Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Clin Endocrinol Metab ; 109(7): 1754-1764, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38242693

RESUMEN

CONTEXT: Regular exercise is a key prevention strategy for obesity and type 2 diabetes (T2D). Exerkines secreted in response to exercise or recovery may contribute to improved systemic metabolism. Conversely, an impaired exerkine response to exercise and recovery may contribute to cardiometabolic diseases. OBJECTIVE: We investigated if the exercise-induced regulation of the exerkine, growth differentiation factor 15 (GDF15) and its putative upstream regulators of the unfolded protein response (UPR)/integrated stress response (ISR) is impaired in skeletal muscle in patients with T2D compared with weight-matched glucose-tolerant men. METHODS: Thirteen male patients with T2D and 14 age- and weight-matched overweight/obese glucose-tolerant men exercised at 70% of VO2max for 1 hour. Blood and skeletal muscle biopsies were sampled before, immediately after, and 3 hours into recovery. Serum and muscle transcript levels of GDF15 and key markers of UPR/ISR were determined. Additionally, protein/phosphorylation levels of key regulators in UPR/ISR were investigated. RESULTS: Acute exercise increased muscle gene expression and serum GDF15 levels in both groups. In recovery, muscle expression of GDF15 decreased toward baseline, whereas serum GDF15 remained elevated. In both groups, acute exercise increased the expression of UPR/ISR markers, including ATF4, CHOP, EIF2K3 (encoding PERK), and PPP1R15A (encoding GADD34), of which only CHOP remained elevated 3 hours into recovery. Downstream molecules of the UPR/ISR including XBP1-U, XBP1-S, and EDEM1 were increased with exercise and 3 hours into recovery in both groups. The phosphorylation levels of eIF2α-Ser51, a common marker of unfolded protein response (UPR) and ISR, increased immediately after exercise in controls, but decreased 3 hours into recovery in both groups. CONCLUSION: In conclusion, exercise-induced regulation of GDF15 and key markers of UPR/ISR are not compromised in patients with T2D compared with weight-matched controls.


Asunto(s)
Diabetes Mellitus Tipo 2 , Ejercicio Físico , Factor 15 de Diferenciación de Crecimiento , Músculo Esquelético , Respuesta de Proteína Desplegada , Humanos , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/sangre , Factor 15 de Diferenciación de Crecimiento/genética , Masculino , Diabetes Mellitus Tipo 2/metabolismo , Respuesta de Proteína Desplegada/fisiología , Músculo Esquelético/metabolismo , Ejercicio Físico/fisiología , Persona de Mediana Edad , Adulto , Estudios de Casos y Controles , Estrés Fisiológico/fisiología
2.
Rev Endocr Metab Disord ; 24(6): 1121-1133, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37558853

RESUMEN

White adipose tissue (WAT) is an important endocrine organ that regulates systemic energy metabolism. In metabolically unhealthy obesity, adipocytes become dysfunctional through hypertrophic mechanisms associated with a reduced endocrine function, reduced mitochondrial function, but increased inflammation, fibrosis, and extracellular remodelling. A pathologic WAT remodelling promotes systemic lipotoxicity characterized by fat accumulation in tissues such as muscle and liver, leading to systemic insulin resistance and type 2 diabetes. Several lines of evidence from human and animal studies suggest a link between unhealthy obesity and adipocyte mitochondrial dysfunction, and interventions that improve mitochondrial function may reduce the risk of obesity-associated diseases. This review discusses the importance of mitochondrial function and metabolism in human adipocyte biology and intercellular communication mechanisms within WAT. Moreover, a selected interventional approach for better adipocyte mitochondrial metabolism in humans is reviewed. A greater understanding of mitochondrial bioenergetics in WAT might provide novel therapeutic opportunities to prevent or restore dysfunctional adipose tissue in obesity-associated diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedades Metabólicas , Animales , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo/metabolismo , Metabolismo Energético , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Mitocondrias/metabolismo
3.
Int J Obes (Lond) ; 47(7): 554-563, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029208

