Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.705
1.
J Physiol ; 602(9): 1967-1986, 2024 May.
Article En | MEDLINE | ID: mdl-38564214

Mitochondria within skeletal muscle cells are located either between the muscle contractile apparatus (interfibrillar mitochondria, IFM) or beneath the cell membrane (subsarcolemmal mitochondria, SSM), with several structural and functional differences reported between IFM and SSM. However, recent 3D imaging studies demonstrate that mitochondria are particularly concentrated in the proximity of capillaries embedded in sarcolemmal grooves rather than in proximity to the sarcolemma itself (paravascular mitochondria, PVM). To evaluate the impact of capillary vs. sarcolemmal proximity, we compared the structure and function of skeletal muscle mitochondria located either lateral to embedded capillaries (PVM), adjacent to the sarcolemma but not in PVM pools (SSM) or interspersed between sarcomeres (IFM). Mitochondrial morphology and interactions were assessed by 3D electron microscopy coupled with machine learning segmentation, whereas mitochondrial energy conversion was assessed by two-photon microscopy of mitochondrial membrane potential, content, calcium, NADH redox and flux in live, intact cells. Structurally, although PVM and SSM were similarly larger than IFM, PVM were larger, rounder and had more physical connections to neighbouring mitochondria compared to both IFM and SSM. Functionally, PVM had similar or greater basal NADH flux compared to SSM and IFM, respectively, despite a more oxidized NADH pool and a greater membrane potential, signifying a greater activation of the electron transport chain in PVM. Together, these data indicate that proximity to capillaries has a greater impact on resting mitochondrial energy conversion and distribution in skeletal muscle than the sarcolemma alone. KEY POINTS: Capillaries have a greater impact on mitochondrial energy conversion in skeletal muscle than the sarcolemma. Paravascular mitochondria are larger, and the outer mitochondrial membrane is more connected with neighbouring mitochondria. Interfibrillar mitochondria are longer and have greater contact sites with other organelles (i.e. sarcoplasmic reticulum and lipid droplets). Paravascular mitochondria have greater activation of oxidative phosphorylation than interfibrillar mitochondria at rest, although this is not regulated by calcium.


Capillaries , Mitochondria, Muscle , Muscle, Skeletal , Sarcolemma , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Sarcolemma/physiology , Animals , Capillaries/physiology , Capillaries/metabolism , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/blood supply , Mice , Energy Metabolism/physiology , Male , Mice, Inbred C57BL , Membrane Potential, Mitochondrial/physiology
2.
J Nutr Biochem ; 100: 108902, 2022 02.
Article En | MEDLINE | ID: mdl-34748920

A diet high in saturated fat leads to skeletal muscle deteriorations including insulin resistance, mitochondrial dysfunction and muscle fiber atrophy. Consumption of long-chain polyunsaturated fatty acids and exercise have shown promise in ameliorating high-fat diet (HFD)-induced oxidative stress and inflammation. However, the impact of extra virgin olive oil (EVOO) on mitochondrial homeostasis in muscle is largely unknown. This study aimed to investigate whether 12 wks of EVOO feeding alone and in conjunction with endurance training could protect against metabolic and mitochondrial dysfunction rat muscle with HFD. Female Sprague-Dawley rats were divided into 4 groups fed a control diet (C), HFD, EVOO diet, and EVOO diet with training (EVOO+T). Mitochondrial enzyme activity and protein content decreased with HFD compared to C, but were restored with EVOO and EVOO+T. EVOO+T elevated muscle cytochrome c and PGC-1α levels. HFD increased muscle proteolytic markers and protein ubiquitination, whereas these effects were not seen in EVOO and EVOO+T. HFD suppressed mitochondrial fusion protein level while increasing fission protein levels, but were restored with EVOO and EVOO+T. Mitophagy marker PINK1 content decreased with HFD, but was unchanged in EVOO and EVOO+T. EVOO+T upregulated autophagy markers, along with decreased phosphorylated/dephosphorylated FoxO3 ratio. Antioxidants enzyme levels were upregulated by EVOO and EVOO+T, and EVOO+T reduced HFD-induced lipid peroxidation. In conclusion, HFD impaired muscle oxidative capacity, promoted protein ubiquitination and mitochondrial fission, and upregulated autophagy markers. Replacement of HFD with EVOO corrected the observed adverse effects, while exercise training in conjunction with EVOO provided additional protection to the muscle.


