Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Biochem Cell Biol ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38843556

RESUMEN

Altered mitochondrial structure and function are implicated in the functional decline of skeletal muscle. Numerous cytoskeletal proteins are known to affect mitochondrial homeostasis, but this complex network is still being unraveled. Here, we investigated mitochondrial alterations in mice lacking the cytoskeletal adapter protein, XIN (XIN-/-). XIN-/- and wild-type littermate male and female mice were fed a chow or high-fat diet (HFD; 60% kcal fat) for 8 weeks before analyses of their skeletal muscles was conducted. Immuno-electron microscopy (EM) and immunofluorescence staining revealed XIN in the mitochondria and peri-mitochondrial areas, as well as the myoplasm. Intermyofibrillar mitochondria in chow-fed XIN-/- mice were notably different from wild-type (large, and/or swollen in appearance). Succinate Dehydrogenase and Cytochrome Oxidase IV staining indicated greater evidence of mitochondrial enzyme activity in XIN-/- mice. No difference in body mass gains or glucose handling was observed between cohorts with HFD. However, EM revealed significantly greater mitochondrial density with evident structural abnormalities (swelling, reduced cristae density) in XIN-/- mice. Absolute Complex I and II-supported respiration was not different between groups, but relative to mitochondrial density, was significantly lower in XIN-/-. These results provide the first evidence for a role of XIN in maintaining mitochondrial morphology and function.

2.
Am J Physiol Cell Physiol ; 321(5): C876-C883, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34586898

RESUMEN

Though preclinical models of type 1 diabetes (T1D) exhibit impaired muscle regeneration, this has yet to be investigated in humans with T1D. Here, we investigated the impact of damaging exercise (eccentric quadriceps contractions) in 18 physically active young adults with and without T1D. Pre- and postexercise (48 h and 96 h), the participants provided blood samples, vastus lateralis biopsies, and performed maximal voluntary quadriceps contractions (MVCs). Skeletal muscle sarcolemmal integrity, extracellular matrix (ECM) content, and satellite cell (SC) content/proliferation were assessed by immunofluorescence. Transmission electron microscopy was used to quantify ultrastructural damage. MVC was comparable between T1D and controls before exercise. Postexercise, MVC was decreased in both groups, but subjects with T1D exhibited moderately lower strength recovery at both 48 h and 96 h. Serum creatine kinase, an indicator of muscle damage, was moderately higher in participants with T1D at rest and exhibited a small elevation 96 h postexercise. Participants with T1D showed lower SC content at all timepoints and demonstrated a moderate delay in SC proliferation after exercise. A greater number of myofibers exhibited sarcolemmal damage (disrupted dystrophin) and increased ECM (laminin) content in participants with T1D despite no differences between groups in ultrastructural damage as assessed by electron microscopy. Finally, transcriptomic analyses revealed dysregulated gene networks involving RNA translation and mitochondrial respiration, providing potential explanations for previous observations of mitochondrial dysfunction in similar cohorts with T1D. Our findings indicate that skeletal muscle in young adults with moderately controlled T1D is altered after damaging exercise, suggesting that longer recovery times following intense exercise may be necessary.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Contracción Muscular , Enfermedades Musculares/etiología , Músculo Cuádriceps/patología , Regeneración , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Proliferación Celular , Creatina Quinasa/sangre , Diabetes Mellitus Tipo 1/diagnóstico , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Densidad Microvascular , Fuerza Muscular , Enfermedades Musculares/sangre , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/fisiopatología , Recuperación de la Función , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Factores de Tiempo , Transcriptoma , Adulto Joven
3.
Diabetologia ; 64(11): 2517-2533, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34392397

