Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Science ; 384(6695): 563-572, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38696572

ABSTRACT

A molecular clock network is crucial for daily physiology and maintaining organismal health. We examined the interactions and importance of intratissue clock networks in muscle tissue maintenance. In arrhythmic mice showing premature aging, we created a basic clock module involving a central and a peripheral (muscle) clock. Reconstituting the brain-muscle clock network is sufficient to preserve fundamental daily homeostatic functions and prevent premature muscle aging. However, achieving whole muscle physiology requires contributions from other peripheral clocks. Mechanistically, the muscle peripheral clock acts as a gatekeeper, selectively suppressing detrimental signals from the central clock while integrating important muscle homeostatic functions. Our research reveals the interplay between the central and peripheral clocks in daily muscle function and underscores the impact of eating patterns on these interactions.


Subject(s)
Aging, Premature , Aging , Brain , Circadian Rhythm , Muscle, Skeletal , Animals , Male , Mice , Aging/genetics , Aging/physiology , Aging, Premature/genetics , Aging, Premature/prevention & control , Brain/physiology , Circadian Clocks/physiology , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Homeostasis , Muscle, Skeletal/physiology , Mice, Knockout , ARNTL Transcription Factors/genetics
2.
Nature ; 629(8010): 154-164, 2024 May.
Article in English | MEDLINE | ID: mdl-38649488

ABSTRACT

Muscle atrophy and functional decline (sarcopenia) are common manifestations of frailty and are critical contributors to morbidity and mortality in older people1. Deciphering the molecular mechanisms underlying sarcopenia has major implications for understanding human ageing2. Yet, progress has been slow, partly due to the difficulties of characterizing skeletal muscle niche heterogeneity (whereby myofibres are the most abundant) and obtaining well-characterized human samples3,4. Here we generate a single-cell/single-nucleus transcriptomic and chromatin accessibility map of human limb skeletal muscles encompassing over 387,000 cells/nuclei from individuals aged 15 to 99 years with distinct fitness and frailty levels. We describe how cell populations change during ageing, including the emergence of new populations in older people, and the cell-specific and multicellular network features (at the transcriptomic and epigenetic levels) associated with these changes. On the basis of cross-comparison with genetic data, we also identify key elements of chromatin architecture that mark susceptibility to sarcopenia. Our study provides a basis for identifying targets in the skeletal muscle that are amenable to medical, pharmacological and lifestyle interventions in late life.


Subject(s)
Aging , Muscle, Skeletal , Single-Cell Analysis , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Young Adult , Aging/genetics , Aging/pathology , Aging/physiology , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin/genetics , Disease Susceptibility , Epigenesis, Genetic , Frailty/genetics , Frailty/pathology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Sarcopenia/genetics , Sarcopenia/pathology , Transcriptome
4.
FEBS J ; 290(5): 1161-1185, 2023 03.
Article in English | MEDLINE | ID: mdl-35811491

ABSTRACT

Cellular senescence is a state of irreversible cell cycle arrest that often emerges after tissue damage and in age-related diseases. Through the production of a multicomponent secretory phenotype (SASP), senescent cells can impact the regeneration and function of tissues. However, the effects of senescent cells and their SASP are very heterogeneous and depend on the tissue environment and type as well as the duration of injury, the degree of persistence of senescent cells and the organism's age. While the transient presence of senescent cells is widely believed to be beneficial, recent data suggest that it is detrimental for tissue regeneration after acute damage. Furthermore, although senescent cell persistence is typically associated with the progression of age-related chronic degenerative diseases, it now appears to be also necessary for correct tissue function in the elderly. Here, we discuss what is currently known about the roles of senescent cells and their SASP in tissue regeneration in ageing and age-related diseases, highlighting their (negative and/or positive) contributions. We provide insight for future research, including the possibility of senolytic-based therapies and cellular reprogramming, with aims ranging from enhancing tissue repair to extending a healthy lifespan.


