Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.656
Filter
1.
Clin Exp Rheumatol ; 42(6): 1215-1223, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38966946

ABSTRACT

OBJECTIVES: The pathogenesis of fibromyalgia (FM), characterised by chronic widespread pain and fatigue, remains notoriously elusive, hampering attempts to develop disease modifying treatments. Mitochondria are the headquarters of cellular energy metabolism, and their malfunction has been proposed to contribute to both FM and chronic fatigue. Thus, the aim of the current pilot study, was to detect structural changes in mitochondria of peripheral blood mononuclear cells (PBMCs) of FM patients, using transmission electron microscopy (TEM). METHODS: To detect structural mitochondrial alterations in FM, we analysed PBMCs from seven patients and seven healthy controls, using TEM. Patients were recruited from a specialised Fibromyalgia Clinic at a tertiary medical centre. After providing informed consent, participants completed questionnaires including the widespread pain index (WPI), symptoms severity score (SSS), fibromyalgia impact questionnaire (FIQ), beck depression inventory (BDI), and visual analogue scale (VAS), to verify a diagnosis of FM according to ACR criteria. Subsequently, blood samples were drawn and PBMCs were collected for EM analysis. RESULTS: TEM analysis of PBMCs showed several distinct mitochondrial cristae patterns, including total loss of cristae in FM patients. The number of mitochondria with intact cristae morphology was reduced in FM patients and the percentage of mitochondria that completely lacked cristae was increased. These results correlated with the WPI severity. Moreover, in the FM patient samples we observed a high percentage of cells containing electron dense aggregates, which are possibly ribosome aggregates. Cristae loss and possible ribosome aggregation were intercorrelated, and thus may represent reactions to a shared cellular stress condition. The changes in mitochondrial morphology suggest that mitochondrial dysfunction, resulting in inefficient oxidative phosphorylation and ATP production, metabolic and redox disorders, and increased reactive oxygen species (ROS) levels, may play a pathogenetic role in FM. CONCLUSIONS: We describe novel morphological changes in mitochondria of FM patients, including loss of mitochondrial cristae. While these observations cannot determine whether the changes are pathogenetic or represent an epiphenomenon, they highlight the possibility that mitochondrial malfunction may play a causative role in the cascade of events leading to chronic pain and fatigue in FM. Moreover, the results offer the possibility of utilising changes in mitochondrial morphology as an objective biomarker in FM. Further understanding the connection between FM and dysfunction of mitochondria physiology, may assist in developing both novel diagnostic tools as well as specific treatments for FM, such as approaches to improve/strengthen mitochondria function.


Subject(s)
Fibromyalgia , Mitochondria , Humans , Fibromyalgia/pathology , Fibromyalgia/physiopathology , Pilot Projects , Mitochondria/ultrastructure , Mitochondria/pathology , Female , Middle Aged , Adult , Case-Control Studies , Male , Microscopy, Electron, Transmission , Leukocytes, Mononuclear/ultrastructure , Leukocytes, Mononuclear/pathology , Severity of Illness Index , Pain Measurement
2.
Cell Biochem Funct ; 42(5): e4085, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38951992

ABSTRACT

This review rigorously investigates the early cerebral changes associated with Alzheimer's disease, which manifest long before clinical symptoms arise. It presents evidence that the dysregulation of calcium (Ca2+) homeostasis, along with mitochondrial dysfunction and aberrant autophagic processes, may drive the disease's progression during its asymptomatic, preclinical stage. Understanding the intricate molecular interplay that unfolds during this critical period offers a window into identifying novel therapeutic targets, thereby advancing the treatment of neurodegenerative disorders. The review delves into both established and emerging insights into the molecular alterations precipitated by the disruption of Ca2+ balance, setting the stage for cognitive decline and neurodegeneration.