RESUMEN

AIMS/HYPOTHESIS: This study interrogated mitochondrial respiratory function and content in skeletal muscle biopsies of healthy adults between 30 and 72 years old with and without uncomplicated type 1 diabetes. METHODS: Participants (12 women/nine men) with type 1 diabetes (48 ± 11 years of age), without overt complications, were matched for age, sex, BMI and level of physical activity to participants without diabetes (control participants) (49 ± 12 years of age). Participants underwent a Bergström biopsy of the vastus lateralis to assess mitochondrial respiratory function using high-resolution respirometry and citrate synthase activity. Electron microscopy was used to quantify mitochondrial content and cristae (pixel) density. RESULTS: Mean mitochondrial area density was 27% lower (p = 0.006) in participants with type 1 diabetes compared with control participants. This was largely due to smaller mitochondrial fragments in women with type 1 diabetes (-18%, p = 0.057), as opposed to a decrease in the total number of mitochondrial fragments in men with diabetes (-28%, p = 0.130). Mitochondrial respiratory measures, whether estimated per milligram of tissue (i.e. mass-specific) or normalised to area density (i.e. intrinsic mitochondrial function), differed between cohorts, and demonstrated sexual dimorphism. Mass-specific mitochondrial oxidative phosphorylation (OXPHOS) capacity with the substrates for complex I and complex II (CI + II) was significantly lower (-24%, p = 0.033) in women with type 1 diabetes compared with control participants, whereas mass-specific OXPHOS capacities with substrates for complex I only (pyruvate [CI pyr] or glutamate [CI glu]) or complex II only (succinate [CII succ]) were not different (p > 0.404). No statistical differences (p > 0.397) were found in mass-specific OXPHOS capacity in men with type 1 diabetes compared with control participants despite a 42% non-significant increase in CI glu OXPHOS capacity (p = 0.218). In contrast, intrinsic CI + II OXPHOS capacity was not different in women with type 1 diabetes (+5%, p = 0.378), whereas in men with type 1 diabetes it was 25% higher (p = 0.163) compared with control participants. Men with type 1 diabetes also demonstrated higher intrinsic OXPHOS capacity for CI pyr (+50%, p = 0.159), CI glu (+88%, p = 0.033) and CII succ (+28%, p = 0.123), as well as higher intrinsic respiratory rates with low (more physiological) concentrations of either ADP, pyruvate, glutamate or succinate (p < 0.012). Women with type 1 diabetes had higher (p < 0.003) intrinsic respiratory rates with low concentrations of succinate only. Calculated aerobic fitness (Physical Working Capacity Test [PWC130]) showed a strong relationship with mitochondrial respiratory function and content in the type 1 diabetes cohort. CONCLUSIONS/INTERPRETATION: In middle- to older-aged adults with uncomplicated type 1 diabetes, we conclude that skeletal muscle mitochondria differentially adapt to type 1 diabetes and demonstrate sexual dimorphism. Importantly, these cellular alterations were significantly associated with our metric of aerobic fitness (PWC130) and preceded notable impairments in skeletal mass and strength.


Asunto(s)
Respiración de la Célula/fisiología , Diabetes Mellitus Tipo 1/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Adulto , Anciano , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosforilación Oxidativa , Consumo de Oxígeno/fisiología , Mecánica Respiratoria
4.
5.
Am J Physiol Endocrinol Metab ; 318(1): E44-E51, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31794260

RESUMEN

Sexual dimorphism in mitochondrial respiratory function has been reported in young women and men without diabetes, which may have important implications for exercise. The purpose of this study was to determine if sexual dimorphism exists in skeletal muscle mitochondrial bioenergetics in people with type 1 diabetes (T1D). A resting muscle microbiopsy was obtained from women and men with T1D (n = 10/8, respectively) and without T1D (control; n = 8/7, respectively). High-resolution respirometry and spectrofluorometry were used to measure mitochondrial respiratory function, hydrogen peroxide (mH2O2) emission and calcium retention capacity (mCRC) in permeabilized myofiber bundles. The impact of T1D on mitochondrial bioenergetics between sexes was interrogated by comparing the change between women and men with T1D relative to the average values of their respective sex-matched controls (i.e., delta). These aforementioned analyses revealed that men with T1D have increased skeletal muscle mitochondrial complex I sensitivity but reduced complex II sensitivity and capacity in comparison to women with T1D. mH2O2 emission was lower in women compared with men with T1D at the level of complex I (succinate driven), whereas mCRC and mitochondrial protein content remained similar between sexes. In conclusion, women and men with T1D exhibit differential responses in skeletal muscle mitochondrial bioenergetics. Although larger cohort studies are certainly required, these early findings nonetheless highlight the importance of considering sex as a variable in the care and treatment of people with T1D (e.g., benefits of different exercise prescriptions).