RESUMEN

A bidirectional relationship exists between adipose tissue metabolism and iron regulation. Total body fat, fat distribution and exercise influence iron status and components of the iron-regulatory pathway, including hepcidin and erythroferrone. Conversely, whole body and tissue iron stores associate with fat mass and distribution and glucose and lipid metabolism in adipose tissue, liver, and muscle. Manipulation of the iron-regulatory proteins erythroferrone and erythropoietin affects glucose and lipid metabolism. Several lines of evidence suggest that iron accumulation and metabolism may play a role in the development of metabolic diseases including obesity, type 2 diabetes, hyperlipidaemia and non-alcoholic fatty liver disease. In this review we summarise the current understanding of the relationship between iron homoeostasis and metabolic disease.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Humanos , Glucosa/metabolismo , Hierro/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Obesidad/metabolismo , Hígado/metabolismo , Metabolismo de los Lípidos/fisiología
4.
Cells ; 11(13)2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35805088

RESUMEN

Insulin resistance in skeletal muscle in type 2 diabetes (T2D) is characterized by more pronounced metabolic and molecular defects than in obesity per se. There is increasing evidence that adipose tissue dysfunction contributes to obesity-induced insulin resistance in skeletal muscle. Here, we used an unbiased approach to examine if adipose tissue dysfunction is exaggerated in T2D and linked to diabetes-related mechanisms of insulin resistance in skeletal muscle. Transcriptional profiling and biological pathways analysis were performed in subcutaneous adipose tissue (SAT) and skeletal muscle biopsies from 17 patients with T2D and 19 glucose-tolerant, age and weight-matched obese controls. Findings were validated by qRT-PCR and western blotting of selected genes and proteins. Patients with T2D were more insulin resistant and had lower plasma adiponectin than obese controls. Transcriptional profiling showed downregulation of genes involved in mitochondrial oxidative phosphorylation and the tricarboxylic-acid cycle and increased expression of extracellular matrix (ECM) genes in SAT in T2D, whereas genes involved in proteasomal degradation were upregulated in the skeletal muscle in T2D. qRT-PCR confirmed most of these findings and showed lower expression of adiponectin in SAT and higher expression of myostatin in muscle in T2D. Interestingly, muscle expression of proteasomal genes correlated positively with SAT expression of ECM genes but inversely with the expression of ADIPOQ in SAT and plasma adiponectin. Protein content of proteasomal subunits and major ubiquitin ligases were unaltered in the skeletal muscle of patients with T2D. A transcriptional signature of exaggerated adipose tissue dysfunction in T2D, compared with obesity alone, is linked to low plasma adiponectin and increased transcriptional activation of proteasomal degradation in skeletal muscle.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Humanos , Resistencia a la Insulina/genética , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Activación Transcripcional
5.
Cell Rep ; 40(4): 111136, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35905723

RESUMEN

Mechanisms governing regional human adipose tissue (AT) development remain undefined. Here, we show that the long non-coding RNA HOTAIR (HOX transcript antisense RNA) is exclusively expressed in gluteofemoral AT, where it is essential for adipocyte development. We find that HOTAIR interacts with polycomb repressive complex 2 (PRC2) and we identify core HOTAIR-PRC2 target genes involved in adipocyte lineage determination. Repression of target genes coincides with PRC2 promoter occupancy and H3K27 trimethylation. HOTAIR is also involved in modifying the gluteal adipocyte transcriptome through alternative splicing. Gluteal-specific expression of HOTAIR is maintained by defined regions of open chromatin across the HOTAIR promoter. HOTAIR expression levels can be modified by hormonal (estrogen, glucocorticoids) and genetic variation (rs1443512 is a HOTAIR eQTL associated with reduced gynoid fat mass). These data identify HOTAIR as a dynamic regulator of the gluteal adipocyte transcriptome and epigenome with functional importance for human regional AT development.