Subject(s)
Cellular Senescence , Longevity , Cellular Senescence/genetics , Phenotype , Biological Transport
5.
Nature ; 613(7942): 169-178, 2023 01.
Article in English | MEDLINE | ID: mdl-36544018

ABSTRACT

Tissue regeneration requires coordination between resident stem cells and local niche cells1,2. Here we identify that senescent cells are integral components of the skeletal muscle regenerative niche that repress regeneration at all stages of life. The technical limitation of senescent-cell scarcity3 was overcome by combining single-cell transcriptomics and a senescent-cell enrichment sorting protocol. We identified and isolated different senescent cell types from damaged muscles of young and old mice. Deeper transcriptome, chromatin and pathway analyses revealed conservation of cell identity traits as well as two universal senescence hallmarks (inflammation and fibrosis) across cell type, regeneration time and ageing. Senescent cells create an aged-like inflamed niche that mirrors inflammation associated with ageing (inflammageing4) and arrests stem cell proliferation and regeneration. Reducing the burden of senescent cells, or reducing their inflammatory secretome through CD36 neutralization, accelerates regeneration in young and old mice. By contrast, transplantation of senescent cells delays regeneration. Our results provide a technique for isolating in vivo senescent cells, define a senescence blueprint for muscle, and uncover unproductive functional interactions between senescent cells and stem cells in regenerative niches that can be overcome. As senescent cells also accumulate in human muscles, our findings open potential paths for improving muscle repair throughout life.


Subject(s)
Aging , Cellular Senescence , Inflammation , Muscle, Skeletal , Regeneration , Stem Cell Niche , Aged , Animals , Humans , Mice , Aging/metabolism , Aging/physiology , Cellular Senescence/physiology , Inflammation/metabolism , Inflammation/physiopathology , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Stem Cells/physiology , Fibrosis/physiopathology , Stem Cell Niche/physiology , Transcriptome , Chromatin/genetics , Geroscience
6.
Nat Aging ; 2: 851-866, 2022 09.
Article in English | MEDLINE | ID: mdl-36438588

ABSTRACT

Cellular senescence is a stable type of cell cycle arrest triggered by different stresses. As such, senescence drives age-related diseases and curbs cellular replicative potential. Here, we show that 3-deazaadenosine (3DA), an S-adenosyl homocysteinase (AHCY) inhibitor, alleviates replicative and oncogene-induced senescence. 3DA-treated senescent cells showed reduced global Histone H3 Lysine 36 trimethylation (H3K36me3), an epigenetic modification that marks the bodies of actively transcribed genes. By integrating transcriptome and epigenome data, we demonstrate that 3DA treatment affects key factors of the senescence transcriptional program. Remarkably, 3DA treatment alleviated senescence and increased the proliferative and regenerative potential of muscle stem cells from very old mice in vitro and in vivo. Moreover, ex vivo 3DA treatment was sufficient to enhance the engraftment of human umbilical cord blood (UCB) cells in immunocompromised mice. Together, our results identify 3DA as a promising drug enhancing the efficiency of cellular therapies by restraining senescence.


Subject(s)
Cellular Senescence , Histones , Humans , Mice , Animals , Histones/genetics , Cellular Senescence/genetics , Tubercidin/pharmacology , Epigenesis, Genetic
8.
Cell Stem Cell ; 29(9): 1298-1314.e10, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35998641

ABSTRACT

Skeletal muscle regeneration depends on the correct expansion of resident quiescent stem cells (satellite cells), a process that becomes less efficient with aging. Here, we show that mitochondrial dynamics are essential for the successful regenerative capacity of satellite cells. The loss of mitochondrial fission in satellite cells-due to aging or genetic impairment-deregulates the mitochondrial electron transport chain (ETC), leading to inefficient oxidative phosphorylation (OXPHOS) metabolism and mitophagy and increased oxidative stress. This state results in muscle regenerative failure, which is caused by the reduced proliferation and functional loss of satellite cells. Regenerative functions can be restored in fission-impaired or aged satellite cells by the re-establishment of mitochondrial dynamics (by activating fission or preventing fusion), OXPHOS, or mitophagy. Thus, mitochondrial shape and physical networking controls stem cell regenerative functions by regulating metabolism and proteostasis. As mitochondrial fission occurs less frequently in the satellite cells in older humans, our findings have implications for regeneration therapies in sarcopenia.


