• Title/Summary/Keyword: cellular dysfunction

Search Result 193, Processing Time 0.02 seconds

Recent Research Trends in Induction of Cellular Senescence by Microplastics (미세플라스틱에 의한 세포 노화 유도의 최근 연구 동향)

  • Yung Hyun Choi
    • Journal of Life Science
    • /
    • v.34 no.8
    • /
    • pp.594-607
    • /
    • 2024
  • Plastic products have long been widely used in both industrial and household applications. However, tiny plastic particles derived from plastic products, such as microplastics (MPs) and nanoplastics (NPs), can infiltrate the human body through inhalation, ingestion, or skin contact. Once inside cells via endocytosis, MPs and NPs (MNPs) can trigger autophagy, but lysosomal dysfunction can block autophagic flux. Accumulating in the cytoplasm, these particles induce cellular stress, including oxidative stress from free radicals, mitochondrial dysfunction, and increased inflammatory response. Meanwhile, cellular senescence is a hallmark of aging and is defined as the stable termination of the cell cycle in response to cell damage and stress. In particular, the accumulation of oxidative stress, a key factor in inducing cellular senescence, induces the expression of major senescence markers. Senescent cells increase the secretion of senescence-associated secretory phenotype, including inflammatory cytokines and chemokines. Despite growing interest in how MNPs induce cellular senescence, there remains a gap regarding their onset and therapeutic targets. Therefore, this review focuses on identifying recent research trends on how MNPs induce cellular aging in key human cell types and proposes future research directions to overcome these challenges.

Exploiting tumor cell senescence in anticancer therapy

  • Lee, Minyoung;Lee, Jae-Seon
    • BMB Reports
    • /
    • v.47 no.2
    • /
    • pp.51-59
    • /
    • 2014
  • Cellular senescence is a physiological process of irreversible cell-cycle arrest that contributes to various physiological and pathological processes of aging. Whereas replicative senescence is associated with telomere attrition after repeated cell division, stress-induced premature senescence occurs in response to aberrant oncogenic signaling, oxidative stress, and DNA damage which is independent of telomere dysfunction. Recent evidence indicates that cellular senescence provides a barrier to tumorigenesis and is a determinant of the outcome of cancer treatment. However, the senescence-associated secretory phenotype, which contributes to multiple facets of senescent cancer cells, may influence both cancer-inhibitory and cancer-promoting mechanisms of neighboring cells. Conventional treatments, such as chemo- and radiotherapies, preferentially induce premature senescence instead of apoptosis in the appropriate cellular context. In addition, treatment-induced premature senescence could compensate for resistance to apoptosis via alternative signaling pathways. Therefore, we believe that an intensive effort to understand cancer cell senescence could facilitate the development of novel therapeutic strategies for improving the efficacy of anticancer therapies. This review summarizes the current understanding of molecular mechanisms, functions, and clinical applications of cellular senescence for anticancer therapy.

Senolytics and Senostatics: A Two-Pronged Approach to Target Cellular Senescence for Delaying Aging and Age-Related Diseases

  • Kang, Chanhee
    • Molecules and Cells
    • /
    • v.42 no.12
    • /
    • pp.821-827
    • /
    • 2019
  • Aging is the most important single risk factor for many chronic diseases such as cancer, metabolic syndrome, and neurodegenerative disorders. Targeting aging itself might, therefore, be a better strategy than targeting each chronic disease individually for enhancing human health. Although much should be achieved for completely understanding the biological basis of aging, cellular senescence is now believed to mainly contribute to organismal aging via two independent, yet not mutually exclusive mechanisms: on the one hand, senescence of stem cells leads to exhaustion of stem cells and thus decreases tissue regeneration. On the other hand, senescent cells secrete many proinflammatory cytokines, chemokines, growth factors, and proteases, collectively termed as the senescence-associated secretory phenotype (SASP), which causes chronic inflammation and tissue dysfunction. Much effort has been recently made to therapeutically target detrimental effects of cellular senescence including selectively eliminating senescent cells (senolytics) and modulating a proinflammatory senescent secretome (senostatics). Here, we discuss current progress and limitations in understanding molecular mechanisms of senolytics and senostatics and therapeutic strategies for applying them. Furthermore, we propose how these novel interventions for aging treatment could be improved, based on lessons learned from cancer treatment.