Subject(s)
Alzheimer Disease , Autophagy , Calcium , Mitochondria , Mitophagy , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mitochondria/metabolism , Mitochondria/pathology , Calcium/metabolism , Animals , Hemostasis , Homeostasis
3.
World J Gastroenterol ; 30(23): 2934-2946, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38946875

ABSTRACT

In this editorial, we comment on an article titled "Morphological and biochemical characteristics associated with autophagy in gastrointestinal diseases", which was published in a recent issue of the World Journal of Gastroenterology. We focused on the statement that "autophagy is closely related to the digestion, secretion, and regeneration of gastrointestinal cells". With advancing research, autophagy, and particularly the pivotal role of the macroautophagy in maintaining cellular equilibrium and stress response in the gastrointestinal system, has garnered extensive study. However, the significance of mitophagy, a unique selective autophagy pathway with ubiquitin-dependent and independent variants, should not be overlooked. In recent decades, mitophagy has been shown to be closely related to the occurrence and development of gastrointestinal diseases, especially inflammatory bowel disease, gastric cancer, and colorectal cancer. The interplay between mitophagy and mitochondrial quality control is crucial for elucidating disease mechanisms, as well as for the development of novel treatment strategies. Exploring the pathogenesis behind gastrointestinal diseases and providing individualized and efficient treatment for patients are subjects we have been exploring. This article reviews the potential mechanism of mitophagy in gastrointestinal diseases with the hope of providing new ideas for diagnosis and treatment.


Subject(s)
Autophagy , Gastrointestinal Diseases , Mitochondria , Mitophagy , Humans , Autophagy/physiology , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/physiopathology , Mitochondria/metabolism , Mitochondria/pathology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/metabolism , Animals
4.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949024

ABSTRACT

Mitochondria-related neurodegenerative diseases have been implicated in the disruption of primary cilia function. Mutation in an intrinsic mitochondrial complex I component NDUFAF2 has been identified in Leigh syndrome, a severe inherited mitochondriopathy. Mutations in ARMC9, which encodes a basal body protein, cause Joubert syndrome, a ciliopathy with defects in the brain, kidney, and eye. Here, we report a mechanistic link between mitochondria metabolism and primary cilia signaling. We discovered that loss of NDUFAF2 caused both mitochondrial and ciliary defects in vitro and in vivo and identified NDUFAF2 as a binding partner for ARMC9. We also found that NDUFAF2 was both necessary and sufficient for cilia formation and that exogenous expression of NDUFAF2 rescued the ciliary and mitochondrial defects observed in cells from patients with known ARMC9 deficiency. NAD+ supplementation restored mitochondrial and ciliary dysfunction in ARMC9-deficient cells and zebrafish and ameliorated the ocular motility and motor deficits of a patient with ARMC9 deficiency. The present results provide a compelling mechanistic link, supported by evidence from human studies, between primary cilia and mitochondrial signaling. Importantly, our findings have significant implications for the development of therapeutic approaches targeting ciliopathies.


Subject(s)
Cilia , Kidney Diseases, Cystic , Leigh Disease , Mitochondria , Zebrafish , Humans , Zebrafish/metabolism , Zebrafish/genetics , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Cilia/metabolism , Cilia/pathology , Cilia/genetics , Animals , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/genetics , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , Retina/metabolism , Retina/pathology , Retina/abnormalities , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Abnormalities/metabolism , Mice , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebellum/abnormalities , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Male
5.
Cancer Res ; 84(14): 2297-2312, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39005053

ABSTRACT

Metabolic reprogramming is a hallmark of cancer and is crucial for cancer progression, making it an attractive therapeutic target. Understanding the role of metabolic reprogramming in cancer initiation could help identify prevention strategies. To address this, we investigated metabolism during acinar-to-ductal metaplasia (ADM), the first step of pancreatic carcinogenesis. Glycolytic markers were elevated in ADM lesions compared with normal tissue from human samples. Comprehensive metabolic assessment in three mouse models with pancreas-specific activation of KRAS, PI3K, or MEK1 using Seahorse measurements, nuclear magnetic resonance metabolome analysis, mass spectrometry, isotope tracing, and RNA sequencing analysis revealed a switch from oxidative phosphorylation to glycolysis in ADM. Blocking the metabolic switch attenuated ADM formation. Furthermore, mitochondrial metabolism was required for de novo synthesis of serine and glutathione (GSH) but not for ATP production. MYC mediated the increase in GSH intermediates in ADM, and inhibition of GSH synthesis suppressed ADM development. This study thus identifies metabolic changes and vulnerabilities in the early stages of pancreatic carcinogenesis. Significance: Metabolic reprogramming from oxidative phosphorylation to glycolysis mediated by MYC plays a crucial role in the development of pancreatic cancer, revealing a mechanism driving tumorigenesis and potential therapeutic targets. See related commentary by Storz, p. 2225.