Subject(s)
Mitochondrial Dynamics , Mitophagy , Aged , Humans , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscles/metabolism , Stem Cells/metabolism
9.
Nat Commun ; 12(1): 5043, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413292

ABSTRACT

Skeletal muscle has a remarkable ability to regenerate owing to its resident stem cells (also called satellite cells, SCs). SCs are normally quiescent; when stimulated by damage, they activate and expand to form new fibers. The mechanisms underlying SC proliferative progression remain poorly understood. Here we show that DHX36, a helicase that unwinds RNA G-quadruplex (rG4) structures, is essential for muscle regeneration by regulating SC expansion. DHX36 (initially named RHAU) is barely expressed at quiescence but is highly induced during SC activation and proliferation. Inducible deletion of Dhx36 in adult SCs causes defective proliferation and muscle regeneration after damage. System-wide mapping in proliferating SCs reveals DHX36 binding predominantly to rG4 structures at various regions of mRNAs, while integrated polysome profiling shows that DHX36 promotes mRNA translation via 5'-untranslated region (UTR) rG4 binding. Furthermore, we demonstrate that DHX36 specifically regulates the translation of Gnai2 mRNA by unwinding its 5' UTR rG4 structures and identify GNAI2 as a downstream effector of DHX36 for SC expansion. Altogether, our findings uncover DHX36 as an indispensable post-transcriptional regulator of SC function and muscle regeneration acting through binding and unwinding rG4 structures at 5' UTR of target mRNAs.


Subject(s)
5' Untranslated Regions , DEAD-box RNA Helicases/metabolism , G-Quadruplexes , Muscles/cytology , Regeneration/physiology , Stem Cells/cytology , Animals , Animals, Genetically Modified , Cells, Cultured , Disease Models, Animal , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation , Humans , Mice , Muscles/metabolism , Myoblasts/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Stem Cells/metabolism
10.
Stem Cell Reports ; 16(9): 2089-2098, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34450038

ABSTRACT

Regeneration of skeletal muscle requires resident stem cells called satellite cells. Here, we report that the chromatin remodeler CHD4, a member of the nucleosome remodeling and deacetylase (NuRD) repressive complex, is essential for the expansion and regenerative functions of satellite cells. We show that conditional deletion of the Chd4 gene in satellite cells results in failure to regenerate muscle after injury. This defect is principally associated with increased stem cell plasticity and lineage infidelity during the expansion of satellite cells, caused by de-repression of non-muscle-cell lineage genes in the absence of Chd4. Thus, CHD4 ensures that a transcriptional program that safeguards satellite cell identity during muscle regeneration is maintained. Given the therapeutic potential of muscle stem cells in diverse neuromuscular pathologies, CHD4 constitutes an attractive target for satellite cell-based therapies.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , DNA Helicases/genetics , Muscle, Skeletal/physiology , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Animals , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Models, Biological , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
11.
Nat Cell Biol ; 22(11): 1307-1318, 2020 11.
Article in English | MEDLINE | ID: mdl-33106654

ABSTRACT

Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.


Subject(s)
Antigens, CD34/metabolism , Cell Proliferation , Cell Self Renewal , Cellular Senescence , Forkhead Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Age Factors , Animals , Cardiotoxins/toxicity , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/transplantation , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/drug effects , Regeneration/genetics , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/transplantation , Signal Transduction , Stem Cell Niche
12.
Cell Rep ; 31(7): 107652, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433961