Alteration of mitochondrial DNA content modulates antioxidant enzyme expressions and oxidative stress in myoblasts

  • Min, Kyung-Ho;Lee, Wan
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.23 no.6
    • /
    • pp.519-528
    • /
    • 2019
  • Mitochondrial dysfunction is closely associated with reactive oxygen species (ROS) generation and oxidative stress in cells. On the other hand, modulation of the cellular antioxidant defense system by changes in the mitochondrial DNA (mtDNA) content is largely unknown. To determine the relationship between the cellular mtDNA content and defense system against oxidative stress, this study examined a set of myoblasts containing a depleted or reverted mtDNA content. A change in the cellular mtDNA content modulated the expression of antioxidant enzymes in myoblasts. In particular, the expression and activity of glutathione peroxidase (GPx) and catalase were inversely correlated with the mtDNA content in myoblasts. The depletion of mtDNA decreased both the reduced glutathione (GSH) and oxidized glutathione (GSSG) slightly, whereas the cellular redox status, as assessed by the GSH/GSSG ratio, was similar to that of the control. Interestingly, the steady-state level of the intracellular ROS, which depends on the reciprocal actions between ROS generation and detoxification, was reduced significantly and the lethality induced by $H_2O_2$ was alleviated by mtDNA depletion in myoblasts. Therefore, these results suggest that the ROS homeostasis and antioxidant enzymes are modulated by the cellular mtDNA content and that the increased expression and activity of GPx and catalase through the depletion of mtDNA are closely associated with an alleviation of the oxidative stress in myoblasts.

FUN14 Domain-Containing Protein 1 Is Involved in Amyloid Beta Peptide-Induced Mitochondrial Dysfunction and Cell Injury in HT-22 Neuronal Cells (HT-22 신경세포에서 아밀로이드 베타 펩티드에 의한 미토콘드리아와 세포 손상 기전에서 FUN14 도메인 함유 단백 1의 역할)

  • Jae Hoon Kang;Jae Suk Woo
    • Journal of Life Science
    • /
    • v.34 no.1
    • /
    • pp.37-47
    • /
    • 2024
  • FUN14 domain-containing protein 1 (FUNDC1), an outer mitochondrial membrane protein, contributes to removal of damaged mitochondria through mitophagy. In this study, to elucidate the role of the FUNDC1 in the amyloid beta peptide (Aβ)-induced neuropathy, changes in the degree of mitochondrial dysfunction and cell injury caused by Aβ treatment were examined in the HT-22 neuronal cells in which the FUNDC1 expression was transiently silenced or overexpressed. We found that Aβ treatment causes a time-dependent decrease of the FUNDC1 expression. In the Aβ-treated cells, there were a drop in MTT reduction ability, depletion of cellular ATP, disruption of mitochondrial membrane potential, stimulation of cellular ROS production, and increased mitochondrial Ca2+ load. Activation of caspase-3 and induction of apoptotic cell death were also observed. Transient silencing of the FUNDC1 expression by transfection with the FUNDC1 small interfering RNA per se caused mitochondrial dysfunction and apoptotic cell death like the effect of Aβ treatment. Conversely, in cells in which the FUNDC1 was transiently overexpressed by FUNDC1-Myc transfection, overexpression itself had no effect on the mitochondrial functional integrity and cell survival but showed a significant prevention effect against mitochondrial and cell injury caused by Aβ treatment. Overall, these results suggest that the FUNDC1 is importantly involved in the Aβ-induced mitochondrial dysfunction and cell injury in the HT-22 neuronal cells.

Enhanced Production of Adenosine Triphosphate by Pharmacological Activation of Adenosine Monophosphate-Activated Protein Kinase Ameliorates Acetaminophen-Induced Liver Injury