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Adulto , Calcio/metabolismo , Estudios de Casos y Controles , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Masculino , Caracteres Sexuales , Factores Sexuales , Adulto Joven
6.
Exp Physiol ; 105(4): 565-570, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31826331

RESUMEN

NEW FINDING: What is the topic of this review? Evidence of impaired mitochondrial functions and/or morphology in people with type 1 diabetes across various organ systems. What advances does it highlight? Impairments to mitochondrial functions and morphology may be a primary mechanism underlying the pathophysiology of various complications in people with type 1 diabetes. ABSTRACT: We recently made the observation that there are significant alterations to the ultrastructure and functions of mitochondria in skeletal muscle of people with type 1 diabetes (T1D). These alterations are proposed to lead to decreased energy production in skeletal muscle during exercise and thus may contribute to the impaired aerobic exercise capacity reported in some people with T1D. This Symposium Review summarizes the evidence that similar alterations also occur in the mitochondria present in organ systems outside skeletal muscle in people with T1D, and that this may contribute to the development and progression of the known complications of T1D, which eventually lead to the reported premature mortality.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Ejercicio Físico/fisiología , Humanos
7.
Exerc Sport Sci Rev ; 47(2): 98-107, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30653028

RESUMEN

Recent evidence reveals impairments to skeletal muscle health in adolescent/young adults with type 1 diabetes (T1D). Interestingly, the observed changes in T1D are not unlike aged muscle, particularly, the alterations to mitochondria. Thus, we put forth the novel hypothesis that T1D may be considered a condition of accelerated muscle aging and that, similar to aging, mitochondrial dysfunction is a primary contributor to this complication.


Asunto(s)
Envejecimiento/patología , Diabetes Mellitus Tipo 1/patología , Mitocondrias Musculares/patología , Músculo Esquelético/patología , Humanos , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
8.
Am J Physiol Cell Physiol ; 315(5): C714-C721, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30257107

RESUMEN

Lipocalin-2 (LCN2) is an adipokine previously described for its contribution to numerous processes, including innate immunity and energy metabolism. LCN2 has also been demonstrated to be an extracellular matrix (ECM) regulator through its association with the ECM protease matrix metalloproteinase-9 (MMP-9). With the global rise in obesity and the associated comorbidities related to increasing adiposity, it is imperative to gain an understanding of the cross talk between adipose tissue and other metabolic tissues, such as skeletal muscle. Given the function of LCN2 on the ECM in other tissues and the importance of matrix remodeling in skeletal muscle regeneration, we examined the localization and expression of LCN2 in uninjured and regenerating wild-type skeletal muscle and assessed the impact of LCN2 deletion (LCN2-/-) on skeletal muscle repair following cardiotoxin injury. Though LCN2 was minimally present in uninjured skeletal muscle, its expression was increased significantly at 1 and 2 days postinjury, with expression present in Pax7-positive satellite cells. Although satellite cell content was unchanged, the ability of quiescent satellite cells to become activated was significantly impaired in LCN2-/- skeletal muscles. Skeletal muscle regeneration was also significantly compromised as evidenced by decreased embryonic myosin heavy chain expression and smaller regenerating myofiber areas. Consistent with a role for LCN2 in MMP-9 regulation, regenerating muscle also displayed a significant increase in fibrosis and lower ( P = 0.07) MMP-9 activity in LCN2-/- mice at 2 days postinjury. These data highlight a novel role for LCN2 in muscle regeneration and suggest that changes in adipokine expression can significantly impact skeletal muscle repair.


Asunto(s)
Lipocalina 2/genética , Metaloproteinasa 9 de la Matriz/genética , Músculo Esquelético/crecimiento & desarrollo , Adipoquinas/genética , Adipoquinas/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Regulación de la Expresión Génica/genética , Humanos , Lipocalina 2/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Mioblastos/patología , Regeneración/genética , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Cicatrización de Heridas/genética
9.
Diabetologia ; 61(2): 433-444, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28965129