Asunto(s)
Complejo Represivo Polycomb 2 , ARN Largo no Codificante/genética , Cromatina , Estrógenos , Humanos , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante/metabolismo , Transcriptoma/genética
6.
Metabolism ; 133: 155240, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35697299

RESUMEN

INTRODUCTION: Non-coding genetic variation at TCF7L2 is the strongest genetic determinant of type 2 diabetes (T2D) risk in humans. TCF7L2 encodes a transcription factor mediating the nuclear effects of WNT signaling in adipose tissue (AT). In vivo studies in transgenic mice have highlighted important roles for TCF7L2 in adipose tissue biology and systemic metabolism. OBJECTIVE: To map the expression of TCF7L2 in human AT, examine its role in human adipose cell biology in vitro, and investigate the effects of the fine-mapped T2D-risk allele at rs7903146 on AT morphology and TCF7L2 expression. METHODS: Ex vivo gene expression studies of TCF7L2 in whole and fractionated human AT. In vitro TCF7L2 gain- and/or loss-of-function studies in primary and immortalized human adipose progenitor cells (APCs) and mature adipocytes (mADs). AT phenotyping of rs7903146 T2D-risk variant carriers and matched controls. RESULTS: Adipose progenitors (APs) exhibited the highest TCF7L2 mRNA abundance compared to mature adipocytes and adipose-derived endothelial cells. Obesity was associated with reduced TCF7L2 transcript levels in whole subcutaneous abdominal AT but paradoxically increased expression in APs. In functional studies, TCF7L2 knockdown (KD) in abdominal APs led to dose-dependent activation of WNT/ß-catenin signaling, impaired proliferation and dose-dependent effects on adipogenesis. Whilst partial KD enhanced adipocyte differentiation, near-total KD impaired lipid accumulation and adipogenic gene expression. Over-expression of TCF7L2 accelerated adipogenesis. In contrast, TCF7L2-KD in gluteal APs dose-dependently enhanced lipid accumulation. Transcriptome-wide profiling revealed that TCF7L2 might modulate multiple aspects of AP biology including extracellular matrix secretion, immune signaling and apoptosis. The T2D-risk allele at rs7903146 was associated with reduced AP TCF7L2 expression and enhanced AT insulin sensitivity. CONCLUSIONS: TCF7L2 plays a complex role in AP biology and has both dose- and depot-dependent effects on adipogenesis. In addition to regulating pancreatic insulin secretion, genetic variation at TCF7L2 might also influence T2D risk by modulating AP function.


Asunto(s)
Tejido Adiposo , Diabetes Mellitus Tipo 2 , Proteína 2 Similar al Factor de Transcripción 7 , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Metabolismo de los Lípidos , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
7.
Acta Physiol (Oxf) ; 234(2): e13766, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34981891

RESUMEN

Exercise activates a plethora of metabolic and signalling pathways in skeletal muscle and other organs causing numerous systemic beneficial metabolic effects. Thus, regular exercise may ameliorate and prevent the development of several chronic metabolic diseases. Skeletal muscle is recognized as an important endocrine organ regulating systemic adaptations to exercise. Skeletal muscle may mediate crosstalk with other organs through the release of exercise-induced cytokines, peptides and proteins, termed myokines, into the circulation. Importantly, other tissues such as the liver and adipose tissue may also release cytokines and peptides in response to exercise. Hence, exercise-released molecules are collectively called exerkines. Moreover, extracellular vesicles (EVs), in the form of exosomes or microvesicles, may carry some of the signals involved in tissue crosstalk. This review focuses on the role of factors potentially mediating crosstalk between muscle and other tissues in response to exercise.