Subject(s)
Metaplasia , Pancreatic Neoplasms , Animals , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Mice , Metaplasia/metabolism , Metaplasia/pathology , Glycolysis , Carcinogenesis/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Oxidative Phosphorylation , Glutathione/metabolism , Cellular Reprogramming , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Male , Mitochondria/metabolism , Mitochondria/pathology , Metabolic Reprogramming
6.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928291

ABSTRACT

The process of aging inevitably leads to an increase in age-related comorbidities, including chronic kidney disease (CKD). In many aspects, CKD can be considered a state of accelerated and premature aging. Aging kidney and CKD have numerous common characteristic features, ranging from pathological presentation and clinical manifestation to underlying mechanisms. The shared mechanisms underlying the process of kidney aging and the development of CKD include the increase in cellular senescence, the decrease in autophagy, mitochondrial dysfunction, and the alterations of epigenetic regulation, suggesting the existence of potential therapeutic targets that are applicable to both conditions. In this review, we provide a comprehensive overview of the common characteristics between aging kidney and CKD, encompassing morphological changes, functional alterations, and recent advancements in understanding the underlying mechanisms. Moreover, we discuss potential therapeutic strategies for targeting senescent cells in both the aging process and CKD.


Subject(s)
Aging , Cellular Senescence , Epigenesis, Genetic , Kidney , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/etiology , Aging/pathology , Kidney/pathology , Kidney/metabolism , Animals , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/pathology , Autophagy
7.
Technol Cancer Res Treat ; 23: 15330338241258570, 2024.
Article in English | MEDLINE | ID: mdl-38832431

ABSTRACT

Background: Colon adenocarcinoma (COAD) has increasing incidence and is one of the most common malignant tumors. The mitochondria involved in cell energy metabolism, oxygen free radical generation, and cell apoptosis play important roles in tumorigenesis and progression. The relationship between mitochondrial genes and COAD remains largely unknown. Methods: COAD data including 512 samples were set out from the UCSC Xena database. The nuclear mitochondrial-related genes (NMRGs)-related risk prognostic model and prognostic nomogram were constructed, and NMRGs-related gene mutation and the immune environment were analyzed using bioinformatics methods. Then, a liver metastasis model of colorectal cancer was constructed and protein expression was detected using Western blot assay. Results: A prognostic model for COAD was constructed. Comparing the prognostic model dataset and the validation dataset showed considerable correlation in both risk grouping and prognosis. Based on the risk score (RS) model, the samples of the prognostic dataset were divided into high risk group and low risk group. Moreover, pathologic N and T stage and tumor recurrence in the two risk groups were significantly different. The four prognostic factors, including age and pathologic T stage in the nomogram survival model also showed excellent predictive performance. An optimal combination of nine differentially expressed NMRGs was finally obtained, including LARS2, PARS2, ETHE1, LRPPRC, TMEM70, AARS2, ACAD9, VARS2, and ATP8A2. The high-RS group had more inflamed immune features, including T and CD4+ memory cell activation. Besides, mitochondria-associated LRPPRC and LARS2 expression levels were increased in vivo xenograft construction and liver metastases assays. Conclusion: This study established a comprehensive prognostic model for COAD, incorporating nine genes associated with nuclear-mitochondrial functions. This model demonstrates superior predictive performance across four prognostic factors: age, pathological T stage, tumor recurrence, and overall prognosis. It is anticipated to be an effective model for enhancing the prognosis and treatment of COAD.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/mortality , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Mice , Animals , Biomarkers, Tumor/genetics , Nomograms , Computational Biology/methods , Genes, Mitochondrial , Disease Models, Animal , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Liver Neoplasms/pathology , Gene Expression Profiling , Neoplasm Staging , Male , Databases, Genetic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Female
8.
Sci Rep ; 14(1): 13789, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877095