RESUMEN

AIMS/HYPOTHESIS: While the underlying mechanisms in the development of insulin resistance remain inconclusive, metabolic dysfunction in both white adipose tissue (WAT) and skeletal muscle have been implicated in the process. Therefore, we investigated the independent and combined effects of α-linolenic acid (ALA) supplementation and exercise training on whole-body glucose homeostasis and mitochondrial bioenergetics within the WAT and skeletal muscle of obese Zucker rats. METHODS: We randomly assigned obese Zucker rats to receive a control diet alone or supplemented with ALA and to remain sedentary or undergo exercise training for 4 weeks (CON-Sed, ALA-Sed, CON-Ex and ALA-Ex groups). Whole-body glucose tolerance was determined in response to a glucose load. Mitochondrial content and bioenergetics were examined in skeletal muscle and epididymal WAT (eWAT). Insulin sensitivity and cellular stress were assessed by western blot. RESULTS: Exercise training independently improved whole-body glucose tolerance as well as insulin-induced signalling in muscle and WAT. However, the consumption of ALA during exercise training prevented exercise-mediated improvements in whole-body glucose tolerance. ALA consumption did not influence exercise-induced adaptations within skeletal muscle, insulin sensitivity and mitochondrial bioenergetics. In contrast, within eWAT, ALA supplementation attenuated insulin signalling, decreased mitochondrial respiration and increased the fraction of electron leak to reactive oxygen species (ROS). CONCLUSIONS/INTERPRETATION: These findings indicate that, in an obese rodent model, consumption of ALA attenuates the favourable adaptive changes of exercise training within eWAT, which consequently impacts whole-body glucose homeostasis. The direct translation to humans, however, remains to be determined.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Glucosa/metabolismo , Ácido alfa-Linolénico/uso terapéutico , Animales , Western Blotting , Metabolismo Energético/efectos de los fármacos , Resistencia a la Insulina , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Condicionamiento Físico Animal , Distribución Aleatoria , Ratas , Ratas Zucker
10.
Diabetologia ; 61(6): 1411-1423, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29666899

RESUMEN

AIMS/HYPOTHESIS: A comprehensive assessment of skeletal muscle ultrastructure and mitochondrial bioenergetics has not been undertaken in individuals with type 1 diabetes. This study aimed to systematically assess skeletal muscle mitochondrial phenotype in young adults with type 1 diabetes. METHODS: Physically active, young adults (men and women) with type 1 diabetes (HbA1c 63.0 ± 16.0 mmol/mol [7.9% ± 1.5%]) and without type 1 diabetes (control), matched for sex, age, BMI and level of physical activity, were recruited (n = 12/group) to undergo vastus lateralis muscle microbiopsies. Mitochondrial respiration (high-resolution respirometry), site-specific mitochondrial H2O2 emission and Ca2+ retention capacity (CRC) (spectrofluorometry) were assessed using permeabilised myofibre bundles. Electron microscopy and tomography were used to quantify mitochondrial content and investigate muscle ultrastructure. Skeletal muscle microvasculature was assessed by immunofluorescence. RESULTS: Mitochondrial oxidative capacity was significantly lower in participants with type 1 diabetes vs the control group, specifically at Complex II of the electron transport chain, without differences in mitochondrial content between groups. Muscles of those with type 1 diabetes also exhibited increased mitochondrial H2O2 emission at Complex III and decreased CRC relative to control individuals. Electron tomography revealed an increase in the size and number of autophagic remnants in the muscles of participants with type 1 diabetes. Despite this, levels of the autophagic regulatory protein, phosphorylated AMP-activated protein kinase (p-AMPKαThr172), and its downstream targets, phosphorylated Unc-51 like autophagy activating kinase 1 (p-ULK1Ser555) and p62, was similar between groups. In addition, no differences in muscle capillary density or platelet aggregation were observed between the groups. CONCLUSIONS/INTERPRETATION: Alterations in mitochondrial ultrastructure and bioenergetics are evident within the skeletal muscle of active young adults with type 1 diabetes. It is yet to be elucidated whether more rigorous exercise may help to prevent skeletal muscle metabolic deficiencies in both active and inactive individuals with type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Adulto , Índice de Masa Corporal , Calcio/química , Diabetes Mellitus Tipo 1/patología , Metabolismo Energético , Ejercicio Físico/fisiología , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Insulina/metabolismo , Masculino , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Mitocondrias/ultraestructura , Músculo Esquelético/patología , Consumo de Oxígeno , Adulto Joven
11.
Am J Physiol Regul Integr Comp Physiol ; 315(2): R191-R204, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29513565

