the roles of mitochondrial dysfunction sheds light on the potential effectiveness of modulating mitochondrial function to overcome AFI, offering new possibilities for therapeutic interventions.

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.

Gateway RFP-Fusion Vectors for High Throughput Functional Analysis of Genes

  • Park, Jae-Yong;Hwang, Eun Mi;Park, Nammi;Kim, Eunju;Kim, Dong-Gyu;Kang, Dawon;Han, Jaehee;Choi, Wan Sung;Ryu, Pan-Dong;Hong, Seong-Geun
    • Molecules and Cells
    • /
    • 제23권3호
    • /
    • pp.357-362
    • /
    • 2007
  • There is an increasing demand for high throughput (HTP) methods for gene analysis on a genome-wide scale. However, the current repertoire of HTP detection methodologies allows only a limited range of cellular phenotypes to be studied. We have constructed two HTP-optimized expression vectors generated from the red fluorescent reporter protein (RFP) gene. These vectors produce RFP-tagged target proteins in a multiple expression system using gateway cloning technology (GCT). The RFP tag was fused with the cloned genes, thereby allowing us localize the expressed proteins in mammalian cells. The effectiveness of the vectors was evaluated using an HTP-screening system. Sixty representative human C2 domains were tagged with RFP and overexpressed in HiB5 neuronal progenitor cells, and we studied in detail two C2 domains that promoted the neuronal differentiation of HiB5 cells. Our results show that the two vectors developed in this study are useful for functional gene analysis using an HTP-screening system on a genome-wide scale.

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.

Mitochondrial fatty acid metabolism in acute kidney injury

  • Jang, Hee-Seong;Padanilam, Babu J.
    • Journal of Medicine and Life Science
    • /
    • 제15권2호
    • /
    • pp.37-41
    • /
    • 2018
  • Mitochondrial injury in renal tubule has been recognized as a major contributor in acute kidney injury (AKI) pathogenesis. Ischemic insult, nephrotoxin, endotoxin and contrast medium destroy mitochondrial structure and function as well as their biogenesis and dynamics, especially in renal proximal tubule, to elicit ATP depletion. Mitochondrial fatty acid ${\beta}$-oxidation (FAO) is the preferred source of ATP in the kidney, and its impairment is a critical factor in AKI pathogenesis. This review explores current knowledge of mitochondrial dysfunction and energy depletion in AKI and prospective views on developing therapeutic strategies targeting mitochondrial dysfunction in AKI.

Senotherapeutics and Their Molecular Mechanism for Improving Aging

  • Park, Jooho;Shin, Dong Wook
    • Biomolecules & Therapeutics
    • /
    • 제30권6호
    • /
    • pp.490-500
    • /
    • 2022
  • Aging is defined as physiological dysfunction of the body and a key risk factor for human diseases. During the aging process, cellular senescence occurs in response to various extrinsic and intrinsic factors such as radiation-induced DNA damage, the activation of oncogenes, and oxidative stress. These senescent cells accumulate in many tissues and exhibit diverse phenotypes, such as resistance to apoptosis, production of senescence-associated secretory phenotype, cellular flattening, and cellular hypertrophy. They also induce abnormal dysfunction of the microenvironment and damage neighboring cells, eventually causing harmful effects in the development of various chronic diseases such as diabetes, cancer, and neurodegenerative diseases. Thus, pharmacological interventions targeting senescent cells, called senotherapeutics, have been extensively studied. These senotherapeutics provide a novel strategy for extending the health span and improving age-related diseases. In this review, we discuss the current progress in understanding the molecular mechanisms of senotherapeutics and provide insights for developing senotherapeutics.

Genetic Variations Leading to Familial Dilated Cardiomyopathy

  • Cho, Kae Won;Lee, Jongsung;Kim, Youngjo
    • Molecules and Cells
    • /
    • 제39권10호
    • /
    • pp.722-727
    • /
    • 2016
  • Cardiomyopathy is a major cause of death worldwide. Based on pathohistological abnormalities and clinical manifestation, cardiomyopathies are categorized into several groups: hypertrophic, dilated, restricted, arrhythmogenic right ventricular, and unclassified. Dilated cardiomyopathy, which is characterized by dilation of the left ventricle and systolic dysfunction, is the most severe and prevalent form of cardiomyopathy and usually requires heart transplantation. Its etiology remains unclear. Recent genetic studies of single gene mutations have provided significant insights into the complex processes of cardiac dysfunction. To date, over 40 genes have been demonstrated to contribute to dilated cardiomyopathy. With advances in genetic screening techniques, novel genes associated with this disease are continuously being identified. The respective gene products can be classified into several functional groups such as sarcomere proteins, structural proteins, ion channels, and nuclear envelope proteins. Nuclear envelope proteins are emerging as potential molecular targets in dilated cardiomyopathy. Because they are not directly associated with contractile force generation and transmission, the molecular pathways through which these proteins cause cardiac muscle disorder remain unclear. However, nuclear envelope proteins are involved in many essential cellular processes. Therefore, integrating apparently distinct cellular processes is of great interest in elucidating the etiology of dilated cardiomyopathy. In this mini review, we summarize the genetic factors associated with dilated cardiomyopathy and discuss their cellular functions.

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.