Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 49
2.
Nature ; 622(7983): 627-636, 2023 Oct.
Article En | MEDLINE | ID: mdl-37821702

Senescent cells drive age-related tissue dysfunction partially through the induction of a chronic senescence-associated secretory phenotype (SASP)1. Mitochondria are major regulators of the SASP; however, the underlying mechanisms have not been elucidated2. Mitochondria are often essential for apoptosis, a cell fate distinct from cellular senescence. During apoptosis, widespread mitochondrial outer membrane permeabilization (MOMP) commits a cell to die3. Here we find that MOMP occurring in a subset of mitochondria is a feature of cellular senescence. This process, called minority MOMP (miMOMP), requires BAX and BAK macropores enabling the release of mitochondrial DNA (mtDNA) into the cytosol. Cytosolic mtDNA in turn activates the cGAS-STING pathway, a major regulator of the SASP. We find that inhibition of MOMP in vivo decreases inflammatory markers and improves healthspan in aged mice. Our results reveal that apoptosis and senescence are regulated by similar mitochondria-dependent mechanisms and that sublethal mitochondrial apoptotic stress is a major driver of the SASP. We provide proof-of-concept that inhibition of miMOMP-induced inflammation may be a therapeutic route to improve healthspan.


Apoptosis , Cellular Senescence , Cytosol , DNA, Mitochondrial , Mitochondria , Animals , Mice , Cytosol/metabolism , DNA, Mitochondrial/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Transmembrane Permeability-Driven Necrosis , Proof of Concept Study , Inflammation/metabolism , Phenotype , Longevity , Healthy Aging
4.
Subcell Biochem ; 102: 77-98, 2023.
Article En | MEDLINE | ID: mdl-36600130

Mitochondria are subcellular organelles present in most eukaryotic cells which play a significant role in numerous aspects of cell biology. These include carbohydrate and fatty acid metabolism to generate cellular energy through oxidative phosphorylation, apoptosis, cell signalling, haem biosynthesis and reactive oxygen species production. Mitochondrial dysfunction is a feature of many human ageing tissues, and since the discovery that mitochondrial DNA mutations were a major underlying cause of changes in oxidative phosphorylation capacity, it has been proposed that they have a role in human ageing. However, there is still much debate on whether mitochondrial DNA mutations play a causal role in ageing or are simply a consequence of the ageing process. This chapter describes the structure of mammalian mitochondria, and the unique features of mitochondrial genetics, and reviews the current evidence surrounding the role of mitochondrial DNA mutations in the ageing process. It then focusses on more recent discoveries regarding the role of mitochondrial dysfunction in stem cell ageing and age-related inflammation.


Aging , DNA, Mitochondrial , Animals , Humans , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Aging/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Reactive Oxygen Species/metabolism , Oxidative Stress , Mammals/genetics
5.
Mol Oncol ; 16(18): 3276-3294, 2022 09.
Article En | MEDLINE | ID: mdl-35842901

Advancing age is a major risk factor for malignant transformation and the development of cancer. As such, over 50% of neoplasms occur in individuals over the age of 70. The pathologies of both ageing and cancer have been characterized by respective groups of molecular hallmarks, and while some features are divergent between the two pathologies, several are shared. Perturbed mitochondrial function is one such common hallmark, and this observation therefore suggests that mitochondrial alterations may be of significance in age-related cancer development. There is now considerable evidence documenting the accumulation of somatic mitochondrial DNA (mtDNA) mutations in ageing human postmitotic and replicative tissues. Similarly, mutations of the mitochondrial genome have been reported in human cancers for decades. The plethora of functions in which mitochondria partake, such as oxidative phosphorylation, redox balance, apoptosis and numerous biosynthetic pathways, manifests a variety of ways in which alterations in mtDNA may contribute to tumour growth. However, the specific mechanisms by which mtDNA mutations contribute to tumour progression remain elusive and often contradictory. This review aims to consolidate current knowledge and describe future direction within the field.