RESUMEN

Supplementation with dietary inorganic nitrate ([Formula: see text]) is increasingly recognized to confer cardioprotective effects in both healthy and clinical populations. While the mechanism(s) remains ambiguous, in skeletal muscle oral consumption of NaNO3 has been shown to improve mitochondrial efficiency. Whether NaNO3 has similar effects on mitochondria within the heart is unknown. Therefore, we comprehensively investigated the effect of NaNO3 supplementation on in vivo left ventricular (LV) function and mitochondrial bioenergetics. Healthy male Sprague-Dawley rats were supplemented with NaNO3 (1 g/l) in their drinking water for 7 days. Echocardiography and invasive hemodynamics were used to assess LV morphology and function. Blood pressure (BP) was measured by tail-cuff and invasive hemodynamics. Mitochondrial bioenergetics were measured in LV isolated mitochondria and permeabilized muscle fibers by high-resolution respirometry and fluorometry. Nitrate decreased ( P < 0.05) BP, LV end-diastolic pressure, and maximal LV pressure. Rates of LV relaxation (when normalized to mean arterial pressure) tended ( P = 0.13) to be higher with nitrate supplementation. However, nitrate did not alter LV mitochondrial respiration, coupling efficiency, or oxygen affinity in isolated mitochondria or permeabilized muscle fibers. In contrast, nitrate increased ( P < 0.05) the propensity for mitochondrial H2O2 emission in the absence of changes in cellular redox state and decreased the sensitivity of mitochondria to ADP (apparent Km). These results add to the therapeutic potential of nitrate supplementation in cardiovascular diseases and suggest that nitrate may confer these beneficial effects via mitochondrial redox signaling.


Asunto(s)
Suplementos Dietéticos , Metabolismo Energético/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Nitratos/farmacología , Adenosina Difosfato/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Masculino , Mitocondrias Cardíacas/metabolismo , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Presión Ventricular/efectos de los fármacos
12.
Curr Opin Neurol ; 30(5): 545-552, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28665810

RESUMEN

PURPOSE OF REVIEW: Here we summarize the evidence from human studies of the impairments to the structural, functional, and metabolic capacities in skeletal muscle in those with type 1 diabetes (T1D) - a condition known as diabetic myopathy. Given the importance of skeletal muscle for blood lipid and glucose management, the development and progression of diabetic myopathy would not only lead to increased insulin resistance, but also impact the ability to mitigate dysglycemic/dyslipidemic burdens. RECENT FINDINGS: Despite the importance of skeletal muscle in whole-body metabolic control, studies investigating diabetic myopathy are startling limited. Recent findings have demonstrated that those with T1D exhibit decreased force production, increased fatigability, loss of muscle stem cells, and a greater reliance on glycolytic metabolism, as a result of reduced mitochondrial capacity. SUMMARY: We propose a mechanistic model for the development of diabetic myopathy based on the human findings to date. This model suggests that repeated insulin injections in those with T1D leads to recurrent periods of intracellular hyperglycemia in myofibers. Resultant reductions in mitochondrial function lead to greater reliance on glycolytic metabolism and a concomitant shift in fiber type composition. Studies defining the scope and magnitude of diabetic myopathy and testing the veracity of this model are urgently needed in order to develop appropriate therapeutic strategies to maximize muscle health in those with T1D.


Asunto(s)
Complicaciones de la Diabetes/genética , Complicaciones de la Diabetes/terapia , Enfermedades Musculares/genética , Enfermedades Musculares/terapia , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/terapia , Complicaciones de la Diabetes/patología , Progresión de la Enfermedad , Humanos , Músculo Esquelético/patología , Enfermedades Musculares/etiología , Enfermedades Neuromusculares/patología
13.
Am J Physiol Regul Integr Comp Physiol ; 311(2): R315-24, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27101294