ABSTRACT

Somatic stem cells expand massively during tissue regeneration, which might require control of cell fitness, allowing elimination of non-competitive, potentially harmful cells. How or if such cells are removed to restore organ function is not fully understood. Here, we show that a substantial fraction of muscle stem cells (MuSCs) undergo necroptosis because of epigenetic rewiring during chronic skeletal muscle regeneration, which is required for efficient regeneration of dystrophic muscles. Inhibition of necroptosis strongly enhances suppression of MuSC expansion in a non-cell-autonomous manner. Prevention of necroptosis in MuSCs of healthy muscles is mediated by the chromatin remodeler CHD4, which directly represses the necroptotic effector Ripk3, while CHD4-dependent Ripk3 repression is dramatically attenuated in dystrophic muscles. Loss of Ripk3 repression by inactivation of Chd4 causes massive necroptosis of MuSCs, abolishing regeneration. Our study demonstrates how programmed cell death in MuSCs is tightly controlled to achieve optimal tissue regeneration.


Subject(s)
Epigenesis, Genetic/genetics , Muscle, Skeletal/metabolism , Necroptosis/genetics , Humans
13.
EMBO Rep ; 21(4): e49075, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32107853

ABSTRACT

Macrophages are characterized by a high plasticity in response to changes in tissue microenvironment, which allows them to acquire different phenotypes and to exert essential functions in complex processes, such as tissue regeneration. Here, we report that the membrane protein Cripto plays a key role in shaping macrophage plasticity in skeletal muscle during regeneration and disease. Conditional deletion of Cripto in the myeloid lineage (CriptoMy-LOF ) perturbs MP plasticity in acutely injured muscle and in mouse models of Duchenne muscular dystrophy (mdx). Specifically, CriptoMy-LOF macrophages infiltrate the muscle, but fail to properly expand as anti-inflammatory CD206+ macrophages, which is due, at least in part, to aberrant activation of TGFß/Smad signaling. This reduction in macrophage plasticity disturbs vascular remodeling by increasing Endothelial-to-Mesenchymal Transition (EndMT), reduces muscle regenerative potential, and leads to an exacerbation of the dystrophic phenotype. Thus, in muscle-infiltrating macrophages, Cripto is required to promote the expansion of the CD206+ anti-inflammatory macrophage type and to restrict the EndMT process, providing a direct functional link between this macrophage population and endothelial cells.


Subject(s)
Endothelial Cells , Muscular Dystrophy, Duchenne , Animals , Macrophages , Mice , Mice, Inbred mdx , Muscle, Skeletal
14.
Nat Commun ; 11(1): 189, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31929511

ABSTRACT

A unique property of skeletal muscle is its ability to adapt its mass to changes in activity. Inactivity, as in disuse or aging, causes atrophy, the loss of muscle mass and strength, leading to physical incapacity and poor quality of life. Here, through a combination of transcriptomics and transgenesis, we identify sestrins, a family of stress-inducible metabolic regulators, as protective factors against muscle wasting. Sestrin expression decreases during inactivity and its genetic deficiency exacerbates muscle wasting; conversely, sestrin overexpression suffices to prevent atrophy. This protection occurs through mTORC1 inhibition, which upregulates autophagy, and AKT activation, which in turn inhibits FoxO-regulated ubiquitin-proteasome-mediated proteolysis. This study reveals sestrin as a central integrator of anabolic and degradative pathways preventing muscle wasting. Since sestrin also protected muscles against aging-induced atrophy, our findings have implications for sarcopenia.


Subject(s)
Heat-Shock Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/prevention & control , Nuclear Proteins/metabolism , Signal Transduction , Aging , Animals , Autophagy , Disease Models, Animal , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression , Heat-Shock Proteins/genetics , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Nuclear Proteins/genetics , Sarcopenia/genetics , Sarcopenia/metabolism , Sarcopenia/pathology , Sarcopenia/prevention & control
15.
Methods Mol Biol ; 2045: 13-23, 2019.
Article in English | MEDLINE | ID: mdl-30771188

ABSTRACT

The maintenance of adult stem cells in their normal quiescent state depends on intrinsic factors and extrinsic signals originating from their microenvironment (also known as the stem cell niche). In skeletal muscle, its stem cells (satellite cells) lose their regenerative potential with aging, and this has been attributed, at least in part, to both age-associated changes in the satellite cells as in the niche cells, which include resident fibro-adipogenic progenitors (FAPs), macrophages, and endothelial cells, among others. To understand the regenerative decline of skeletal muscle with aging, there is a need for methods to specifically isolate stem and niche cells from resting muscle. Here we describe a fluorescence-activated cell sorting (FACS) protocol to simultaneously isolate discrete populations of satellite cells and niche cells from skeletal muscle of aging mice.


Subject(s)
Adult Stem Cells/metabolism , Flow Cytometry/methods , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Stem Cell Niche/genetics , Adult Stem Cells/cytology , Aging , Animals , Antibodies , Endothelial Cells/cytology , Endothelial Cells/metabolism , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Mice , Muscle, Skeletal/cytology , Real-Time Polymerase Chain Reaction , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Niche/physiology , Workflow
16.
Curr Opin Pharmacol ; 40: 147-155, 2018 06.
Article in English | MEDLINE | ID: mdl-29883814

ABSTRACT

Aging is the prime risk factor for the broad-based development of diseases. Frailty is a phenotypical hallmark of aging and is often used to assess whether the predicted benefits of a therapy outweigh the risks for older patients. Senescent cells form as a consequence of unresolved molecular damage and persistently secrete molecules that can impair tissue function. Recent evidence shows senescent cells can chronically interfere with stem cell function and drive aging of the musculoskeletal system. In addition, targeted apoptosis of senescent cells can restore tissue homeostasis in aged animals. Thus, targeting cellular senescence provides new therapeutic opportunities for the intervention of frailty-associated pathologies and could have pleiotropic health benefits.


Subject(s)
Cellular Senescence/drug effects , Molecular Targeted Therapy/methods , Musculoskeletal Diseases/drug therapy , Musculoskeletal System/drug effects , Regeneration/drug effects , Age Factors , Animals , Apoptosis/drug effects , Bone Remodeling/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Bone and Bones/physiopathology , Cartilage/drug effects , Cartilage/metabolism , Cartilage/pathology , Cartilage/physiopathology , Chondrogenesis/drug effects , Drug Design , Humans , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Musculoskeletal Diseases/metabolism , Musculoskeletal Diseases/pathology , Musculoskeletal Diseases/physiopathology , Musculoskeletal System/metabolism , Musculoskeletal System/pathology , Musculoskeletal System/physiopathology , Signal Transduction/drug effects
17.
Cell ; 170(4): 678-692.e20, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28802040

ABSTRACT

Normal homeostatic functions of adult stem cells have rhythmic daily oscillations that are believed to become arrhythmic during aging. Unexpectedly, we find that aged mice remain behaviorally circadian and that their epidermal and muscle stem cells retain a robustly rhythmic core circadian machinery. However, the oscillating transcriptome is extensively reprogrammed in aged stem cells, switching from genes involved in homeostasis to those involved in tissue-specific stresses, such as DNA damage or inefficient autophagy. Importantly, deletion of circadian clock components did not reproduce the hallmarks of this reprogramming, underscoring that rewiring, rather than arrhythmia, is associated with physiological aging. While age-associated rewiring of the oscillatory diurnal transcriptome is not recapitulated by a high-fat diet in young adult mice, it is significantly prevented by long-term caloric restriction in aged mice. Thus, stem cells rewire their diurnal timed functions to adapt to metabolic cues and to tissue-specific age-related traits.


Subject(s)
Adult Stem Cells/pathology , Cellular Senescence , Circadian Rhythm , Epidermis/pathology , Muscle, Skeletal/pathology , Adult Stem Cells/physiology , Animals , Autophagy , Caloric Restriction , Circadian Clocks , DNA Damage , Diet, High-Fat , Homeostasis , Mice , Stress, Physiological , Transcriptome
18.
Dev Cell ; 42(2): 114-116, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28743000

ABSTRACT

Fibro/adipogenic progenitors (FAPs) are emerging as crucial regulators of fibrous and fat deposits during skeletal muscle regeneration. In a recent issue of Cell, Kopinke et al. (2017) report that primary cilia induce the adipogenic fate of FAPs in injured and diseased muscle by restraining Hedgehog signaling.