Diet, High-Fat/adverse effects , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Olive Oil , Physical Conditioning, Animal , Animals , Antioxidants/metabolism , Autophagy , Body Weight , Cholesterol/blood , Female , Insulin/blood , Mitochondria, Muscle/ultrastructure , Mitochondrial Dynamics , Muscle, Skeletal/ultrastructure , Oxidation-Reduction , Proteolysis , Rats , Rats, Sprague-Dawley , Ubiquitination
3.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article En | MEDLINE | ID: mdl-34502433

Diabetes mellitus is a systemic metabolic disorder associated with mitochondrial dysfunction, with mitochondrial permeability transition (MPT) pore opening being recognized as one of its pathogenic mechanisms. Alisporivir has been recently identified as a non-immunosuppressive analogue of the MPT pore blocker cyclosporin A and has broad therapeutic potential. The purpose of the present work was to study the effect of alisporivir (2.5 mg/kg/day i.p.) on the ultrastructure and functions of the skeletal muscle mitochondria of mice with diabetes mellitus induced by a high-fat diet combined with streptozotocin injections. The glucose tolerance tests indicated that alisporivir increased the rate of glucose utilization in diabetic mice. An electron microscopy analysis showed that alisporivir prevented diabetes-induced changes in the ultrastructure and content of the mitochondria in myocytes. In diabetes, the ADP-stimulated respiration, respiratory control, and ADP/O ratios and the level of ATP synthase in the mitochondria decreased, whereas alisporivir treatment restored these indicators. Alisporivir eliminated diabetes-induced increases in mitochondrial lipid peroxidation products. Diabetic mice showed decreased mRNA levels of Atp5f1a, Ant1, and Ppif and increased levels of Ant2 in the skeletal muscles. The skeletal muscle mitochondria of diabetic animals were sensitized to the MPT pore opening. Alisporivir normalized the expression level of Ant2 and mitochondrial susceptibility to the MPT pore opening. In parallel, the levels of Mfn2 and Drp1 also returned to control values, suggesting a normalization of mitochondrial dynamics. These findings suggest that the targeting of the MPT pore opening by alisporivir is a therapeutic approach to prevent the development of mitochondrial dysfunction and associated oxidative stress in the skeletal muscles in diabetes.


Cyclosporine/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Mitochondria, Muscle/drug effects , Animals , Cyclosporine/pharmacology , Diet, High-Fat , Drug Evaluation, Preclinical , Male , Mice, Inbred C57BL , Mitochondria, Muscle/ultrastructure , Mitochondrial Dynamics/drug effects , Mitochondrial Permeability Transition Pore
4.
Physiol Rep ; 9(16): e15016, 2021 08.
Article En | MEDLINE | ID: mdl-34427401

This study aimed to examine the effects of voluntary wheel running on cancer cachexia-induced mitochondrial alterations in mouse skeletal muscle. Mice bearing colon 26 adenocarcinoma (C26) were used as a model of cancer cachexia. C26 mice showed a lower gastrocnemius and plantaris muscle weight, but 4 weeks of voluntary exercise rescued these changes. Further, voluntary exercise attenuated observed declines in the levels of oxidative phosphorylation proteins and activities of citrate synthase and cytochrome c oxidase in the skeletal muscle of C26 mice. Among mitochondrial morphology regulatory proteins, mitofusin 2 (Mfn2) and dynamin-related protein 1 (Drp1) were decreased in the skeletal muscle of C26 mice, but exercise resulted in similar improvements as seen in markers of mitochondrial content. In isolated mitochondria, 4-hydroxynonenal and protein carbonyls were elevated in C26 mice, but exercise blunted the increases in these markers of oxidative stress. In addition, electron microscopy revealed that exercise alleviated the observed increase in the percentage of damaged mitochondria in C26 mice. These results suggest that voluntary exercise effectively counteracts mitochondrial dysfunction to mitigate muscle loss in cachexia.


Cachexia/prevention & control , Mitochondria, Muscle/ultrastructure , Neoplasms/complications , Physical Conditioning, Animal/methods , Animals , Cachexia/etiology , Citrate (si)-Synthase/metabolism , Dynamins/metabolism , Electron Transport Complex IV/metabolism , GTP Phosphohydrolases/metabolism , Male , Mice , Mitochondria, Muscle/metabolism , Motor Activity , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Oxidative Stress , Protein Carbonylation
5.
Elife ; 102021 08 27.
Article En | MEDLINE | ID: mdl-34448452

Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.


Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Microtubules/metabolism , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Neuromuscular Junction/metabolism , Organelle Biogenesis , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Excitation Contraction Coupling , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microtubules/genetics , Microtubules/ultrastructure , Mitochondria, Muscle/genetics , Mitochondria, Muscle/ultrastructure , Muscle Fatigue , Muscle Fibers, Skeletal/ultrastructure , Muscle Strength , Myoblasts, Skeletal/ultrastructure , Neuromuscular Junction/genetics , Neuromuscular Junction/ultrastructure , Time Factors
6.
PLoS Genet ; 17(3): e1009488, 2021 03.
Article En | MEDLINE | ID: mdl-33780446

Mitochondria are essential for maintaining skeletal muscle metabolic homeostasis during adaptive response to a myriad of physiologic or pathophysiological stresses. The mechanisms by which mitochondrial function and contractile fiber type are concordantly regulated to ensure muscle function remain poorly understood. Evidence is emerging that the Folliculin interacting protein 1 (Fnip1) is involved in skeletal muscle fiber type specification, function, and disease. In this study, Fnip1 was specifically expressed in skeletal muscle in Fnip1-transgenic (Fnip1Tg) mice. Fnip1Tg mice were crossed with Fnip1-knockout (Fnip1KO) mice to generate Fnip1TgKO mice expressing Fnip1 only in skeletal muscle but not in other tissues. Our results indicate that, in addition to the known role in type I fiber program, FNIP1 exerts control upon muscle mitochondrial oxidative program through AMPK signaling. Indeed, basal levels of FNIP1 are sufficient to inhibit AMPK but not mTORC1 activity in skeletal muscle cells. Gain-of-function and loss-of-function strategies in mice, together with assessment of primary muscle cells, demonstrated that skeletal muscle mitochondrial program is suppressed via the inhibitory actions of FNIP1 on AMPK. Surprisingly, the FNIP1 actions on type I fiber program is independent of AMPK and its downstream PGC-1α. These studies provide a vital framework for understanding the intrinsic role of FNIP1 as a crucial factor in the concerted regulation of mitochondrial function and muscle fiber type that determine muscle fitness.


AMP-Activated Protein Kinases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Female , Gene Expression Profiling , Male , Mice , Mice, Transgenic , Mitochondria, Muscle/ultrastructure , Muscle Fibers, Skeletal/ultrastructure , Organ Specificity , Oxidation-Reduction , Oxidative Stress
8.
Science ; 371(6528)2021 01 29.
Article En | MEDLINE | ID: mdl-33303683

Treatments are lacking for sarcopenia, a debilitating age-related skeletal muscle wasting syndrome. We identifed increased amounts of 15-hydroxyprostaglandin dehydrogenase (15-PGDH), the prostaglandin E2 (PGE2)-degrading enzyme, as a hallmark of aged tissues, including skeletal muscle. The consequent reduction in PGE2 signaling contributed to muscle atrophy in aged mice and results from 15-PGDH-expressing myofibers and interstitial cells, such as macrophages, within muscle. Overexpression of 15-PGDH in young muscles induced atrophy. Inhibition of 15-PGDH, by targeted genetic depletion or a small-molecule inhibitor, increased aged muscle mass, strength, and exercise performance. These benefits arise from a physiological increase in PGE2 concentrations, which augmented mitochondrial function and autophagy and decreased transforming growth factor-ß signaling and activity of ubiquitin-proteasome pathways. Thus, PGE2 signaling ameliorates muscle atrophy and rejuvenates muscle function, and 15-PGDH may be a suitable therapeutic target for countering sarcopenia.


Aging/metabolism , Dinoprostone/metabolism , Hydroxyprostaglandin Dehydrogenases/physiology , Muscle, Skeletal/pathology , Rejuvenation , Sarcopenia/enzymology , Animals , Autophagic Cell Death/genetics , Autophagic Cell Death/physiology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/genetics , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/ultrastructure , Muscle Strength/genetics , Muscle Strength/physiology , Muscle, Skeletal/enzymology , Myofibrils/enzymology , Sarcopenia/genetics
9.
Toxicol Appl Pharmacol ; 411: 115366, 2021 01 15.
Article En | MEDLINE | ID: mdl-33316273