RESUMEN

High-fat diets rapidly cause weight gain and glucose intolerance. We sought to determine whether these changes could be mitigated with prior exercise training. Male C57BL/6J mice were exercise-trained by treadmill running (1 h/day, 5 days/wk) for 4 wk. Twenty-four hours after the final bout of exercise, mice were provided with a high-fat diet (HFD; 60% kcal from lard) for 4 days, with no further exercise. In mice fed the HFD prior to exercise training, the results were blunted weight gain, reduced fat mass, and a slight attenuation in glucose intolerance that was mirrored by greater insulin-induced Akt phosphorylation in skeletal muscle compared with sedentary mice fed the HFD. When ad libitum-fed sedentary mice were compared with sedentary high-fat fed mice that were calorie restricted (-30%) to match the weight gain of the previously trained high-fat fed mice, the same attenuated impairments in glucose tolerance were found. Blunted weight gain was associated with a greater capacity to increase energy expenditure in trained compared with sedentary mice when challenged with a HFD. Although mitochondrial enzymes in white adipose tissue and UCP-1 protein content in brown adipose tissue were increased in previously exercised compared with sedentary mice fed a HFD, ex vivo mitochondrial respiration was not increased in either tissue. Our data suggest that prior exercise training attenuates high-fat diet-induced weight gain and glucose intolerance and is associated with a greater ability to increase energy expenditure in response to a high-fat diet.


Asunto(s)
Dieta Alta en Grasa/métodos , Grasas de la Dieta/farmacocinética , Metabolismo Energético/fisiología , Condicionamiento Físico Animal/métodos , Aumento de Peso/fisiología , Animales , Glucosa/farmacocinética , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL
14.
J Diabetes Complications ; 38(8): 108798, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38991492

RESUMEN

AIMS: Type 1 diabetes has been associated with mitochondrial dysfunction. However, the mechanism of this dysfunction in adults remains unclear. METHODS: A secondary analysis was conducted using data from several clinical trials measuring in-vivo and ex-vivo mitochondrial function in adults with type 1 diabetes (n = 34, age 38.8 ± 14.6 years) and similarly aged controls (n = 59, age 44.6 ± 13.9 years). In-vivo mitochondrial function was assessed before, during, and after isometric exercise with 31phosphorous magnetic resonance spectroscopy. High resolution respirometry of vastus lateralis muscle tissue was used to assess ex-vivo measures. RESULTS: In-vivo data showed higher rates of anaerobic glycolysis (p = 0.013), and a lower maximal mitochondrial oxidative capacity (p = 0.012) and mitochondrial efficiency (p = 0.024) in adults with type 1 diabetes. After adjustment for age and percent body fat maximal mitochondrial capacity (p = 0.014) continued to be lower and anaerobic glycolysis higher (p = 0.040) in adults with type 1 diabetes. Ex-vivo data did not demonstrate significant differences between the two groups. CONCLUSIONS: The in-vivo analysis demonstrates that adults with type 1 diabetes have mitochondrial dysfunction. This builds on previous research showing in-vivo mitochondrial dysfunction in youths with type 1 diabetes and suggests that defects in substrate or oxygen delivery may play a role in in-vivo dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 1 , Mitocondrias Musculares , Humanos , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/fisiopatología , Adulto , Masculino , Femenino , Persona de Mediana Edad , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Glucólisis/fisiología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Enfermedades Mitocondriales/complicaciones , Estudios de Casos y Controles , Espectroscopía de Resonancia Magnética , Adulto Joven , Ejercicio Físico/fisiología
15.
J Clin Endocrinol Metab ; 106(8): 2405-2422, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-33890059

RESUMEN

CONTEXT: Previous investigations on skeletal muscle health in type 1 diabetes (T1D) have generally focused on later stages of disease progression where comorbidities are present and are posited as a primary mechanism of muscle dysfunction. OBJECTIVE: To investigate skeletal muscle function and morphology across the adult lifespan in those with and without T1D. DESIGN: Participants underwent maximal contraction (MVC) testing, resting muscle biopsy, and venous blood sampling. SETTING: Procedures in this study were undertaken at the McMaster University Medical Centre. PARTICIPANTS: Sixty-five healthy adult (18-78 years old) men/males and women/females (T1D = 34; control = 31) matched for age/biological sex/body mass index; self-reported physical activity levels were included. MAIN OUTCOME MEASURES: Our primary measure in this study was MVC, with supporting histological/immunofluorescent measures. RESULTS: After 35 years of age ("older adults"), MVC declined quicker in T1D subjects compared to controls. Loss of strength in T1D was accompanied by morphological changes associated with accelerated aging. Type 1 myofiber grouping was higher in T1D, and the groups were larger and more numerous than in controls. Older T1D females exhibited more myofibers expressing multiple myosin heavy chain isoforms (hybrid fibers) than controls, another feature of accelerated aging. Conversely, T1D males exhibited a shift toward type 2 fibers, with less evidence of myofiber grouping or hybrid fibers. CONCLUSIONS: These data suggest impairments to skeletal muscle function and morphology exist in T1D. The decline in strength with T1D is accelerated after 35 years of age and may be responsible for the earlier onset of frailty, which characterizes those with diabetes.