Asunto(s)
Ejercicio Físico , Vesículas Extracelulares , Tejido Adiposo/metabolismo , Citocinas/metabolismo , Ejercicio Físico/fisiología , Vesículas Extracelulares/metabolismo , Humanos , Músculo Esquelético/metabolismo
8.
Front Endocrinol (Lausanne) ; 12: 681290, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34456860

RESUMEN

Obesity is characterized by pathological adipose tissue (AT) expansion. While healthy AT expansion enhances systemic insulin sensitivity, unhealthy AT expansion through increased adipocyte size is associated with insulin resistance, fibrosis, hypoxia, and reduced adipose-derived adiponectin secretion. The mechanisms causing the unhealthy AT expansion are not fully elucidated; yet, dysregulated crosstalk between cells within the AT is an important contributor. Evidence from animal and human studies suggests a crucial role of the crosstalk between vascular endothelium (the innermost cell type in blood vessels) and adipocytes for metabolic homeostasis. Arterial endothelial cells are directly involved in maintaining normal organ functions through local blood flow regulation. The endothelial-dependent regulation of blood flow in AT is hampered in obesity, which negatively affects the adipocyte. Moreover, endothelial cells secrete extracellular vesicles (EVs) that target adipocytes in vivo. The endothelial EVs secretion is hampered in obesity and may be affected by the adipocyte-derived adipokine adiponectin. Adiponectin targets the vascular endothelium, eliciting organ-protective functions through binding to T-cadherin. The reduced obesity-induced adiponectin binding of T-cadherin reduces endothelial EV secretion. This affects endothelial health and cell-cell communication between AT cells and distant organs, influencing systemic energy homeostasis. This review focuses on the current understanding of endothelial and adipocyte crosstalk. We will discuss how obesity changes the AT environment and how these changes contribute to obesity-associated metabolic disease in humans. Particularly, we will describe and discuss the EV-dependent communication and regulation between adipocytes, adiponectin, and the endothelial cells regulating systemic energy homeostasis in health and metabolic disease in humans.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Obesidad/metabolismo , Animales , Humanos
9.
Metabolism ; 112: 154347, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32853647

RESUMEN

BACKGROUND: Long-term testosterone replacement therapy (TRT) increases muscle mass in elderly men with subnormal testosterone levels. However, the molecular mechanisms underlying this effect of TRT on protein balance in human skeletal muscle in vivo remain to be established. METHODS: Here, we examined skeletal muscle biopsies obtained before and 24-h after the last dose of treatment with either testosterone gel (n = 12) or placebo (n = 13) for 6 months in aging men with subnormal bioavailable testosterone levels. The placebo-controlled, testosterone-induced changes (ß-coefficients) in mRNA levels, protein expression and phosphorylation were examined by quantitative real-time PCR and western blotting. RESULTS: Long-term TRT increased muscle mass by ß = 1.6 kg (p = 0.01) but had no significant effect on mRNA levels of genes involved in myostatin/activin/SMAD or IGF1/FOXO3 signalling, muscle-specific E3-ubiquitin ligases, upstream transcription factors (MEF2C, PPARGC1A-4) or myogenic factors. However, TRT caused a sustained decrease in protein expression of SMAD2 (ß = -36%, p = 0.004) and SMAD3 (ß = -32%, p = 0.001), which was accompanied by reduced protein expression of the muscle-specific E3-ubiquitin ligases, MuRF1 (ß = -26%, p = 0.004) and Atrogin-1/MAFbx (ß = -20%, p = 0.04), but with no changes in FOXO3 signalling. Importantly, TRT did not affect muscle fibre type distribution between slow-oxidative (type 1), fast-oxidative (type 2a) and fast-glycolytic (type 2×) muscle fibres. CONCLUSIONS: Our results indicate that long-term TRT of elderly men with subnormal testosterone levels increases muscle mass, at least in part, by decreasing protein breakdown through the ubiquitin proteasome pathway mediated by a sustained suppression of SMAD-signalling and muscle-specific E3-ubiquitin ligases.