Mitochondrial dysfunction is a well-characterized consequence of spinal cord injury (SCI). We previously reported that treatment with the FDA-approved ß2-adrenergic receptor agonist formoterol beginning 8 h post-SCI induces mitochondrial biogenesis (MB) and improves body composition and locomotor recovery in female mice. To determine the time-to-treatment window of formoterol, female mice were subjected to 80 kdyn contusion SCI and daily administration of vehicle or formoterol (0.3 mg/kg) beginning 24 h after injury. This delayed treatment paradigm improved body composition in female mice by 21 DPI, returning body weight to pre-surgery weight and restoring gastrocnemius mass to sham levels; however, there was no effect on locomotor recovery, as measured by the Basso-Mouse Scale (BMS), or lesion volume. To assess the cross-sex potential of formoterol, injured male mice were treated with vehicle or formoterol (0.3 or 1.0 mg/kg) beginning 8 h after SCI. Formoterol also improved body composition post-SCI in male mice, restoring body weight and muscle mass regardless of dose. Interestingly, however, improved BMS scores and decreased lesion volume was observed only in male mice treated with 0.3 mg/kg. Additionally, 0.3 mg/kg formoterol induced MB in the gastrocnemius and injured spinal cord, as evidenced by increased MB protein expression and mitochondrial number. These data indicate that formoterol treatment improves recovery post-SCI in both male and female mice in a dose- and initiation time-dependent manner. Furthermore, formoterol-induced functional recovery post-SCI is not directly associated with peripheral effects, such as muscle mass and body weight.


Adrenergic beta-2 Receptor Agonists/administration & dosage , Formoterol Fumarate/administration & dosage , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Organelle Biogenesis , Receptors, Adrenergic, beta-2/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Time-to-Treatment , Animals , Body Composition/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Male , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Receptors, Adrenergic, beta-2/metabolism , Recovery of Function , Sex Factors , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors
10.
Mitochondrion ; 56: 82-90, 2021 01.
Article En | MEDLINE | ID: mdl-33220503

The efficient production of energy via oxidative phosphorylation is essential to the growth, survival, and reproduction of eukaryotes. The behavior (position of, and communication between, mitochondria) and morphology of mitochondria play key roles in efficient energy production and are influenced by oxidative stressors such as ultraviolet (UV) radiation. We tested the hypothesis that mitochondria change their behavior and morphology to meet energetic demands of responding to changes in oxidative stress. Specifically, we predicted that UV irradiation would increase the density of inner mitochondrial membrane and proportion of inter-mitochondrial junctions to influence whole-animal metabolic rate. Using transmission electron microscopy, we found that both three and six hours of UV-A/B irradiation (0.5 W/m2) increased the proportion of inter-mitochondrial junctions (with increasing mitochondrial aspect ratio) and the density of inner mitochondrial membrane in myocytes of Tigriopus californicus copepods. Mitochondrial density increased following both irradiation treatments, but mitochondrial size decreased under the six hour treatment. Metabolic rate was maintained under three hours of irradiation but decreased following six hours of exposure. These observations demonstrate that the density of inner mitochondrial membrane and proportion of inter-mitochondrial junctions can play formative roles in maintaining whole-animal metabolic rate, and ultimately organismal performance, under exposure to an oxidative stressor.


Copepoda/cytology , Mitochondria, Muscle/ultrastructure , Mitochondrial Membranes/ultrastructure , Ultraviolet Rays/adverse effects , Animals , Copepoda/radiation effects , Energy Metabolism/radiation effects , Female , Male , Microscopy, Electron, Transmission , Mitochondria, Muscle/radiation effects , Mitochondrial Membranes/radiation effects , Oxidative Phosphorylation , Oxidative Stress
11.
Sci Rep ; 10(1): 21045, 2020 12 03.
Article En | MEDLINE | ID: mdl-33273629

Mitochondria are dynamic organelles that change morphology to adapt to cellular energetic demands under both physiological and stress conditions. Cardiomyopathies and neuronal disorders are associated with structure-related dysfunction in mitochondria, but three-dimensional characterizations of the organelles are still lacking. In this study, we combined high-resolution imaging and 3D electron density information provided by cryo-soft X-ray tomography to characterize mitochondria cristae morphology isolated from murine. Using the linear attenuation coefficient, the mitochondria were identified (0.247 ± 0.04 µm-1) presenting average dimensions of 0.90 ± 0.20 µm in length and 0.63 ± 0.12 µm in width. The internal mitochondria structure was successfully identified by reaching up the limit of spatial resolution of 35 nm. The internal mitochondrial membranes invagination (cristae) complexity was calculated by the mitochondrial complexity index (MCI) providing quantitative and morphological information of mitochondria larger than 0.90 mm in length. The segmentation to visualize the cristae invaginations into the mitochondrial matrix was possible in mitochondria with MCI ≥ 7. Altogether, we demonstrated that the MCI is a valuable quantitative morphological parameter to evaluate cristae modelling and can be applied to compare healthy and disease state associated to mitochondria morphology.