  • Hwang, Jung Hwan;Kim, Yong-Hoon;Noh, Jung-Ran;Choi, Dong-Hee;Kim, Kyoung-Shim;Lee, Chul-Ho
    • Molecules and Cells
    • /
    • v.38 no.10
    • /
    • pp.843-850
    • /
    • 2015
  • The1hepatic cell death induced by acetaminophen (APAP) is closely related to cellular adenosine triphosphate (ATP) depletion, which is mainly caused by mitochondrial dysfunction. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a key sensor of low energy status. AMPK regulates metabolic homeostasis by stimulating catabolic metabolism and suppressing anabolic pathways to increase cellular energy levels. We found that the decrease in active phosphorylation of AMPK in response to APAP correlates with decreased ATP levels, in vivo. Therefore, we hypothesized that the enhanced production of ATP via AMPK stimulation can lead to amelioration of APAP-induced liver failure. A769662, an allosteric activator of AMPK, produced a strong synergistic effect on AMPK Thr172 phosphorylation with APAP in primary hepatocytes and liver tissue. Interestingly, activation of AMPK by A769662 ameliorated the APAP-induced hepatotoxicity in C57BL/6N mice treated with APAP at a dose of 400 mg/kg intraperitoneally. However, mice treated with APAP alone developed massive centrilobular necrosis, and APAP increased their serum alanine aminotransferase and aspartate aminotransferase levels. Furthermore, A769662 administration prevented the loss of intracellular ATP without interfering with the APAP-mediated reduction of mitochondrial dysfunction. In contrast, inhibition of glycolysis by 2-deoxy-glucose eliminated the beneficial effects of A769662 on APAP-mediated liver injury. In conclusion, A769662 can effectively protect mice against APAP-induced liver injury through ATP synthesis by anaerobic glycolysis. Furthermore, stimulation of AMPK may have potential therapeutic application for APAP overdose.

Apolipoprotein E in Synaptic Plasticity and Alzheimer's Disease: Potential Cellular and Molecular Mechanisms

  • Kim, Jaekwang;Yoon, Hyejin;Basak, Jacob;Kim, Jungsu
    • Molecules and Cells
    • /
    • v.37 no.11
    • /
    • pp.767-776
    • /
    • 2014
  • Alzheimer's disease (AD) is clinically characterized with progressive memory loss and cognitive decline. Synaptic dysfunction is an early pathological feature that occurs prior to neurodegeneration and memory dysfunction. Mounting evidence suggests that aggregation of amyloid-${\alpha}$ ($A{\alpha}$) and hyperphosphorylated tau leads to synaptic deficits and neurodegeneration, thereby to memory loss. Among the established genetic risk factors for AD, the ${\varepsilon}4$ allele of apolipoprotein E (APOE) is the strongest genetic risk factor. We and others previously demonstrated that apoE regulates $A{\alpha}$ aggregation and clearance in an isoform-dependent manner. While the effect of apoE on $A{\alpha}$ may explain how apoE isoforms differentially affect AD pathogenesis, there are also other underexplored pathogenic mechanisms. They include differential effects of apoE on cerebral energy metabolism, neuroinflammation, neurovascular function, neurogenesis, and synaptic plasticity. ApoE is a major carrier of cholesterols that are required for neuronal activity and injury repair in the brain. Although there are a few conflicting findings and the underlying mechanism is still unclear, several lines of studies demonstrated that apoE4 leads to synaptic deficits and impairment in long-term potentiation, memory and cognition. In this review, we summarize current understanding of apoE function in the brain, with a particular emphasis on its role in synaptic plasticity and the underlying cellular and molecular mechanisms, involving low-density lipoprotein receptor-related protein 1 (LRP1), syndecan, and LRP8/ApoER2.

Endothelin Receptor Overexpression Alters Diastolic Function in Cultured Rat Ventricular Myocytes

  • Kang, Mi-Suk;Walker, Jeffery W.;Chung, Ka-Young
    • Biomolecules & Therapeutics
    • /
    • v.20 no.4
    • /
    • pp.386-392
    • /
    • 2012
  • The endothelin (ET) signaling pathway controls many physiological processes in myocardium and often becomes upregulated in heart diseases. The aim of the present study was to investigate the effects of ET receptor upregulation on the contractile function of adult ventricular myocytes. Primary cultured adult rat ventricular myocytes were used as a model system of ET receptor overexpression in the heart. Endothelin receptor type A ($ET_A$) or type B ($ET_B$) was overexpressed by Adenoviral infection, and the twitch responses of infected ventricular myocytes were measured after ET-1 stimulation. Overexpression of $ET_A$ exaggerated positive inotropic effect (PIE) and diastolic shortening of ET-1, and induced a new twitch response including twitch broadening. On the contrary, overexpression of $ET_B$ increased PIE of ET-1, but did not affect other two twitch responses. Control myocytes expressing endogenous receptors showed a parallel increase in twitch amplitude and systolic $Ca^{2+}$ in response to ET-1. However, intracellular $Ca^{2+}$ did not change in proportion to the changes in contractility in myocytes overexpressing $ET_A$. Overexpression of $ET_A$ enhanced both systolic and diastolic contractility without parallel changes in $Ca^{2+}$. Differential regulation of this nature indicates that upregulation of $ET_A$ may contribute to diastolic myocardial dysfunction by selectively targeting myofilament proteins that regulate resting cell length, twitch duration and responsiveness to prevailing $Ca^{2+}$.