Asunto(s)
Terapia de Reemplazo de Hormonas , Músculo Esquelético/efectos de los fármacos , Testosterona/administración & dosificación , Anciano , Envejecimiento , Composición Corporal/efectos de los fármacos , Humanos , Masculino , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Fosforilación/efectos de los fármacos , Globulina de Unión a Hormona Sexual/metabolismo , Testosterona/sangre , Resultado del Tratamiento
10.
Acta Physiol (Oxf) ; 228(1): e13346, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31334916

RESUMEN

Urinary extracellular vesicles (uEVs) are a heterogenous group of vesicles consisting mainly of microvesicles and exosomes that originate predominantly (99.96%) from kidney, the urinary tract epithelium and the male reproductive tract. Secreted EVs contain molecular cargo from parental cells and provide an attractive source for biomarkers, a potential readout of physiological and pathophysiological mechanisms, and events associated with the urinary system. uEVs are readily enriched and isolated from urine samples and we review 6 standard methods that allow for downstream analysis of the uEV cargo. Although the use of uEVs as a surrogate readout for physiological changes in tissue protein levels is widespread, the protein abundance in uEVs is affected significantly by mechanisms that regulate protein sorting and secretion in uEVs. Data suggest that baseline kidney tissue and uEV levels of apical membrane-associated electrolyte transport proteins are not directly related in human patients. Recent evidence indicates that EVs may contribute to physiological and pathophysiological intercellular signalling and EVs confer protection against renal ischemia-reperfusion injury. The therapeutic use of EVs as information carriers has mainly been explored in vitro and a major hurdle lies in the translation of the in vitro findings into an in vivo setting. Thus, the EV research field is moving from a technical focus to a more physiological focus, allowing for a deeper understanding of human physiology, development of diagnostic tools and potential treatment strategies for precision medicine.


Asunto(s)
Vesículas Extracelulares/fisiología , Animales , Biomarcadores/orina , Enfermedades Urológicas/diagnóstico , Enfermedades Urológicas/orina
11.
Am J Physiol Renal Physiol ; 317(3): F560-F571, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31241991

RESUMEN

Human urinary extracellular vesicles (uEVs) contain proteins from all nephron segments. An assumption for years has been that uEVs might provide a noninvasive liquid biopsy that reflect physiological regulation of transporter protein expression in humans. We hypothesized that protein abundance in human kidney tissue and uEVs are directly related and tested this in paired collections of nephrectomy tissue and urine sample from 12 patients. Kidney tissue was fractioned into total kidney protein, crude membrane (plasma membrane and large intracellular vesicles)-enriched, and intracellular vesicle-enriched fractions as well as sections for immunolabeling. uEVs were isolated from spot urine samples. Antibodies were used to quantify six segment-specific proteins [proximal tubule-expressed Na+-phosphate cotransporters (NaPi-2a), thick ascending limb-expressed Tamm-Horsfall protein and renal outer medullary K+ channels, distal convoluted tubule-expressed NaCl cotransporters, intercalated cell-expressed V-type H+-ATPase subunit G3 (ATP6V1G3), and principal cell-expressed aquaporin 2] and three uEV markers (exosomal CD63, microvesicle marker vesicle-associated membrane protein 3, and ß-actin) in each fraction. By Western blot analysis and immunofluorescence labeling, we found significant positive correlations between the abundance of CD63, NaCl cotransporters, aquaporin 2, and ATP6V1G3, respectively, within the different kidney-derived fractions. We detected all nine proteins in uEVs, but their level did not correlate with kidney tissue protein abundance. uEV protein levels showed higher interpatient variability than kidney-derived fractions, indicating that factors, besides kidney protein abundance, contribute to the uEV protein level. Our data suggest that, in a random sample of nephrectomy patients, uEV protein level is not a predictor of kidney protein abundance.