Asunto(s)
Envejecimiento/fisiología , Diabetes Mellitus Tipo 1/fisiopatología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/fisiopatología , Adolescente , Adulto , Anciano , Diabetes Mellitus Tipo 1/patología , Ejercicio Físico/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Factores Sexuales , Adulto Joven
16.
J Neurol Sci ; 413: 116816, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32272361

RESUMEN

Lipofuscin aggregation may result from incomplete degradation of damaged mitochondria by autophagy-lysosome pathway, and intra-mitochondrial lipofuscin aggregation may exacerbate mitochondrial abnormalities in mitochondrial myopathy (MM) and mitochondrial disease. We examined vastus lateralis muscle biopsies from 24 patients with pathologically diagnosed MM and clinically diagnosed chronic progressive external ophthalmoplegia, in comparison to the biopsies from 3 other groups:10 patients with inclusion body myositis (IBM), 11 younger adults, and 10 older subjects with no to minimal myopathic changes. Lipofuscin aggregation in muscle fibres was assessed on autofluorescence microscopy, some histochemical stains, and electron microscopy (EM). EM analyses demonstrated intra-mitochondrial lipofuscin aggregates, spherical dense bodies (SDBs), and paracrystalline inclusions (PCIs) which were semi-quantitatively assessed. Intra-mitochondrial lipofuscin aggregates showed no significant differences between groups of MM patients and older subjects or IBM patients, but significant differences between groups of younger adults and others with associated age-related changes. Intra-mitochondrial SDBs were significantly more in MM patients than in older subjects, IBM patients, and younger adults. There was a significant positive correlation between intra-mitochondrial lipofuscin aggregates and SDBs. These findings suggest that intra-mitochondrial formation of lipofuscin SDBs is more in MM and contributing to the pathophysiology of mitochondrial disease.


Asunto(s)
Miopatías Mitocondriales , Miositis por Cuerpos de Inclusión , Oftalmoplejía Externa Progresiva Crónica , Adulto , Anciano , Humanos , Lipofuscina , Mitocondrias
17.
Physiol Rep ; 8(13): e14500, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32652899

RESUMEN

Type 1 diabetes (T1D) has been reported to negatively affect the health of skeletal muscle, though the underlying mechanisms are unknown. Myostatin, a myokine whose increased expression is associated with muscle-wasting diseases, has not been reported in humans with T1D but has been demonstrated to be elevated in preclinical diabetes models. Thus, the purpose of this study was to determine if there is an elevated expression of myostatin in the serum and skeletal muscle of persons with T1D compared to controls. Secondarily, we aimed to explore relationships between myostatin expression and clinically important metrics (e.g., HbA1c , strength, lean mass) in women and men with (N = 31)/without T1D (N = 24) between 18 and 72 years old. Body composition, baseline strength, blood sample and vastus lateralis muscle biopsy were evaluated. Serum, but not muscle, myostatin expression was significantly elevated in those with T1D versus controls, and to a greater degree in T1D women than T1D men. Serum myostatin levels were not significantly associated with HbA1c nor disease duration. A significant correlation between serum myostatin expression and maximal voluntary contraction (MVC) and body fat mass was demonstrated in control subjects, but these correlations did not reach significance in those with T1D (MVC: R = 0.64 controls vs. R = 0.37 T1D; Body fat: R = -0.52 controls/R = -0.02 T1D). Collectively, serum myostatin was correlated with lean mass (R = 0.45), and while this trend was noted in both groups separately, neither reached statistical significance (R = 0.47 controls/R = 0.33 T1D). Overall, while those with T1D exhibited elevated serum myostatin levels (particularly females) myostatin expression was not correlated with clinically relevant metrics despite some of these relationships existing in controls (e.g., lean/fat mass). Future studies will be needed to fully understand the mechanisms underlying increased myostatin in T1D, with relationships to insulin dosing being particularly important to elucidate.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Adiposidad , Adolescente , Adulto , Anciano , Diabetes Mellitus Tipo 1/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Contracción Muscular , Músculo Esquelético/fisiopatología , Miostatina/sangre , Miostatina/genética , Factores Sexuales
18.
Appl Physiol Nutr Metab ; 45(9): 1049-1053, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32379978