Imaging, Three-Dimensional/methods , Mitochondria, Muscle/ultrastructure , X-Ray Microtomography/methods , Animals , Cells, Cultured , Cryopreservation/methods , Imaging, Three-Dimensional/standards , Limit of Detection , Myocytes, Smooth Muscle/ultrastructure , Rats , X-Ray Microtomography/standards
12.
Nutr Res ; 84: 42-52, 2020 12.
Article En | MEDLINE | ID: mdl-33189431

Glutamine and glucose are both oxidized in the mitochondria to supply the majority of usable energy for processes of cellular function. Low levels of plasma and skeletal muscle glutamine are associated with severe illness. We hypothesized that glutamine deficiency would disrupt mitochondrial integrity and impair cell function. C2C12 mouse myoblasts were cultured in control media supplemented with 5.6 mmol/L glucose and 2 mmol/L glutamine, glutamine depletion (Gln-) or glucose depletion (Glc-) media. We compared mitochondrial morphology and function, as well as cell proliferation, myogenic differentiation, and heat-shock response in these cells. Glc- cells exhibited slightly elongated mitochondrial networks and increased mitochondrial mass, with normal membrane potential (ΔΨm). Mitochondria in Gln- cells became hyperfused and swollen, which were accompanied by severe disruption of cristae and decreases in ΔΨm, mitochondrial mass, the inner mitochondrial membrane remodeling protein OPA1, electron transport chain complex IV protein expression, and markers of mitochondrial biogenesis and bioenergetics. In addition, Gln- increased the autophagy marker LC3B-II on the mitochondrial membrane. Notably, basal mitochondrial respiration was increased in Glc- cells as compared to control cells, whereas maximal respiration remained unchanged. In contrast, basal respiration, maximal respiration and reserve capacity were all decreased in Gln- cells. Consistent with the aforementioned mitochondrial deficits, Gln- cells had lower growth rates and myogenic differentiation, as well as a higher rate of cell death under heat stress conditions than Glc- and control cells. We conclude that glutamine is essential for mitochondrial integrity and function; glutamine depletion impairs myoblast proliferation, differentiation, and the heat-shock response.


Glutamine/metabolism , Heat-Shock Response , Mitochondria, Muscle/metabolism , Myoblasts/physiology , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Energy Metabolism , Mice , Mitochondria, Muscle/ultrastructure , Mitophagy , Muscle Development , Myoblasts/cytology , Myoblasts/metabolism , Myoblasts/ultrastructure , Organelle Biogenesis , Oxygen Consumption
13.
Biomed Pharmacother ; 129: 110482, 2020 Sep.
Article En | MEDLINE | ID: mdl-32768964

The Qiangji Jianli Decoction (QJJLD) is an effective Chinese medicine formula for treating Myasthenia gravis (MG) in the clinic. QJJLD has been proven to regulate mitochondrial fusion and fission of skeletal muscle in myasthenia gravis. In this study, we investigated whether QJJLD plays a therapeutic role in regulating mitochondrial biogenesis in MG and explored the underlying mechanism. Rats were experimentally induced to establish autoimmune myasthenia gravis (EAMG) by subcutaneous immunization with R97-116 peptides. The treatment groups were administered three different dosages of QJJLD respectively. After the intervention of QJJLD, the pathological changes of gastrocnemius muscle in MG rats were significantly improved; SOD, GSH-Px, Na+-K+ ATPase and Ca2+-Mg2+ ATPase activities were increased; and MDA content was decreased in the gastrocnemius muscle. Moreover, AMPK, p38MAPK, PGC-1α, NRF-1, Tfam and COX IV mRNA and protein expression levels were also reversed by QJJLD. These results implied that QJJLD may provide a potential therapeutic strategy through promoting mitochondrial biogenesis to alleviate MG via activating the AMPK/PGC-1α signaling pathway.


AMP-Activated Protein Kinases/metabolism , Drugs, Chinese Herbal/pharmacology , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Female , Gene Expression Regulation , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/genetics , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/enzymology , Muscle, Skeletal/ultrastructure , Myasthenia Gravis, Autoimmune, Experimental/enzymology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , Peptide Fragments , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Rats, Inbred Lew , Receptors, Cholinergic , Signal Transduction
14.
Cell Rep ; 32(5): 107989, 2020 08 04.
Article En | MEDLINE | ID: mdl-32755582