Asunto(s)
Células Epiteliales/química , Vesículas Extracelulares/química , Túbulos Renales/química , Proteínas de Transporte de Membrana/orina , Biomarcadores/orina , Humanos , Túbulos Renales/cirugía , Nefrectomía
12.
J Clin Endocrinol Metab ; 104(10): 4909-4920, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31135885

RESUMEN

CONTEXT: Type 2 diabetes (T2D) is characterized by insulin resistance in skeletal muscle. Regular exercise improves insulin sensitivity, mitochondrial function, and energy metabolism. Thus, an impaired response to exercise may contribute to insulin resistance. OBJECTIVE: We hypothesized that key transcriptional regulators of metabolic adaptation to exercise show an attenuated response in skeletal muscle in T2D. DESIGN AND PATIENTS: Skeletal muscle biopsies were obtained from 13 patients with T2D and 14 age- and weight-matched controls before, immediately after 1 hour acute exercise (70% maximal pulmonary oxygen uptake), and 3 hours into recovery to examine mRNA expression of key transcription factors and downstream targets and activity of key upstream kinases underlying the metabolic adaptation to exercise. RESULTS: Acute exercise increased gene expression of the nuclear hormone receptor 4A (NR4A) subfamily (∼4- to 36-fold) and other key transcription factors, including ATF3, EGR1, JUNB, SIK1, PPARA, and PPARG (∼1.5- to 12-fold), but with no differences between groups. The expression of NR4A1 (approximately eightfold) and NR4A3 (∼75-fold) was further increased 3 hours into recovery, whereas most muscle transcripts sustained elevated or returned to basal levels, again with no differences between groups. Muscle expression of HKII and SLC2A4 and hexokinase II protein content were reduced in patients with T2D. The phosphorylation of p38 MAPK, Erk1/2, Ca2+/calmodulin-dependent kinase II, and cAMP-responsive element-binding protein was equally increased in response to exercise and/or recovery in both groups. CONCLUSION: Acute exercise elicits a pronounced and overall similar increase in expression of key transcription factors and activation of key upstream kinases involved in muscle metabolic adaptation to exercise in patients with T2D and weight-matched controls.


Asunto(s)
Adaptación Fisiológica/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Ejercicio Físico/fisiología , Músculo Esquelético/metabolismo , Factores de Transcripción/genética , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica/genética , Humanos , Resistencia a la Insulina/genética , Masculino , Persona de Mediana Edad , Factores de Transcripción/metabolismo
13.
J Clin Endocrinol Metab ; 104(10): 4804-4814, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30933285

RESUMEN

CONTEXT: Microvesicles (MVs) are a class of membrane particles shed by any cell in the body in physiological and pathological conditions. They are considered to be key players in intercellular communication, and with a molecular content reflecting the composition of the cell of origin, they have recently emerged as a promising source of biomarkers in a number of diseases. OBJECTIVE: The effects of acute exercise on the plasma concentration of skeletal muscle-derived MVs (SkMVs) carrying metabolically important membrane proteins were examined. PARTICIPANTS: Thirteen men with obesity and type 2 diabetes mellitus (T2DM) and 14 healthy male controls with obesity exercised on a cycle ergometer for 60 minutes. INTERVENTIONS: Muscle biopsies and blood samples-obtained before exercise, immediately after exercise, and 3 hours into recovery-were collected for the analysis of long-chain fatty acid (LCFA) transport proteins CD36 (a scavenger receptor class B protein) and fatty acid transport protein 4 (FATP4) mRNA content in muscle and for flow cytometric studies on circulating SkMVs carrying either LCFA transport protein. RESULTS: Besides establishing a flow cytometric approach for the detection of circulating SkMVs and subpopulations carrying either CD36 or FATP4 and thereby adding proof to their existence, we demonstrated an overall exercise-induced change of SkMVs carrying these LCFA transport proteins. A positive correlation between exercise-induced changes in skeletal muscle CD36 mRNA expression and concentrations of SkMVs carrying CD36 was found in T2DM only. CONCLUSIONS: This approach could add important real-time information about the abundance of LCFA transport proteins present on activated muscle cells in subjects with impaired glucose metabolism.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Ejercicio Físico/fisiología , Proteínas de Transporte de Ácidos Grasos/metabolismo , Músculo Esquelético/metabolismo , Biopsia , Antígenos CD36/genética , Antígenos CD36/metabolismo , Estudios de Casos y Controles , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Transporte de Ácidos Grasos/genética , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , ARN Mensajero/metabolismo
14.
Physiol Rep ; 6(12): e13723, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29924476