ABSTRACT

Mitochondrial function is critical to continued cellular vitality and is an important contributor to a growing number of human diseases. Mitochondrial dysfunction is typically heterogeneous, mediated through the clonal expansion of mitochondrial DNA (mtDNA) variants in a subset of cells in a given tissue. To date, our understanding of the dynamics of clonal expansion of mtDNA variants has been technically limited to the single cell-level. Here, we report the use of nanobiopsy for subcellular sampling from human tissues, combined with next-generation sequencing to assess subcellular mtDNA mutation load in human tissue from mitochondrial disease patients. The ability to map mitochondrial mutation loads within individual cells of diseased tissue samples will further our understanding of mitochondrial genetic diseases.


Subject(s)
DNA, Mitochondrial , Heteroplasmy , High-Throughput Nucleotide Sequencing , Mutation , Humans , DNA, Mitochondrial/genetics , Heteroplasmy/genetics , High-Throughput Nucleotide Sequencing/methods , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Diseases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology
9.
Arch Dermatol Res ; 316(7): 412, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878082

ABSTRACT

Keloid scars and folliculitis keloidalis nuchae (FKN) are benign fibroproliferative dermal lesions of unknown aetiology and ill-defined treatment, which typically present in genetically susceptible individuals. Their pathognomonic hallmarks include local aggressive invasive behaviour plus high recurrence post-therapy. In view of this, we investigated proliferative and key parameters of bioenergetic cellular characteristics of site-specific keloid-derived fibroblasts (intra(centre)- and peri(margin)-lesional) and FKN compared to normal skin and normal flat non-hypertrophic scar fibroblasts as negative controls.The results showed statistically significant (P < 0.01) and variable growth dynamics with increased proliferation and migration in keloid fibroblasts, while FKN fibroblasts showed a significant (P < 0.001) increase in proliferation but similar migration profile to controls. A statistically significant metabolic switch towards aerobic glycolysis in the fibroblasts from the disease conditions was noted. Furthermore, an increase in basal glycolysis with a concomitant increase in the cellular maximum glycolytic capacity was also demonstrated in perilesional keloid and FKN fibroblasts (P < 0.05). Mitochondrial function parameters showed increased oxidative phosphorylation in the disease conditions (P < 0.05) indicating functional mitochondria. These findings further suggest that Keloids and FKN demonstrate a switch to a metabolic phenotype of aerobic glycolysis. Increased glycolytic flux inhibition is a potential mechanistic basis for future therapy.


Subject(s)
Cell Proliferation , Fibroblasts , Folliculitis , Glycolysis , Keloid , Humans , Keloid/metabolism , Keloid/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Folliculitis/metabolism , Folliculitis/pathology , Mitochondria/metabolism , Mitochondria/pathology , Cells, Cultured , Oxidative Phosphorylation , Cell Movement , Adult , Skin/pathology , Skin/metabolism , Energy Metabolism , Female , Male
10.
PeerJ ; 12: e17551, 2024.
Article in English | MEDLINE | ID: mdl-38887622