RESUMEN

Dietary nitrate has been shown to increase cytosolic calcium concentrations within the heart, which would necessitate greater calcium sequestration for relaxation. In the present study we demonstrate that while nitrate supplementation reduced blood pressure, calcium-handling protein content, sarco(endo)plasmic reticulum Ca-ATPase 2a (SERCA) enzymatic properties, and left ventricular function were not altered. In addition, nitrite did not alter in vitro SERCA activity. Combined, these data suggest that in healthy rats, dietary nitrate does not increase left ventricle SERCA-related calcium-handling properties. Novelty Dietary nitrate decreases blood pressure but does not alter left ventricular calcium-handling protein content or SERCA activity in healthy rats.


Asunto(s)
Nitratos/administración & dosificación , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Función Ventricular , Animales , Presión Sanguínea , Calcio , Dieta , Ventrículos Cardíacos , Masculino , Ratas , Ratas Sprague-Dawley
19.
J Cachexia Sarcopenia Muscle ; 10(3): 643-661, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30938481

RESUMEN

BACKGROUND: Muscle wasting and weakness in Duchenne muscular dystrophy (DMD) causes severe locomotor limitations and early death due in part to respiratory muscle failure. Given that current clinical practice focuses on treating secondary complications in this genetic disease, there is a clear need to identify additional contributions in the aetiology of this myopathy for knowledge-guided therapy development. Here, we address the unresolved question of whether the complex impairments observed in DMD are linked to elevated mitochondrial H2 O2 emission in conjunction with impaired oxidative phosphorylation. This study performed a systematic evaluation of the nature and degree of mitochondrial-derived H2 O2 emission and mitochondrial oxidative dysfunction in a mouse model of DMD by designing in vitro bioenergetic assessments that attempt to mimic in vivo conditions known to be critical for the regulation of mitochondrial bioenergetics. METHODS: Mitochondrial bioenergetics were compared with functional and histopathological indices of myopathy early in DMD (4 weeks) in D2.B10-DMDmdx /2J mice (D2.mdx)-a model that demonstrates severe muscle weakness. Adenosine diphosphate's (ADP's) central effect of attenuating H2 O2 emission while stimulating respiration was compared under two models of mitochondrial-cytoplasmic phosphate exchange (creatine independent and dependent) in muscles that stained positive for membrane damage (diaphragm, quadriceps, and white gastrocnemius). RESULTS: Pathway-specific analyses revealed that Complex I-supported maximal H2 O2 emission was elevated concurrent with a reduced ability of ADP to attenuate emission during respiration in all three muscles (mH2 O2 : +17 to +197% in D2.mdx vs. wild type). This was associated with an impaired ability of ADP to stimulate respiration at sub-maximal and maximal kinetics (-17 to -72% in D2.mdx vs. wild type), as well as a loss of creatine-dependent mitochondrial phosphate shuttling in diaphragm and quadriceps. These changes largely occurred independent of mitochondrial density or abundance of respiratory chain complexes, except for quadriceps. This muscle was also the only one exhibiting decreased calcium retention capacity, which indicates increased sensitivity to calcium-induced permeability transition pore opening. Increased H2 O2 emission was accompanied by a compensatory increase in total glutathione, while oxidative stress markers were unchanged. Mitochondrial bioenergetic dysfunctions were associated with induction of mitochondrial-linked caspase 9, necrosis, and markers of atrophy in some muscles as well as reduced hindlimb torque and reduced respiratory muscle function. CONCLUSIONS: These results provide evidence that Complex I dysfunction and loss of central respiratory control by ADP and creatine cause elevated oxidant generation during impaired oxidative phosphorylation. These dysfunctions may contribute to early stage disease pathophysiology and support the growing notion that mitochondria are a potential therapeutic target in this disease.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Peróxido de Hidrógeno/metabolismo , Mitocondrias/patología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Animales , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/citología , Distrofia Muscular de Duchenne/genética , Oxidación-Reducción , Fosforilación Oxidativa , Estrés Oxidativo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA