scispace - formally typeset
Search or ask a question

How does mitochondrial biogenesis dysfunction effects on cellular homeostasis and physiology? 


Best insight from top research papers

Mitochondrial biogenesis dysfunction significantly impacts cellular homeostasis and physiology. Impaired protein import into mitochondria due to various stressors leads to the accumulation of precursor proteins in the cytosol, activating stress response pathways. This disruption triggers a network of processes involving transcription, translation, and protein clearance to restore cellular protein homeostasis. Additionally, mitochondrial dysfunction, particularly protein import defects, results in the formation of aberrant protein deposits in the cytosol, leading to the aggregation of disease-related proteins like α-synuclein and amyloid β, causing a cytosolic protein homeostasis imbalance. Mitochondrial dysfunction also affects cellular functions like oxidative phosphorylation, calcium homeostasis, and biomolecule synthesis, ultimately impacting overall cellular health.

Answers from top 5 papers

More filters
Papers (5)Insight
Mitochondrial biogenesis dysfunction disrupts cellular proteostasis, leading to impaired mitochondrial function, increased reactive oxygen species, and potential cell death, impacting cellular homeostasis and physiology.
Mitochondrial biogenesis dysfunction disrupts cellular homeostasis, leading to metabolic diseases and aging. Exercise helps prevent and delay such dysfunction by enhancing mitochondrial metabolism and quality control mechanisms.
Mitochondrial protein import failures lead to cytosolic aggregation, disrupting protein homeostasis. This triggers further aggregation of mitochondrial and disease-related proteins, impacting cellular homeostasis and potentially contributing to neurodegenerative diseases.
Mitochondrial biogenesis dysfunction disrupts cellular homeostasis by impairing functions like oxidative phosphorylation, calcium regulation, and biomolecule synthesis, impacting overall cellular physiology and contributing to neurodegenerative conditions.
Mitochondrial biogenesis dysfunction disrupts cellular protein homeostasis, triggering stress responses involving transcription, translation, and protein clearance mechanisms to restore cellular equilibrium.