ABSTRACT

Background: Keloid is a chronic proliferative fibrotic disease caused by abnormal fibroblasts proliferation and excessive extracellular matrix (ECM) production. Numerous fibrotic disorders are significantly influenced by ferroptosis, and targeting ferroptosis can effectively mitigate fibrosis development. This study aimed to investigate the role and mechanism of ferroptosis in keloid development. Methods: Keloid tissues from keloid patients and normal skin tissues from healthy controls were collected. Iron content, lipid peroxidation (LPO) level, and the mRNA and protein expression of ferroptosis-related genes including solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), transferrin receptor (TFRC), and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined. Mitochondrial morphology was observed using transmission electron microscopy (TEM). Keloid fibroblasts (KFs) were isolated from keloid tissues, and treated with ferroptosis inhibitor ferrostatin-1 (fer-1) or ferroptosis activator erastin. Iron content, ferroptosis-related marker levels, LPO level, mitochondrial membrane potential, ATP content, and mitochondrial morphology in KFs were detected. Furthermore, the protein levels of α-smooth muscle actin (α-SMA), collagen I, and collagen III were measured to investigate whether ferroptosis affect fibrosis in KFs. Results: We found that iron content and LPO level were substantially elevated in keloid tissues and KFs. SLC7A11, GPX4, and Nrf2 were downregulated and TFRC was upregulated in keloid tissues and KFs. Mitochondria in keloid tissues and KFs exhibited ferroptosis-related pathology. Fer-1 treatment reduced iron content, restrained ferroptosis and mitochondrial dysfunction in KFs, Moreover, ferrostatin-1 restrained the protein expression of α-SMA, collagen I, and collagen III in KFs. Whereas erastin treatment showed the opposite results. Conclusion: Ferroptosis exists in keloid. Ferrostatin-1 restrained ECM deposition and fibrosis in keloid through inhibiting ferroptosis, and erastin induced ECM deposition and fibrosis through intensifying ferroptosis.


Subject(s)
Cyclohexylamines , Ferroptosis , Fibroblasts , Fibrosis , Keloid , NF-E2-Related Factor 2 , Phenylenediamines , Phospholipid Hydroperoxide Glutathione Peroxidase , Humans , Ferroptosis/drug effects , Keloid/pathology , Keloid/metabolism , Keloid/drug therapy , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Cyclohexylamines/pharmacology , Fibrosis/metabolism , Fibrosis/pathology , Phenylenediamines/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Male , Lipid Peroxidation/drug effects , Female , Adult , Iron/metabolism , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Piperazines/pharmacology , Actins/metabolism , Actins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Membrane Potential, Mitochondrial/drug effects
11.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892349

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease affecting mostly women of child-bearing age. Immune dysfunction in SLE results from disrupted apoptosis which lead to an unregulated interferon (IFN) stimulation and the production of autoantibodies, leading to immune complex formation, complement activation, and organ damage. Lupus nephritis (LN) is a common and severe complication of SLE, impacting approximately 30% to 40% of SLE patients. Recent studies have demonstrated an alteration in mitochondrial homeostasis in SLE patients. Mitochondrial dysfunction contributes significantly to SLE pathogenesis by enhancing type 1 IFN production through various pathways involving neutrophils, platelets, and T cells. Defective mitophagy, the process of clearing damaged mitochondria, exacerbates this cycle, leading to increased immune dysregulation. In this review, we aim to detail the physiopathological link between mitochondrial dysfunction and disease activity in SLE. Additionally, we will explore the potential role of mitochondria as biomarkers and therapeutic targets in SLE, with a specific focus on LN. In LN, mitochondrial abnormalities are observed in renal cells, correlating with disease progression and renal fibrosis. Studies exploring cell-free mitochondrial DNA as a biomarker in SLE and LN have shown promising but preliminary results, necessitating further validation and standardization. Therapeutically targeting mitochondrial dysfunction in SLE, using drugs like metformin or mTOR inhibitors, shows potential in modulating immune responses and improving clinical outcomes. The interplay between mitochondria, immune dysregulation, and renal involvement in SLE and LN underscores the need for comprehensive research and innovative therapeutic strategies. Understanding mitochondrial dynamics and their impact on immune responses offers promising avenues for developing personalized treatments and non-invasive biomarkers, ultimately improving outcomes for LN patients.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Mitochondria , Humans , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/etiology , Mitochondria/metabolism , Mitochondria/pathology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/immunology , DNA, Mitochondrial/metabolism , Animals , Biomarkers , Mitophagy
12.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892357