DNA, Mitochondrial , Neoplasms , Aging/genetics , Aging/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Mitochondria/metabolism , Mutation/genetics , Neoplasms/pathology
6.
Sci Rep ; 12(1): 6660, 2022 04 22.
Article En | MEDLINE | ID: mdl-35459777

Advances in multiplex immunofluorescence (mIF) and digital image analysis has enabled simultaneous assessment of protein defects in electron transport chain components. However, current manual methodology is time consuming and labour intensive. Therefore, we developed an automated high-throughput mIF workflow for quantitative single-cell level assessment of formalin fixed paraffin embedded tissue (FFPE), leveraging tyramide signal amplification on a Ventana Ultra platform coupled with automated multispectral imaging on a Vectra 3 platform. Utilising this protocol, we assessed the mitochondrial oxidative phosphorylation (OXPHOS) protein alterations in a cohort of benign and malignant prostate samples. Mitochondrial OXPHOS plays a critical role in cell metabolism, and OXPHOS perturbation is implicated in carcinogenesis. Marked inter-patient, intra-patient and spatial cellular heterogeneity in OXPHOS protein abundance was observed. We noted frequent Complex IV loss in benign prostate tissue and Complex I loss in age matched prostate cancer tissues. Malignant regions within prostate cancer samples more frequently contained cells with low Complex I & IV and high mitochondrial mass in comparison to benign-adjacent regions. This methodology can now be applied more widely to study the frequency and distribution of OXPHOS alterations in formalin-fixed tissues, and their impact on long-term clinical outcomes.


Fluorescent Antibody Technique , Prostate , Prostatic Neoplasms , Electron Transport Complex IV , Fluorescent Antibody Technique/methods , Formaldehyde , Humans , Male , Oxidative Phosphorylation , Paraffin Embedding , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Tissue Fixation
7.
Mol Metab ; 60: 101489, 2022 06.
Article En | MEDLINE | ID: mdl-35390502

OBJECTIVE: There is strong evidence that mitochondrial DNA mutations and mitochondrial dysfunction play a role in diabetes pathogenesis. The homozygous knock-in mtDNA mutator mouse is a model of premature aging due to the accumulation of mitochondrial DNA mutations. We used this mouse model to investigate the relationship between mitochondrial subunit expression and pancreatic islet cell composition. METHODS: Quadruple immunofluorescence was used to quantify mitochondrial subunit expression (complex I and IV) and cell composition in pancreatic islets from mitochondrial DNA mutator mice (PolgAmut/mut) and control C57BL/6 mice at 12 and 44 weeks of age. RESULTS: Mitochondrial complex I subunit expression was decreased in islets from 12 week PolgAmut/mut mice. This complex I deficiency persisted with age and was associated with decreased insulin staining intensity at 44 weeks. Complex I deficiency was greater in α-cells compared with ß-cells in islets from 44 week PolgAmut/mut mice. Islet cell composition was normal in 12 week PolgAmut/mut mice, but the ß: α cell ratio was decreased in islets from 44 week PolgAmut/mut mice. This was due to an increase in α-cell number linked to an increase in α-cell proliferation. CONCLUSION: Complex I deficiency promotes α-cell proliferation and alters islet cell composition.


Mitochondrial Diseases , Animals , Cell Proliferation , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Electron Transport Complex I/deficiency , Mice , Mice, Inbred C57BL
8.
Aging Cell ; 20(3): e13321, 2021 03.
Article En | MEDLINE | ID: mdl-33626245

One of the hallmarks of aging is an accumulation of cells with defects in oxidative phosphorylation (OXPHOS) due to mutations of mitochondrial DNA (mtDNA). Rapidly dividing tissues maintained by stem cells, such as the colonic epithelium, are particularly susceptible to accumulation of OXPHOS defects over time; however, the effects on the stem cells are unknown. We have crossed a mouse model in which intestinal stem cells are labelled with EGFP (Lgr5-EGFP-IRES-creERT2) with a model of accelerated mtDNA mutagenesis (PolgAmut/mut ) to investigate the effect of OXPHOS dysfunction on colonic stem cell proliferation. We show that a reduction in complex I protein levels is associated with an increased rate of stem cell cycle re-entry. These changes in stem cell homeostasis could have significant implications for age-associated intestinal pathogenesis.