Related Questions

What is the role of mitochondrial biogenesis in atherosclerosis progression?4 answersMitochondrial biogenesis plays a pivotal role in the progression of atherosclerosis through several interconnected mechanisms. The process of mitochondrial dynamics, which includes biogenesis, is crucial for maintaining cardiovascular homeostasis. Impairment in these dynamics can lead to myocardial damage and cardiac disease progression, suggesting that mitochondrial biogenesis is a fundamental process in maintaining the integrity and function of mitochondria in the cardiovascular system. Mitochondrial dysfunction, characterized by altered biogenesis, contributes to the initiation and progression of atherosclerosis by elevating the production of reactive oxygen species (ROS), altering mitochondrial dynamics, and promoting inflammation. This dysfunction affects arterial wall cells, including macrophages and endothelial cells, causing oxidative stress, chronic inflammation, and intracellular lipid deposition, which are key factors in atherosclerotic plaque progression. Furthermore, mitochondrial homeostasis, maintained through biogenesis, is essential for many cellular processes. Under stress conditions, disruptions in mitochondrial dynamics can lead to increased ROS production, inflammation, and tissue damage, which are closely related to the progression of cardiovascular diseases. Vascular smooth muscle cell (VSMC) proliferation and migration, critical events in atherosclerosis, are also influenced by mitochondrial dynamics and biogenesis. Altered mitochondrial function, potentially stemming from compromised biogenesis, is recognized as an important factor in atherosclerosis initiation and progression. Innovative therapeutic strategies, such as mitochondrial transplantation, aim to regulate macrophage bioenergetics by improving mitochondrial function, thereby attenuating inflammatory processes and preventing plaque progression. The identification of mitochondrial DNA (mtDNA) mutations associated with atherosclerosis underscores the importance of mitochondrial biogenesis in the disease's pathogenesis. Understanding the role of mitochondrial metabolism in cardiovascular disease progression highlights the significance of mitochondrial biogenesis in developing preventive and therapeutic methods. Lastly, mitochondrial dysfunction, including impaired biogenesis, contributes to lesion development and progression in atherosclerosis by affecting cellular homeostasis and metabolic functions.
How does mitochondrial function impact cellular metabolism and energy production?4 answersMitochondria play a crucial role in cellular metabolism and energy production. These organelles are known as the cellular powerhouses due to their involvement in oxidative phosphorylation (OXPHOS), which is the primary process for energy generation. Mitochondria communicate with the nucleus and other organelles to maintain cellular homeostasis, adapt to stress, and influence cell fate decisions. Dysregulation of mitochondrial function is linked to various prevalent diseases like type 2 diabetes, cardiovascular disease, cancer, and neurodegenerative disorders. Additionally, mitochondria are essential for reactive oxygen species production, fatty acid metabolism, and epigenetic remodeling, impacting stem cell fate decisions and tissue regeneration. The balance between glycolysis and OXPHOS is crucial for ATP generation, with mitochondrial dysfunction contributing significantly to disease pathogenesis.
Mitochondrial biogenesis is a potential target for OA treatment?5 answersMitochondrial biogenesis has been identified as a potential target for the treatment of osteoarthritis (OA). Recent studies have shown that mitochondrial dysfunction plays a significant role in the pathogenesis of OA, and maintaining normal mitochondrial quantity and function is crucial for chondrocytes. Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) is the central regulator of mitochondrial biogenesis, and a regulatory network involving various signaling molecules has been reported. Additionally, substances such as puerarin and omentin-1 have shown the potential to activate damaged mitochondrial biogenesis in OA chondrocytes and retard the development of OA. Furthermore, research on exogenous drugs and factors like stem cells and exosomes, which can improve mitochondrial function, has been conducted. Therefore, targeting mitochondrial biogenesis may offer new therapeutic strategies for the treatment of OA.
What role does mitochondrial biogenesis play in OA?5 answersMitochondrial biogenesis plays an important role in the pathogenesis of osteoarthritis (OA). It has been shown that mitochondria are significant energy metabolic centers in chondrocytes, and dysfunction of mitochondria is considered an essential mechanism in the development of OA. Mitochondrial biogenesis is a key process that maintains the normal quantity and function of mitochondria, and the central regulator of this process is peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α). There is a regulatory network of mitochondrial biogenesis involving various signaling molecules, such as adenosine monophosphate-activated protein kinase, sirtuin1/3, and cyclic adenosine monophosphate response element-binding protein. Furthermore, substances like puerarin and omentin-1 have shown potential in activating damaged mitochondrial biogenesis in OA chondrocytes, suggesting their potential use in OA treatment. The mitochondrial genetic background also influences the prevalence, severity, incidence, and progression of OA, possibly through the regulation of energy production, reactive oxygen species generation, apoptosis, and inflammation. Therefore, mitochondrial biogenesis is a promising therapeutic target for OA.
Does mitochondrial dysfunction lead to chondrocyte dysfunction?5 answersMitochondrial dysfunction has been shown to contribute to chondrocyte dysfunction in osteoarthritis (OA). Studies have demonstrated that mitochondrial dysfunction in chondrocytes is associated with OA and can lead to increased chondrocyte apoptosis, decreased type II collagen secretion, and cartilage degeneration. Furthermore, mitochondrial dysfunction-induced activation of the catabolic response in chondrocytes has been observed, resulting in the expression of catabolic genes involved in OA pathogenesis. Mechanical stress, a major risk factor for OA, has also been shown to affect mitochondrial function in chondrocytes, with moderate stress maintaining mitochondrial function and reducing apoptosis, while excessive stress leading to mitochondrial dysfunction and apoptosis. These findings highlight the importance of mitochondrial homeostasis in chondrocyte function and suggest that targeting mitochondrial dysfunction may be a potential therapeutic strategy for OA management.
Does mitochondrial dysfunction contribute to the development of diet-induced insulin resistance?2 answersMitochondrial dysfunction has been shown to contribute to the development of diet-induced insulin resistance. Studies have demonstrated that impaired mitochondrial function, including reduced ATP production, citrate synthase activity, and mitochondrial respiration, occurs coincidently with insulin resistance in skeletal muscle. Additionally, mitochondrial dysfunction has been associated with insulin resistance in adipose tissue, liver, and skeletal muscle, which are all involved in food metabolism. Furthermore, improving mitochondrial function has been found to attenuate insulin resistance, suggesting that targeting mitochondrial dysfunction may have therapeutic potential for treating insulin resistance. The toxic effects of drugs and pollutants on mitochondria have also been reported to potentially compromise insulin sensitivity, highlighting the importance of understanding drug-induced mitochondrial toxicity in the development of insulin resistance. In summary, the evidence suggests that mitochondrial dysfunction plays a significant role in the development of diet-induced insulin resistance.

See what other people are reading

Can DNAJB1 bind to tau amyloids?
5 answers
DNAJB1, a chaperone protein, has been primarily associated with binding to Huntingtin (HTT) amyloids. However, other DNAJB proteins, such as DNAJB6 and DNAJB8, have been shown to interact with disease-related protein aggregates and prevent their formation. DNAJB6b, for instance, plays a crucial role in preventing aggregation of intrinsically disordered proteins. While DNAJB1's specific interaction with tau amyloids is not explicitly mentioned in the provided contexts, the broader role of DNAJB proteins in suppressing protein aggregation suggests that DNAJB1 may have the potential to bind to tau amyloids, similar to its interactions with other amyloidogenic proteins. Further research focusing on DNAJB1's interaction with tau amyloids specifically would be beneficial to confirm this potential binding capability.
Ampk activation in L6 cells via mitochondria
4 answers
AMPK activation in L6 cells can be linked to mitochondria through various mechanisms. AMPK activation, induced by compounds like AICAR and caffeine, has been shown to increase mitochondrial biogenesis in L6 myotubes. Additionally, acetic acid has been found to enhance glucose uptake and fatty acid metabolism in skeletal muscle cells through AMPK activation, leading to increased expression of mitochondrial markers like GLUT4 and myoglobin. Furthermore, flavonoids from Psidium guajava leaves have demonstrated significant AMPK activation in L6 cells, suggesting a potential role in regulating mitochondrial function. Overall, AMPK activation in L6 cells appears to play a crucial role in promoting mitochondrial biogenesis and metabolic adaptations, highlighting the intricate connection between AMPK signaling and mitochondrial function in these cells.
What is the role of mitochondrial ribosomal proteins in cancer development?
5 answers
Mitochondrial ribosomal proteins play a crucial role in cancer development by influencing energy metabolism, apoptosis regulation, and protein synthesis. These proteins are involved in synthesizing mitochondrial inner membrane oxidative phosphorylation (OXPHOS)-related proteins, contributing to tumor cell survival in challenging conditions. Alterations in the expression of mitochondrial ribosomal proteins have been linked to changes in oxidative energy metabolism, tumor heterogeneity, and epithelial-mesenchymal transition markers in breast cancer. Additionally, the dysregulation of mitochondrial ribosomal proteins is associated with tumorigenesis and can serve as diagnostic and prognostic markers in various types of cancer. Understanding the functions and expressions of these proteins provides insights into cancer progression, highlighting their potential as biomarkers and therapeutic targets in cancer treatment.
How does the UPR of mitochondria recognize unfolded proteins?
5 answers
The recognition of unfolded proteins by the mitochondrial unfolded protein response (UPRmt) involves the integration of signals from mitochondrial misfolding stress (MMS) into the nucleus. The UPRmt is activated by the release of two key signals: cytosolic-mitochondrial reactive oxygen species (mtROS) and the accumulation of mitochondrial protein precursors in the cytosol (c-mtProt). Mitochondrial stress leads to the release of mtROS into the cytosol and causes defects in mitochondrial protein import, resulting in c-mtProt accumulation. These signals converge to activate the UPRmt pathway, where mtROS oxidize cytosolic HSP40 protein DNAJA1, enhancing the recruitment of cytosolic HSP70 to c-mtProt. This ultimately leads to the translocation of HSF1 to the nucleus, initiating the transcription of UPRmt genes.
How does the mtUPR correlate to aging?
5 answers
The mitochondrial unfolded protein response (mtUPR) plays a crucial role in aging by promoting cell homeostasis and improving lifespan. Activation of the mtUPR through mild intracellular stress leads to an overcompensation mechanism that enhances proteostasis and antioxidant systems, ultimately benefiting longevity and mitigating age-related diseases. Studies have shown that interventions like MnSOD overexpression, hormesis-type environmental stress, and knockdown of ETC and MRP genes activate the mtUPR, indicating a common mechanism in extending lifespan across species. Additionally, the mtUPR is implicated in mitochondrial dysfunction, a key factor in aging and age-related diseases, highlighting its significance in maintaining cellular fitness and survival throughout the aging process.
What is the relationship between brain mitochondrial function and mental health outcomes?
5 answers
Brain mitochondrial function plays a crucial role in mental health outcomes. Mitochondria are essential for energy synthesis, reactive oxygen species regulation, and neurotransmission, with dysfunction implicated in various mental disorders. Mitochondrial abnormalities and oxidative stress are linked to neuropsychiatric conditions like schizophrenia and bipolar disorder, affecting cognitive processes and behavior. Studies on children with mitochondrial disease show impairments in cognitive and mental health functioning, emphasizing the importance of assessing these aspects in patient care. In mood disorders like major depressive disorder and bipolar disorder, lower brain cytochrome-c-oxidase activity and abnormal mitochondrial parameters are associated with depression severity, potentially serving as biomarkers for antidepressant effects. Mitochondrial disturbances are observed in individuals with mood disorders, highlighting the intricate relationship between mitochondrial function and mental health.
What are the specific molecular mechanisms by which HSP40 contributes to the development of ALS?
5 answers
HSP40, a member of the heat shock protein family, plays a crucial role in ALS development through its involvement in protein homeostasis. Studies have shown that HSP40 proteins assist in protein folding, clearance of degraded proteins, and maintenance of cellular proteostasis. In ALS, the presence of ultra-rare protein-truncating variants in genes like DNAJC7, a member of the HSP40 family, has been observed, indicating a potential link to disease pathology. Furthermore, disruptions in intracellular Ca2+ homeostasis and mitochondrial dysfunction are prominent features in ALS pathogenesis. HSP40's function in facilitating protein folding and degradation processes is crucial in preventing misfolding and accumulation of proteins, which are key factors in neurodegenerative diseases like ALS.
What are the metabolic pathways that areactivated during football practice and compitition?
5 answers
Football practice and competition activate various metabolic pathways. Studies have shown that football participation induces changes in metabolites related to xanthine, fatty acid (acyl choline), medium chain fatty acid, primary bile acid, and glycolysis, gluconeogenesis, and pyruvate metabolism. Additionally, tyrosine metabolism is significantly altered during soccer matches, affecting pathways such as homogentisate, DOPA, and hawkinsin. Long-term training in football players enhances antioxidant defenses and mitochondrial biogenesis through pathways involving NF-κB, PGC1α, and UCPs. The external load in football training is associated with metabolic changes in steroid hormone biosynthesis, tyrosine, tryptophan metabolism, and phenylalanine pathways. These findings collectively suggest that football practice and competition trigger a complex interplay of metabolic pathways to meet the energetic demands and adapt to the physical stresses of the sport.
What are downsides of ethanol treatment on cancer cells?
5 answers
Ethanol treatment poses several downsides on cancer cells. Firstly, ethanol induces oxidative stress in cancer-associated fibroblasts, promoting myofibroblastic differentiation and autophagy in the tumor microenvironment. This oxidative stress leads to ketone production in fibroblasts and ketone re-utilization in epithelial cancer cells, fueling tumor growth via mitochondrial metabolism. Additionally, heavy alcohol consumption increases cancer stem cell (CSC) populations in various cancers, potentially enhancing tumor promotion and aggressiveness. Ethanol's metabolism generates free radicals that interfere with essential cellular functions, affecting growth and differentiation processes, including disturbances in CSCs. Furthermore, ethanol treatment alters protein expression in fibroblast cells, resembling patterns seen in nasopharyngeal carcinoma-associated stromal fibroblasts, hinting at potential cancer-related pathways being targeted by ethanol.
Does insulin inhibit mitochondrial function?
4 answers
Insulin plays a crucial role in regulating mitochondrial function. Insulin deficiency, as seen in diabetes, can lead to decreased mitochondrial ATP production and altered mitochondrial fusion and fission protein levels in various brain regions. Both insulin deficiency and insulin resistance in diabetes are associated with impaired mitochondrial function and increased reactive oxygen species (ROS) production. In skeletal muscle, acute insulin deprivation reduces mitochondrial oxidative capacity and increases mitochondrial-derived H2O2 emission, affecting fatty acid oxidation regulation. Insulin impairment in type 1 and insulin resistance in type 2 diabetes can disrupt mitochondrial biogenesis and function in organs like the liver, heart, kidneys, and brain. Additionally, insulin can enhance mitochondrial biogenesis and function in hepatocytes, with butyrate promoting mitochondrial energy metabolism under high insulin conditions.
How is the influence of physical exercise training on HIF activation?
5 answers
Physical exercise training has a significant impact on hypoxia-inducible factor (HIF) activation. Studies show that exercise stimulates and activates the HIF-1 pathway, including vascular endothelial growth factor (VEGF) signaling, leading to angiogenesis and increased capillary growth. Endurance training under hypoxic conditions enhances the activity of the HIF pathway in skeletal muscle, improving aspects of muscle oxygen transport and metabolism. Additionally, increased levels of reactive oxygen species (ROS) due to exercise induce the expression of PGC-1α, which regulates mitochondrial biogenesis and increases VEGF expression, promoting angiogenesis independently of HIF-1α. Activation of the HIF pathway, even without exposure to atmospheric hypoxia, enhances the effects of running training, indicating the importance of combining training with HIF activation for maximal training benefits.