Subject(s)
Adipocytes , Cilia , Adipogenesis , Cell Differentiation , Humans , Muscle, Skeletal
19.
Stem Cells ; 35(7): 1687-1703, 2017 07.
Article in English | MEDLINE | ID: mdl-28472853

ABSTRACT

Coenzyme Q10 (CoQ10 ) plays a crucial role in mitochondria as an electron carrier within the mitochondrial respiratory chain (MRC) and is an essential antioxidant. Mutations in genes responsible for CoQ10 biosynthesis (COQ genes) cause primary CoQ10 deficiency, a rare and heterogeneous mitochondrial disorder with no clear genotype-phenotype association, mainly affecting tissues with high-energy demand including brain and skeletal muscle (SkM). Here, we report a four-year-old girl diagnosed with minor mental retardation and lethal rhabdomyolysis harboring a heterozygous mutation (c.483G > C (E161D)) in COQ4. The patient's fibroblasts showed a decrease in [CoQ10 ], CoQ10 biosynthesis, MRC activity affecting complexes I/II + III, and respiration defects. Bona fide induced pluripotent stem cell (iPSCs) lines carrying the COQ4 mutation (CQ4-iPSCs) were generated, characterized and genetically edited using the CRISPR-Cas9 system (CQ4ed -iPSCs). Extensive differentiation and metabolic assays of control-iPSCs, CQ4-iPSCs and CQ4ed -iPSCs demonstrated a genotype association, reproducing the disease phenotype. The COQ4 mutation in iPSC was associated with CoQ10 deficiency, metabolic dysfunction, and respiration defects. iPSC differentiation into SkM was compromised, and the resulting SkM also displayed respiration defects. Remarkably, iPSC differentiation in dopaminergic or motor neurons was unaffected. This study offers an unprecedented iPSC model recapitulating CoQ10 deficiency-associated functional and metabolic phenotypes caused by COQ4 mutation. Stem Cells 2017;35:1687-1703.


Subject(s)
Ataxia/genetics , Intellectual Disability/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Rhabdomyolysis/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Ataxia/enzymology , Ataxia/pathology , CRISPR-Cas Systems , Cell Differentiation , Child, Preschool , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Fatal Outcome , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Editing/methods , Gene Expression , Genes, Lethal , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Intellectual Disability/enzymology , Intellectual Disability/pathology , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , Mitochondrial Proteins/deficiency , Motor Neurons/cytology , Motor Neurons/metabolism , Muscle Weakness/enzymology , Muscle Weakness/pathology , Primary Cell Culture , Rhabdomyolysis/enzymology , Rhabdomyolysis/pathology , Ubiquinone/genetics
20.
Methods Mol Biol ; 1556: 3-19, 2017.
Article in English | MEDLINE | ID: mdl-28247342

ABSTRACT

Skeletal muscle stem cells, originally termed satellite cells for their position adjacent to differentiated muscle fibers, are absolutely required for the process of skeletal muscle repair and regeneration. In the last decade, satellite cells have become one of the most studied adult stem cell systems and have emerged as a standard model not only in the field of stem cell-driven tissue regeneration but also in stem cell dysfunction and aging. Here, we provide background in the field and discuss recent advances in our understanding of muscle stem cell function and dysfunction, particularly in the case of aging, and the potential involvement of muscle stem cells in genetic diseases such as the muscular dystrophies.


Subject(s)
Muscle, Skeletal/cytology , Stem Cells/cytology , Stem Cells/physiology , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Biomarkers , Cell Differentiation , Cellular Senescence/genetics , Humans , Muscular Dystrophies/etiology , Muscular Dystrophies/metabolism , Phenotype , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...