ABSTRACT

Currently, there is an increase in the aging of the population, which represents a risk factor for many diseases, including sarcopenia. Sarcopenia involves progressive loss of mass, strength, and function of the skeletal muscle. Some mechanisms include alterations in muscle structure, reduced regenerative capacity, oxidative stress, mitochondrial dysfunction, and inflammation. The zebrafish has emerged as a new model for studying skeletal muscle aging because of its numerous advantages, including histological and molecular similarity to human skeletal muscle. In this study, we used fish of 2, 10, 30, and 60 months of age. The older fish showed a higher frailty index with a value of 0.250 ± 0.000 because of reduced locomotor activity and alterations in biometric measurements. We observed changes in muscle structure with a decreased number of myocytes (0.031 myocytes/µm2 ± 0.004 at 60 months) and an increase in collagen with aging up to 15% ± 1.639 in the 60-month group, corresponding to alterations in the synthesis, degradation, and differentiation pathways. These changes were accompanied by mitochondrial alterations, such as a nearly 50% reduction in the number of intermyofibrillar mitochondria, 100% mitochondrial damage, and reduced mitochondrial dynamics. Overall, we demonstrated a similarity in the aging processes of muscle aging between zebrafish and mammals.


Subject(s)
Aging , Frailty , Muscle, Skeletal , Sarcopenia , Zebrafish , Sarcopenia/metabolism , Sarcopenia/pathology , Animals , Humans , Aging/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Frailty/metabolism , Disease Models, Animal , Mitochondria/metabolism , Mitochondria/pathology
13.
J Pediatr Hematol Oncol ; 46(5): e338-e347, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38857202

ABSTRACT

Primary mitochondrial disorders (PMDs) are known for their pleiotropic manifestations in humans, affecting almost any organ or system at any time. Hematologic manifestations, such as cytopenias and sideroblastic anemia, occur in 10% to 30% of patients with confirmed PMDs. These can be the initial presenting features or complications that develop over time. Surveillance for these manifestations allows for prompt identification and treatment. This article provides an overview of the pathophysiology underpinning the hematologic effects of mitochondrial dysfunction, discussing the 3 key roles of the mitochondria in hematopoiesis: providing energy for cell differentiation and function, synthesizing heme, and generating iron-sulfur clusters. Subsequently, the diagnosis and management of mitochondrial disorders are discussed, focusing on hematologic manifestations and the specific conditions commonly associated with them. Through this, we aimed to provide a concise point of reference for those considering a mitochondrial cause for a patient's hematologic abnormality, or for those considering a hematologic manifestation in a patient with known or suspected mitochondrial disease.


Subject(s)
Hematologic Diseases , Mitochondrial Diseases , Humans , Mitochondrial Diseases/complications , Hematologic Diseases/blood , Hematologic Diseases/complications , Hematologic Diseases/pathology , Mitochondria/pathology , Hematopoiesis , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/therapy
14.
Cells ; 13(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38891059

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a group of sporadic and genetic neurodegenerative disorders that result in losses of upper and lower motor neurons. Treatment of ALS is limited, and survival is 2-5 years after disease onset. While ALS can occur in younger individuals, the risk significantly increases with advancing age. Notably, both sporadic and genetic forms of ALS share pathophysiological features overlapping hallmarks of aging including genome instability/DNA damage, mitochondrial dysfunction, inflammation, proteostasis, and cellular senescence. This review explores chronological and biological aging in the context of ALS onset and progression. Age-related muscle weakness and motor unit loss mirror aspects of ALS pathology and coincide with peak ALS incidence, suggesting a potential link between aging and disease development. Hallmarks of biological aging, including DNA damage, mitochondrial dysfunction, and cellular senescence, are implicated in both aging and ALS, offering insights into shared mechanisms underlying disease pathogenesis. Furthermore, senescence-associated secretory phenotype and senolytic treatments emerge as promising avenues for ALS intervention, with the potential to mitigate neuroinflammation and modify disease progression.


Subject(s)
Aging , Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/therapy , Humans , Aging/pathology , Senotherapeutics/pharmacology , Senotherapeutics/therapeutic use , Animals , Cellular Senescence , Mitochondria/metabolism , Mitochondria/pathology , DNA Damage
15.
Acta Neuropathol Commun ; 12(1): 88, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38840253

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by an expanded CAG repeat in the coding sequence of huntingtin protein. Initially, it predominantly affects medium-sized spiny neurons (MSSNs) of the corpus striatum. No effective treatment is still available, thus urging the identification of potential therapeutic targets. While evidence of mitochondrial structural alterations in HD exists, previous studies mainly employed 2D approaches and were performed outside the strictly native brain context. In this study, we adopted a novel multiscale approach to conduct a comprehensive 3D in situ structural analysis of mitochondrial disturbances in a mouse model of HD. We investigated MSSNs within brain tissue under optimal structural conditions utilizing state-of-the-art 3D imaging technologies, specifically FIB/SEM for the complete imaging of neuronal somas and Electron Tomography for detailed morphological examination, and image processing-based quantitative analysis. Our findings suggest a disruption of the mitochondrial network towards fragmentation in HD. The network of interlaced, slim and long mitochondria observed in healthy conditions transforms into isolated, swollen and short entities, with internal cristae disorganization, cavities and abnormally large matrix granules.


Subject(s)
Disease Models, Animal , Huntington Disease , Imaging, Three-Dimensional , Mitochondria , Animals , Huntington Disease/pathology , Huntington Disease/genetics , Huntington Disease/metabolism , Mitochondria/ultrastructure , Mitochondria/pathology , Mitochondria/metabolism , Imaging, Three-Dimensional/methods , Mice , Mice, Transgenic , Brain/pathology , Brain/ultrastructure , Brain/metabolism , Microscopy, Electron/methods , Male , Neurons/pathology , Neurons/ultrastructure , Neurons/metabolism
16.
Acta Neuropathol Commun ; 12(1): 90, 2024 06 08.
Article in English | MEDLINE | ID: mdl-38851733

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Fibroblasts , Mitochondria , Mitophagy , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Fibroblasts/pathology , Fibroblasts/metabolism , Aged , Female , Mitochondria/pathology , Mitochondria/metabolism , Male , Mitophagy/physiology , Middle Aged , Aged, 80 and over , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Cognitive Dysfunction/pathology , Cognitive Dysfunction/metabolism , Autophagy/physiology
17.
Nature ; 630(8016): 475-483, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839958

ABSTRACT

Senescence is a cellular state linked to ageing and age-onset disease across many mammalian species1,2. Acutely, senescent cells promote wound healing3,4 and prevent tumour formation5; but they are also pro-inflammatory, thus chronically exacerbate tissue decline. Whereas senescent cells are active targets for anti-ageing therapy6-11, why these cells form in vivo, how they affect tissue ageing and the effect of their elimination remain unclear12,13. Here we identify naturally occurring senescent glia in ageing Drosophila brains and decipher their origin and influence. Using Activator protein 1 (AP1) activity to screen for senescence14,15, we determine that senescent glia can appear in response to neuronal mitochondrial dysfunction. In turn, senescent glia promote lipid accumulation in non-senescent glia; similar effects are seen in senescent human fibroblasts in culture. Targeting AP1 activity in senescent glia mitigates senescence biomarkers, extends fly lifespan and health span, and prevents lipid accumulation. However, these benefits come at the cost of increased oxidative damage in the brain, and neuronal mitochondrial function remains poor. Altogether, our results map the trajectory of naturally occurring senescent glia in vivo and indicate that these cells link key ageing phenomena: mitochondrial dysfunction and lipid accumulation.


Subject(s)
Aging , Brain , Cellular Senescence , Drosophila melanogaster , Lipid Metabolism , Mitochondria , Neuroglia , Animals , Female , Humans , Male , Aging/metabolism , Aging/pathology , Brain/metabolism , Brain/pathology , Brain/cytology , Drosophila melanogaster/metabolism , Drosophila melanogaster/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Longevity , Mitochondria/metabolism , Mitochondria/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Transcription Factor AP-1/metabolism , Lipids , Inflammation/metabolism , Inflammation/pathology
18.
Mol Biomed ; 5(1): 24, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38937317