Aging/pathology , Colon/pathology , Electron Transport Complex I/deficiency , Mitochondrial Diseases/pathology , Stem Cells/pathology , Animals , Cell Proliferation , Female , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Phosphorylation , Thymidine/metabolism
10.
Nat Cancer ; 1(10): 976-989, 2020 10.
Article En | MEDLINE | ID: mdl-33073241

Oxidative phosphorylation (OXPHOS) defects caused by somatic mitochondrial DNA (mtDNA) mutations increase with age in human colorectal epithelium and are prevalent in colorectal tumours, but whether they actively contribute to tumorigenesis remains unknown. Here we demonstrate that mtDNA mutations causing OXPHOS defects are enriched during the human adenoma/carcinoma sequence, suggesting they may confer a metabolic advantage. To test this we deleted the tumour suppressor Apc in OXPHOS deficient intestinal stem cells in mice. The resulting tumours were larger than in control mice due to accelerated cell proliferation and reduced apoptosis. We show that both normal crypts and tumours undergo metabolic remodelling in response to OXPHOS deficiency by upregulating the de novo serine synthesis pathway (SSP). Moreover, normal human colonic crypts upregulate the SSP in response to OXPHOS deficiency prior to tumorigenesis. Our data show that age-associated OXPHOS deficiency causes metabolic remodelling that can functionally contribute to accelerated intestinal cancer development.


Intestinal Neoplasms , Mitochondrial Diseases , Animals , Cell Transformation, Neoplastic/genetics , DNA, Mitochondrial/genetics , Intestinal Neoplasms/genetics , Mice , Mitochondria/genetics , Mutation
11.
Sci Rep ; 10(1): 11643, 2020 07 15.
Article En | MEDLINE | ID: mdl-32669663

The pathogenesis of declining bone mineral density, a universal feature of ageing, is not fully understood. Somatic mitochondrial DNA (mtDNA) mutations accumulate with age in human tissues and mounting evidence suggests that they may be integral to the ageing process. To explore the potential effects of mtDNA mutations on bone biology, we compared bone microarchitecture and turnover in an ageing series of wild type mice with that of the PolgAmut/mut mitochondrial DNA 'mutator' mouse. In vivo analyses showed an age-related loss of bone in both groups of mice; however, it was significantly accelerated in the PolgAmut/mut mice. This accelerated rate of bone loss is associated with significantly reduced bone formation rate, reduced osteoblast population densities, increased osteoclast population densities, and mitochondrial respiratory chain deficiency in osteoblasts and osteoclasts in PolgAmut/mut mice compared with wild-type mice. In vitro assays demonstrated severely impaired mineralised matrix formation and increased osteoclast resorption by PolgAmut/mut cells. Finally, application of an exercise intervention to a subset of PolgAmut/mut mice showed no effect on bone mass or mineralised matrix formation in vitro. Our data demonstrate that mitochondrial dysfunction, a universal feature of human ageing, impairs osteogenesis and is associated with accelerated bone loss.


Aging/genetics , Bone Resorption/genetics , DNA Polymerase gamma/genetics , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Animals , Bone Density/physiology , Bone Resorption/metabolism , Bone Resorption/physiopathology , Calcification, Physiologic , Cell Count , DNA Polymerase gamma/deficiency , DNA, Mitochondrial/metabolism , Electron Transport Complex I/deficiency , Electron Transport Complex I/genetics , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Female , Femur/metabolism , Femur/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Mutation , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoporosis/metabolism , Osteoporosis/physiopathology , Physical Conditioning, Animal
12.
Biogerontology ; 21(4): 445-459, 2020 08.
Article En | MEDLINE | ID: mdl-31802313

Alterations in mitochondrial metabolism have been described as one of the major hallmarks of both ageing cells and cancer. Age is the biggest risk factor for the development of a significant number of cancer types and this therefore raises the question of whether there is a link between age-related mitochondrial dysfunction and the advantageous changes in mitochondrial metabolism prevalent in cancer cells. A common underlying feature of both ageing and cancer cells is the presence of somatic mutations of the mitochondrial genome (mtDNA) which we postulate may drive compensatory alterations in mitochondrial metabolism that are advantageous for tumour growth. In this review, we discuss basic mitochondrial functions, mechanisms of mtDNA mutagenesis and their metabolic consequences, and review the evidence for and against a role for mtDNA mutations in cancer development.


Aging , Mitochondria , Neoplasms , Aging/pathology , Cellular Senescence , DNA, Mitochondrial/genetics , Humans , Mitochondria/genetics , Mitochondria/pathology , Mutation , Neoplasms/pathology
13.
EMBO J ; 38(5)2019 03 01.
Article En | MEDLINE | ID: mdl-30737259

Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age-related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post-mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age-related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent-like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length-independent telomere damage in cardiomyocytes activates the classical senescence-inducing pathways, p21CIP and p16INK4a, and results in a non-canonical senescence-associated secretory phenotype, which is pro-fibrotic and pro-hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age-related myocardial dysfunction and in the wider setting to ageing in post-mitotic tissues.


Cardiomegaly/pathology , Cellular Senescence , DNA Damage , Fibrosis/pathology , Mitosis , Myocytes, Cardiac/pathology , Telomere Shortening , Aging , Animals , Cardiomegaly/etiology , Female , Fibrosis/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monoamine Oxidase/physiology , Myocytes, Cardiac/metabolism , Phenotype , RNA/physiology , Rats, Sprague-Dawley , Telomerase/physiology
14.
J Pathol ; 246(4): 427-432, 2018 12.
Article En | MEDLINE | ID: mdl-30146801

Inherited mitochondrial DNA (mtDNA) mutations cause mitochondrial disease, but mtDNA mutations also occur somatically and accumulate during ageing. Studies have shown that the mutation load of some inherited mtDNA mutations decreases over time in blood, suggesting selection against the mutation. However, it is unknown whether such selection occurs in other mitotic tissues, and where it occurs within the tissue. Gastrointestinal epithelium is a canonical mitotic tissue rapidly renewed by stem cells. Intestinal crypts (epithelium) undergo monoclonal conversion with a single stem cell taking over the niche and producing progeny. We show: (1) that there is a significantly lower mtDNA mutation load in the mitotic epithelium of the gastrointestinal tract when compared to the smooth muscle in the same tissue in patients with the pathogenic m.3243A>G and m.8344A>G mutations; (2) that there is considerable variation seen in individual crypts, suggesting changes in the stem cell population; (3) that this lower mutation load is reflected in the absence of a defect in oxidative phosphorylation in the epithelium. This suggests that there is selection against inherited mtDNA mutations in the gastrointestinal stem cells that is in marked contrast to the somatic mtDNA mutations that accumulate with age in epithelial stem cells leading to a biochemical defect. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


DNA, Mitochondrial/genetics , Epithelial Cells/chemistry , Gastric Mucosa/chemistry , Intestinal Mucosa/chemistry , Mitochondria/genetics , Mitochondrial Myopathies/genetics , Mutation , Stem Cells/chemistry , Adult , Case-Control Studies , Cellular Senescence/genetics , Epithelial Cells/pathology , Female , Gastric Mucosa/pathology , Genetic Predisposition to Disease , Heredity , Humans , Intestinal Mucosa/pathology , Middle Aged , Mitochondria/pathology , Mitochondrial Myopathies/pathology , Mitosis , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/pathology , Oxidative Phosphorylation , Pedigree , Phenotype , RNA, Transfer, Leu/genetics , RNA, Transfer, Lys/genetics , Selection, Genetic , Stem Cells/pathology
15.
EBioMedicine ; 31: 166-173, 2018 May.
Article En | MEDLINE | ID: mdl-29748033

Stem cell (SC) dynamics within the human colorectal crypt SC niche remain poorly understood, with previous studies proposing divergent hypotheses on the predominant mode of SC self-renewal and the rate of SC replacement. Here we use age-related mitochondrial oxidative phosphorylation (OXPHOS) defects to trace clonal lineages within human colorectal crypts across the adult life-course. By resolving the frequency and size distribution of OXPHOS-deficient clones, quantitative analysis shows that, in common with mouse, long-term maintenance of the colonic epithelial crypt relies on stochastic SC loss and replacement mediated by competition for limited niche access. We find that the colonic crypt is maintained by ~5 effective SCs. However, with a SC loss/replacement rate estimated to be slower than once per year, our results indicate that the vast majority of individual SC divisions result in asymmetric fate outcome. These findings provide a quantitative platform to detect and study deviations from human colorectal crypt SC niche homeostasis during the process of colorectal carcinogenesis.


Aging/metabolism , Colon/metabolism , Intestinal Mucosa/metabolism , Stem Cell Niche , Stem Cells/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aging/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Intestinal Mucosa/pathology , Middle Aged , Oxidative Phosphorylation , Stem Cells/pathology
16.
J Pathol ; 245(3): 311-323, 2018 07.
Article En | MEDLINE | ID: mdl-29660116

Defects in the respiratory chain, interfering with energy production in the cell, are major underlying causes of mitochondrial diseases. In spite of this, the surprising variety of clinical symptoms, disparity between ages of onset, as well as the involvement of mitochondrial impairment in ageing and age-related diseases continue to challenge our understanding of the pathogenic processes. This complexity can be in part attributed to the unique metabolic needs of organs or of various cell types. In this view, it remains essential to investigate mitochondrial dysfunction at the cellular level. For this purpose, we developed a novel enzyme histochemical method that enables precise quantification in fresh-frozen tissues using competing redox reactions which ultimately lead to the reduction of tetrazolium salts and formazan deposition in cytochrome c oxidase-deficient mitochondria. We demonstrate that the loss of oxidative activity is detected at very low levels - this achievement is unequalled by previous techniques and opens up new opportunities for the study of early disease processes or comparative investigations. Moreover, human biopsy samples of mitochondrial disease patients of diverse genotypic origins were used and the successful detection of COX-deficient cells suggests a broad application for this new method. Lastly, the assay can be adapted to a wide range of tissues in the mouse and extends to other animal models, which we show here with the fruit fly, Drosophila melanogaster. Overall, the new assay provides the means to quantify and map, on a cell-by-cell basis, the full extent of COX deficiency in tissues, thereby expending new possibilities for future investigation. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Cytochrome-c Oxidase Deficiency/diagnosis , Electron Transport Complex IV/metabolism , Single-Cell Analysis/methods , Staining and Labeling/methods , Animals , Cytochrome-c Oxidase Deficiency/enzymology , Cytochrome-c Oxidase Deficiency/genetics , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster , Energy Metabolism , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Methylphenazonium Methosulfate/chemistry , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Mutation , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Nitroblue Tetrazolium/chemistry , Oxidation-Reduction , Predictive Value of Tests , RNA, Transfer, Ala/genetics
17.
Genes (Basel) ; 9(4)2018 Mar 27.
Article En | MEDLINE | ID: mdl-29584704

Mitochondrial DNA (mtDNA) mutations accumulate in somatic stem cells during ageing and cause mitochondrial dysfunction. In this review, we summarize the studies that link mtDNA mutations to stem cell ageing. We discuss the age-related behaviours of the somatic mtDNA mutations in stem cell populations and how they potentially contribute to stem cell ageing by altering mitochondrial properties in humans and in mtDNA-mutator mice. We also draw attention to the diverse fates of the mtDNA mutations with different origins during ageing, with potential selective pressures on the germline inherited but not the somatic mtDNA mutations.

18.
J Gerontol A Biol Sci Med Sci ; 73(5): 571-578, 2018 04 17.
Article En | MEDLINE | ID: mdl-29045670

Mitochondrial dysfunction is prevalent in the aging gastrointestinal tract. We investigated whether mitochondrial function in aging colonic crypts and exercise influences microbial gut communities in mice. Twelve PolgAmut/mut mice were randomly divided into a sedentary and exercise group at 4 months. Seven-aged matched PolgA+/+ mice remained sedentary throughout. Stool samples were collected at 4, 7, and 11 months, and bacterial profiling was achieved through 16S rRNA sequencing profiling. Mitochondrial enzyme activity was assessed in colonic epithelial crypts at 11 months for PolgAmut/mut and PolgA+/+ mice. Sedentary and exercised PolgAmut/mut mice had significantly higher levels of mitochondrial dysfunction than PolgA+/+ mice (78%, 77%, and 1% of crypts, respectively). Bacterial profiles of sedentary PolgAmut/mut mice were significantly different from the sedentary PolgA+/+ mice, with increases in Lactobacillus and Mycoplasma, and decreases in Alistipes, Odoribacter, Anaeroplasma, Rikenella, Parabacteroides, and Allobaculum in the PolgAmut/mut mice. Exercise did not have any impact upon gut mitochondrial dysfunction; however, exercise did increase gut microbiota diversity and significantly increased bacterial genera Mucispirillum and Desulfovibrio. Mitochondrial dysfunction is associated with changes in the gut microbiota. Endurance exercise moderated some of these changes, establishing that environmental factors can influence gut microbiota, despite mitochondrial dysfunction.


Aging , Gastrointestinal Microbiome , Mitochondrial Diseases , Physical Conditioning, Animal , Animals , Mice , Aging/physiology , Feces/microbiology , Mitochondrial Diseases/physiopathology , Random Allocation , Sedentary Behavior
19.
Cell Rep ; 16(11): 2980-2990, 2016 09 13.
Article En | MEDLINE | ID: mdl-27626666

Mutations of mtDNA are an important cause of human disease, but few animal models exist. Because mammalian mitochondria cannot be transfected, the development of mice with pathogenic mtDNA mutations has been challenging, and the main strategy has therefore been to introduce mutations found in cell lines into mouse embryos. Here, we describe a phenotype-driven strategy that is based on detecting clonal expansion of pathogenic mtDNA mutations in colonic crypts of founder mice derived from heterozygous mtDNA mutator mice. As proof of concept, we report the generation of a mouse line transmitting a heteroplasmic pathogenic mutation in the alanine tRNA gene of mtDNA displaying typical characteristics of classic mitochondrial disease. In summary, we describe a straightforward and technically simple strategy based on mouse breeding and histology to generate animal models of mtDNA-mutation disease, which will be of great importance for studies of disease pathophysiology and preclinical treatment trials.


DNA, Mitochondrial/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , Animals , Breeding , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Clone Cells , Disease Models, Animal , Female , Mice, Inbred C57BL , Mitochondrial Diseases/physiopathology , Phenotype , Protein Biosynthesis , RNA, Transfer, Ala/genetics
20.
Sci Rep ; 6: 31907, 2016 08 24.
Article En | MEDLINE | ID: mdl-27553587

Fragility fractures caused by osteoporosis affect millions of people worldwide every year with significant levels of associated morbidity, mortality and costs to the healthcare economy. The pathogenesis of declining bone mineral density is poorly understood but it is inherently related to increasing age. Growing evidence in recent years, especially that provided by mouse models, suggest that accumulating somatic mitochondrial DNA mutations may cause the phenotypic changes associated with the ageing process including osteoporosis. Methods to study mitochondrial abnormalities in individual osteoblasts, osteoclasts and osteocytes are limited and impair our ability to assess the changes seen with age and in animal models of ageing. To enable the assessment of mitochondrial protein levels, we have developed a quadruple immunofluorescence method to accurately quantify the presence of mitochondrial respiratory chain components within individual bone cells. We have applied this technique to a well-established mouse model of ageing and osteoporosis and show respiratory chain deficiency.


Aging/genetics , DNA Polymerase gamma/genetics , Electron Transport Chain Complex Proteins/metabolism , Osteoblasts/metabolism , Osteoporosis/genetics , Aging/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Mice , Mice, Transgenic , Mitochondrial Proteins/metabolism , Mutation , Osteoblasts/pathology , Osteoporosis/metabolism
...