Effects of Scutellariae Radix on Locomotor Dysfunction of Contusive Spinal Cord Injury Induced Rats (황금(黃芩)이 척수압박손상 흰쥐의 운동기능장애에 미치는 영향)

  • Kim, Eun-Seok;Kim, Pyung-Soo;Kim, Bum-Hoi;Shin, Jung-Won;Kim, Seong-Joon;Sohn, Nak-Won
    • Journal of Acupuncture Research
    • /
    • v.28 no.3
    • /
    • pp.33-42
    • /
    • 2011
  • Objectives : This study was performed to evaluate the effects of Scutellariae radix (SR) water extract on locomotor dysfunction induced by spinal cord injury (SCI) in rats. Methods : SCI was induced mechanical contusion following laminectomy of 10 th thoracic vertebra in Sprague-Dawley rats. SR was orally given once a day for 7 days after SCI. Neurological behavior was examined with the Basso-Beattie-Bresnahan locomotor rating scale. Tissue damage and nerve fiber degeneration were examined with cresyl violet and luxol fast blue (LFB) histochemistry. Using immunohistochemistry, cellular damages to neurons and nerve fibers were examined MAP-2. Results : 1. SR significantly ameliorated the locomotor dysfunction of the SCI-induced rats. 2. SR significantly reduced the number of motor neurons in the ventral horn of the SCI-induced rat spinal cord. 3. SR attenuated the reduction of nerve fiber shirnakage and degeneration of the SCI-induced rat spinal cord. 4. SR attenuated the reduction of MAP-2 positive cells in the peri-lesion of the SCI-induced rat spinal cord. Conclusions : These results suggest that SR improves the locomotor dysfunction of SCI by reducing degeneration of nerve fibers and motor neuron shrinkage in the ventral horn.

Actin Dysfunction Induces Cell Cycle Delay at G2/M with Sustained ERK and RSK Activation in IMR-90 Normal Human Fibroblasts

  • Shrestha, Deepmala;Choi, Daeun;Song, Kiwon
    • Molecules and Cells
    • /
    • v.41 no.5
    • /
    • pp.436-443
    • /
    • 2018
  • The actin cytoskeleton plays a key role in the entry of mitosis as well as in cytokinesis. In a previous study, we showed that actin disruption delays mitotic entry at G2/M by sustained activation of extracellular signal-related kinase 1/2 (ERK1/2) in primary cells but not in transformed cancer cell lines. Here, we examined the mechanism of cell cycle delay at G2/M by actin dysfunction in IMR-90 normal human fibroblasts. We observed that de-polymerization of actin with cytochalasin D (CD) constitutively activated ribosomal S6 kinase (RSK) and induced inhibitory phosphorylation of Cdc2 (Tyr 15) in IMR-90 cells. In the presence of an actin defect in IMR-90 cells, activating phosphorylation of Wee1 kinase (Ser 642) and inhibitory phosphorylation of Cdc25C (Ser 216) was also maintained. However, when kinase-dead RSK (DN-RSK) was overexpressed, we observed sustained activation of ERK1/2, but no delay in the G2/M transition, demonstrating that RSK functions downstream of ERK in cell cycle delay by actin dysfunction. In DN-RSK overexpressing IMR-90 cells treated with CD, phosphorylation of Cdc25C (Ser 216) was blocked and phosphorylation of Cdc2 (Tyr 15) was decreased, but the phosphorylation of Wee1 (Ser 642) was maintained, demonstrating that RSK directly controls phosphorylation of Cdc25C (Ser 216), but not the activity of Wee1. These results strongly suggest that actin dysfunction in primary cells activates ERK1/2 to inhibit Cdc2, delaying the cell cycle at G2/M by activating downstream RSK, which phosphorylates and blocks Cdc25C, and by directly activating Wee1.