Amyotrophic lateral sclerosis (ALS) manifests pathological changes in motor neurons and various other cell types. Compared to motor neurons, the contribution of the other cell types to the ALS phenotypes is understudied. G4C2 repeat expansion in C9ORF72 is the most common genetic cause of ALS along with frontotemporal dementia (C9-ALS/FTD), with increasing evidence supporting repeat-encoded poly(GR) in disease pathogenesis. Here, we show in Drosophila muscle that poly(GR) enters mitochondria and interacts with components of the Mitochondrial Contact Site and Cristae Organizing System (MICOS), altering MICOS dynamics and intra-subunit interactions. This impairs mitochondrial inner membrane structure, ion homeostasis, mitochondrial metabolism, and muscle integrity. Similar mitochondrial defects are observed in patient fibroblasts. Genetic manipulation of MICOS components or pharmacological restoration of ion homeostasis with nigericin effectively rescue the mitochondrial pathology and disease phenotypes in both systems. These results implicate MICOS-regulated ion homeostasis in C9-ALS pathogenesis and suggest potential new therapeutic strategies.


Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , DNA Repeat Expansion , Frontotemporal Dementia/genetics , Homeostasis , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/metabolism , Animals , Animals, Genetically Modified , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Fibroblasts/metabolism , Fibroblasts/pathology , HEK293 Cells , HeLa Cells , Humans , Ions , Male , Mitochondria, Muscle/ultrastructure , Nigericin/pharmacology , Protein Binding
15.
Am J Physiol Endocrinol Metab ; 318(6): E886-E889, 2020 06 01.
Article En | MEDLINE | ID: mdl-32255679

The measurement of mitochondrial content is essential for bioenergetic research, as it provides a tool to evaluate whether changes in mitochondrial function are strictly due to changes in content or other mechanisms that influence function. In this perspective, we argue that commonly used biomarkers of mitochondrial content may possess limited utility for capturing changes in content with physiological intervention. Moreover, we argue that they may not provide reliable estimates of content in certain pathological situations. Finally, we discuss potential solutions to overcome issues related to the utilization of biomarkers of mitochondrial content. Shedding light on this important issue will hopefully aid conclusions about the mitochondrial structure-function relationship.


Cardiolipins/metabolism , Citrate (si)-Synthase/metabolism , DNA, Mitochondrial/metabolism , Electron Transport Chain Complex Proteins/metabolism , Exercise/physiology , Mitochondria, Muscle/ultrastructure , Mitochondrial Turnover , Muscle Fibers, Skeletal/ultrastructure , Biomarkers , Humans , Microscopy, Electron, Transmission , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Peripheral Arterial Disease/metabolism , Reproducibility of Results
16.
Acta Neuropathol Commun ; 8(1): 32, 2020 03 13.
Article En | MEDLINE | ID: mdl-32169121

Charcot-Marie-Tooth disease (CMT) is a group of inherited neurological disorders of the peripheral nervous system. CMT is subdivided into two main types: a demyelinating form, known as CMT1, and an axonal form, known as CMT2. Nearly 30 genes have been identified as a cause of CMT2. One of these is the 'dehydrogenase E1 and transketolase domain containing 1' (DHTKD1) gene. We previously demonstrated that a nonsense mutation [c.1455 T > G (p.Y485*)] in exon 8 of DHTKD1 is one of the disease-causing mutations in CMT2Q (MIM 615025). The aim of the current study was to investigate whether human disease-causing mutations in the Dhtkd1 gene cause CMT2Q phenotypes in a mouse model in order to investigate the physiological function and pathogenic mechanisms associated with mutations in the Dhtkd1 gene in vivo. Therefore, we generated a knock-in mouse model with the Dhtkd1Y486* point mutation. We observed that the Dhtkd1 expression level in sciatic nerve of knock-in mice was significantly lower than in wild-type mice. Moreover, a histopathological phenotype was observed, reminiscent of a peripheral neuropathy, including reduced large axon diameter and abnormal myelination in peripheral nerves. The knock-in mice also displayed clear sensory defects, while no abnormalities in the motor performance were observed. In addition, accumulation of mitochondria and an elevated energy metabolic state was observed in the knock-in mice. Taken together, our study indicates that the Dhtkd1Y486* knock-in mice partially recapitulate the clinical phenotypes of CMT2Q patients and we hypothesize that there might be a compensatory effect from the elevated metabolic state in the knock-in mice that enables them to maintain their normal locomotor function.


Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Ketoglutarate Dehydrogenase Complex/genetics , Mice , Mitochondria/pathology , Sciatic Nerve/metabolism , Somatosensory Disorders/genetics , Animals , Axons/pathology , Axons/ultrastructure , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/physiopathology , Codon, Nonsense , Energy Metabolism , Gene Knock-In Techniques , Ketoglutarate Dehydrogenase Complex/metabolism , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Neural Conduction , Nonsense Mediated mRNA Decay/genetics , Peripheral Nerves/pathology , Peripheral Nerves/ultrastructure , Phenotype , Point Mutation , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Somatosensory Disorders/pathology , Somatosensory Disorders/physiopathology
17.
Cells ; 9(3)2020 02 28.
Article En | MEDLINE | ID: mdl-32121096

Mitochondrial dysfunction has been implicated as a central mechanism in the metabolic myopathy accompanying critical limb ischemia (CLI). However, whether mitochondrial dysfunction is directly related to lower extremity ischemia and the structural and molecular mechanisms underpinning mitochondrial dysfunction in CLI patients is not understood. Here, we aimed to study whether mitochondrial dysfunction is a distinctive characteristic of CLI myopathy by assessing mitochondrial respiration in gastrocnemius muscle from 14 CLI patients (65.3 ± 7.8 y) and 15 matched control patients (CON) with a similar comorbidity risk profile and medication regimen but without peripheral ischemia (67.4 ± 7.4 y). Furthermore, we studied potential structural and molecular mechanisms of mitochondrial dysfunction by measuring total, sub-population, and fiber-type-specific mitochondrial volumetric content and cristae density with transmission electron microscopy and by assessing mitophagy and fission/fusion-related protein expression. Finally, we asked whether commonly used biomarkers of mitochondrial content are valid in patients with cardiovascular disease. CLI patients exhibited inferior mitochondrial respiration compared to CON. This respiratory deficit was not related to lower whole-muscle mitochondrial content or cristae density. However, stratification for fiber types revealed ultrastructural mitochondrial alterations in CLI patients compared to CON. CLI patients exhibited an altered expression of mitophagy-related proteins but not fission/fusion-related proteins compared to CON. Citrate synthase, cytochrome c oxidase subunit IV (COXIV), and 3-hydroxyacyl-CoA dehydrogenase (ß-HAD) could not predict mitochondrial content. Mitochondrial dysfunction is a distinctive characteristic of CLI myopathy and is not related to altered organelle content or cristae density. Our results link this intrinsic mitochondrial deficit to dysregulation of the mitochondrial quality control system, which has implications for the development of therapeutic strategies.


Extremities/blood supply , Ischemia/complications , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscular Diseases/complications , Muscular Diseases/metabolism , Muscular Diseases/pathology , Aged , Biomarkers/metabolism , Cell Respiration , Extremities/pathology , Female , Humans , Male , Mitochondria, Muscle/ultrastructure , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism
18.
EBioMedicine ; 52: 102637, 2020 Feb.
Article En | MEDLINE | ID: mdl-31981975

BACKGROUND: Canagliflozin (CANA) administration increases the risk of lower limb amputation in the clinic. The present study aimed to investigate whether and how CANA interferes with the intracellular physiological processes of bone marrow derived mesenchymal stem cells (BM-MSCs) and its contribution to ischaemic lower limb. METHODS: The in vivo blood flow recovery in ischaemic lower limbs following CANA treatment was evaluated. The cellular function of BM-MSCs after CANA treatment were also assessed in vitro. In silico docking analysis and mutant substitution assay were conducted to confirm the interaction of CANA with glutamate dehydrogenase 1 (GDH1). FINDINGS: Following CANA treatment, attenuated angiogenesis and hampered blood flow recovery in the ischaemic region were detected in diabetic and non-diabetic mice, and inhibition of the proliferation and migration of BM-MSCs were also observed. CANA was involved in mitochondrial respiratory malfunction in BM-MSCs and the inhibition of ATP production, cytochrome c release and vessel endothelial growth factor A (VEGFA) secretion, which may contribute to reductions in the tissue repair capacity of BM-MSCs. The detrimental effects of CANA on MSCs result from the inhibition of GDH1 by CANA (evidenced by in silico docking analysis and H199A-GDH1/N392A-GDH1 mutant substitution). INTERPRETATION: Our work highlights that the inhibition of GDH1 activity by CANA interferes with the metabolic activity of the mitochondria, and this interference deteriorates the retention of and VEGFA secretion by MSCs. FUNDING: National Natural Science Foundation of China, Natural Science Foundation of Zhejiang Province and Wenzhou Science and Technology Bureau Foundation.