ABSTRACT

Chronic kidney disease (CKD) poses a significant global health dilemma, emerging from complex causes. Although our prior research has indicated that a deficiency in Reticulon-3 (RTN3) accelerates renal disease progression, a thorough examination of RTN3 on kidney function and pathology remains underexplored. To address this critical need, we generated Rtn3-null mice to study the consequences of RTN3 protein deficiency on CKD. Single-cell transcriptomic analyses were performed on 47,885 cells from the renal cortex of both healthy and Rtn3-null mice, enabling us to compare spatial architectures and expression profiles across 14 distinct cell types. Our analysis revealed that RTN3 deficiency leads to significant alterations in the spatial organization and gene expression profiles of renal cells, reflecting CKD pathology. Specifically, RTN3 deficiency was associated with Lars2 overexpression, which in turn caused mitochondrial dysfunction and increased reactive oxygen species levels. This shift induced a transition in renal epithelial cells from a functional state to a fibrogenic state, thus promoting renal fibrosis. Additionally, RTN3 deficiency was found to drive the endothelial-to-mesenchymal transition process and disrupt cell-cell communication, further exacerbating renal fibrosis. Immunohistochemistry and Western-Blot techniques were used to validate these observations, reinforcing the critical role of RTN3 in CKD pathogenesis. The deficiency of RTN3 protein in CKD leads to profound changes in cellular architecture and molecular profiles. Our work seeks to elevate the understanding of RTN3's role in CKD's narrative and position it as a promising therapeutic contender.


Subject(s)
Disease Progression , Fibrosis , Gene Expression Profiling , Renal Insufficiency, Chronic , Single-Cell Analysis , Animals , Mice , Fibrosis/pathology , Fibrosis/metabolism , Fibrosis/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Kidney/pathology , Kidney/metabolism , Transcriptome , Reactive Oxygen Species/metabolism , Epithelial-Mesenchymal Transition/genetics , Disease Models, Animal , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/genetics
19.
Nature ; 630(8017): 720-727, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839949

ABSTRACT

Spermatozoa harbour a complex and environment-sensitive pool of small non-coding RNAs (sncRNAs)1, which influences offspring development and adult phenotypes1-7. Whether spermatozoa in the epididymis are directly susceptible to environmental cues is not fully understood8. Here we used two distinct paradigms of preconception acute high-fat diet to dissect epididymal versus testicular contributions to the sperm sncRNA pool and offspring health. We show that epididymal spermatozoa, but not developing germ cells, are sensitive to the environment and identify mitochondrial tRNAs (mt-tRNAs) and their fragments (mt-tsRNAs) as sperm-borne factors. In humans, mt-tsRNAs in spermatozoa correlate with body mass index, and paternal overweight at conception doubles offspring obesity risk and compromises metabolic health. Sperm sncRNA sequencing of mice mutant for genes involved in mitochondrial function, and metabolic phenotyping of their wild-type offspring, suggest that the upregulation of mt-tsRNAs is downstream of mitochondrial dysfunction. Single-embryo transcriptomics of genetically hybrid two-cell embryos demonstrated sperm-to-oocyte transfer of mt-tRNAs at fertilization and suggested their involvement in the control of early-embryo transcription. Our study supports the importance of paternal health at conception for offspring metabolism, shows that mt-tRNAs are diet-induced and sperm-borne and demonstrates, in a physiological setting, father-to-offspring transfer of sperm mitochondrial RNAs at fertilization.


Subject(s)
Diet, High-Fat , Epigenesis, Genetic , Mitochondria , RNA, Mitochondrial , Spermatozoa , Animals , Female , Humans , Male , Mice , Body Mass Index , Diet, High-Fat/adverse effects , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Epididymis/cytology , Epigenesis, Genetic/genetics , Fertilization/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Obesity/genetics , Obesity/metabolism , Obesity/etiology , Oocytes/metabolism , Overweight/genetics , Overweight/metabolism , Paternal Inheritance/genetics , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Spermatozoa/metabolism , Testis/cytology , Transcription, Genetic
20.
Neurosci Biobehav Rev ; 163: 105767, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38885889

ABSTRACT

Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.


Subject(s)
Mitochondria , Multiple Sclerosis , Nogo Proteins , Humans , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Mitochondria/metabolism , Mitochondria/pathology , Nogo Proteins/metabolism , Animals , Signal Transduction/physiology , Axons/metabolism , Axons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...