Canagliflozin/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Paracrine Communication/drug effects , Reperfusion Injury/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Binding Sites , Canagliflozin/chemistry , Cell Cycle/drug effects , Cell Movement , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glutamate Dehydrogenase/chemistry , Glutamate Dehydrogenase/metabolism , Humans , Lower Extremity/blood supply , Mesenchymal Stem Cell Transplantation , Mice , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Models, Molecular , Neovascularization, Physiologic/drug effects , Protein Binding , Reperfusion Injury/drug therapy , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Structure-Activity Relationship
19.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165674, 2020 05 01.
Article En | MEDLINE | ID: mdl-31926263

Duchenne muscular dystrophy (DMD) is characterized by a pronounced and progressive degradation of the structure of skeletal muscles, which decreases their strength and lowers endurance of the organism. At muscular dystrophy, mitochondria are known to undergo significant functional changes, which is manifested in a decreased efficiency of oxidative phosphorylation and impaired energy metabolism of the cell. It is believed that the DMD-induced functional changes of mitochondria are mainly associated with the dysregulation of Ca2+ homeostasis. This work examines the kinetic parameters of Ca2+ transport and the opening of the Ca2+-dependent MPT pore in the skeletal-muscle mitochondria of the dystrophin-deficient C57BL/10ScSn-mdx mice. As compared to the organelles of wild-type animals, skeletal-muscle mitochondria of mdx mice have been found to be much less efficient in respect to Ca2+ uniport, with the kinetics of Na+-dependent Ca2+ efflux not changing. The data obtained indicate that the decreased rate of Ca2+ uniport in the mitochondria of mdx mice may be associated with the increased level of the dominant negative subunit of Ca2+ uniporter (MCUb). The experiments have also shown that in mdx mice, skeletal-muscle mitochondria have low resistance to the induction of MPT, which may be related to a significantly increased expression of adenylate translocator (ANT2), a possible structural element of the MPT pore. The paper discusses how changes in the expression of calcium uniporter and putative components of the MPT pore caused by the development of DMD can affect Ca2+ homeostasis of skeletal-muscle mitochondria.


Calcium/metabolism , Mitochondria, Muscle/pathology , Mitochondrial Transmembrane Permeability-Driven Necrosis/genetics , Muscular Dystrophy, Duchenne/pathology , Adenine Nucleotide Translocator 2/genetics , Adenine Nucleotide Translocator 2/metabolism , Animals , Cations, Divalent/metabolism , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Humans , Ion Transport/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred mdx , Microscopy, Electron , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Muscular Dystrophy, Duchenne/genetics , Oxidative Phosphorylation
20.
FASEB J ; 33(11): 12087-12098, 2019 11.
Article En | MEDLINE | ID: mdl-31398297

The main objective of this work was to investigate whether mitochondrial fusion occurs in the skeletal muscle of well-trained athletes in response to high-intensity exercise. Well-trained swimmers (n = 9) performed a duration-matched sprint interval training (SIT) and high-intensity high-volume training (HIHVT) session on separate days. Muscle samples from triceps brachii were taken before, immediately after, and 3 h after the training sessions. Transmission electron microscopy (TEM) was applied to assess mitochondrial morphology. Moreover, expression of genes coding for regulators of mitochondrial fusion and fission were assessed by real-time quantitative PCR. In addition, mitofusin (MFN)2 and optic atrophy 1 (OPA1) were quantified by Western blot analysis. TEM analyses showed that mitochondrial morphology remained altered for 3 h after HIHVT, whereas SIT-induced changes were only evident immediately after exercise. Only SIT increased MFN1 and MFN2 mRNA expression, whereas SIT and HIHVT both increased MFN2 protein content 3 h after exercise. Notably, only HIHVT increased OPA1 protein content. Mitochondrial morphologic changes that suggest fusion occurs in well-adapted athletes during exercise. However, HIHVT appears as a more robust inducer of mitochondrial fusion events than SIT. Indeed, SIT induces a rapid and transient change in mitochondrial morphology.-Huertas, J. R., Ruiz-Ojeda, F. J., Plaza-Díaz, J., Nordsborg, N. B., Martín-Albo, J., Rueda-Robles, A., Casuso, R. A. Human muscular mitochondrial fusion in athletes during exercise.


Athletes , Exercise/physiology , Mitochondria, Muscle/metabolism , Mitochondrial Dynamics/physiology , Muscle, Skeletal/metabolism , Cross-Over Studies , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression , High-Intensity Interval Training , Humans , Male , Microscopy, Electron, Transmission , Mitochondria, Muscle/ultrastructure , Mitochondrial Dynamics/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Swimming , Young Adult
...