RESUMEN

Regular exercise plays an important role in the prevention and treatment of type 2 diabetes (T2D). The synthesis and secretion of myokines in response to contraction may contribute to the beneficial metabolic effects of exercise. However, some exercise-induced responses may be attenuated in T2D. Here, we investigated whether the effect of acute exercise on selected myokines are impaired in T2D. Skeletal muscle biopsies and blood samples were obtained from 13 men with T2D and 14 weight-matched, glucose-tolerant men before, immediately after and 3-h after acute exercise (60 min cycling) to examine muscle expression and plasma/serum levels of selected myokines. One-hour of exercise increased muscle expression of IL6, FGF21, ANGPTL4, CHI3L1, CTGF and CYR61, of which FGF21, ANGPTL4 and CHI3L1 increased further 3-h into recovery, whereas expression of IL6, CYR61, and CTGF returned to baseline levels. There was no immediate effect of exercise on IL15 expression, but it decreased 3-h into recovery. Plasma IL-6 increased robustly, whereas circulating levels of FGF21, ANGPTL4, IL-15, and CHI3L1 increased only modestly in response to exercise. All returned toward baseline levels 3-h into recovery except for plasma ANGPTL4, which increased further. No significant differences in these responses to exercise were observed between the groups. Our results demonstrate that muscle expression and circulating levels of selected known and putative myokines were equally regulated by acute exercise in patients with T2D and weight-matched controls. This suggests that the potential beneficial metabolic effects of these myokines are not impaired in patients with T2D.


Asunto(s)
Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ejercicio Físico/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteína 4 Similar a la Angiopoyetina/genética , Proteína 4 Similar a la Angiopoyetina/metabolismo , Estudios de Casos y Controles , Proteína 1 Similar a Quitinasa-3/genética , Proteína 1 Similar a Quitinasa-3/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/metabolismo , Citocinas/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Interleucina-15/genética , Interleucina-15/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética
15.
Mol Genet Metab ; 119(3): 258-269, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27595546

RESUMEN

Fabry disease is an X-linked recessive inborn disorder of the glycosphingolipid metabolism, caused by total or partial deficiency of the lysosomal α-galactosidase A enzyme due to mutations in the GLA gene. The prevalent c.639+919 G>A mutation in GLA leads to pathogenic insertion of a 57bp pseudoexon sequence from intron 4, which is responsible for the cardiac variant phenotype. In this study we investigate the splicing regulatory mechanism leading to GLA pseudoexon activation. Splicing analysis of GLA minigenes revealed that pseudoexon activation is influenced by cell-type. We demonstrate that the wild-type sequence harbors an hnRNP A1 and hnRNP A2/B1-binding exonic splicing silencer (ESS) overlapping the 5'splice site (5'ss) that prevents pseudoexon inclusion. The c.639+919 G>A mutation disrupts this ESS allowing U1 snRNP recognition of the 5'ss. We show that the wild-type GLA 5'ss motif with the ESS is also able to inhibit inclusion of an unrelated pseudoexon in the FGB gene, and that also in the FGB context inactivation of the ESS by the c.639+919 G>A mutation causes pseudoexon activation, underscoring the universal nature of the ESS. Finally, we demonstrate that splice switching oligonucleotide (SSO) mediated blocking of the pseudoexon 3'ss and 5'ss effectively restores normal GLA splicing. This indicates that SSO based splicing correction may be a therapeutic alternative in the treatment of Fabry disease.


Asunto(s)
Enfermedad de Fabry/genética , Ribonucleoproteína Nuclear Heterogénea A1/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , alfa-Galactosidasa/genética , Exones/genética , Enfermedad de Fabry/patología , Células HeLa , Humanos , Intrones , Mutación , Sitios de Empalme de ARN , Empalme del ARN/genética , ARN Mensajero/genética , Elementos Silenciadores Transcripcionales/genética
16.
Diabetes ; 65(5): 1219-30, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26822091

RESUMEN

Current evidence on exercise-mediated AMPK regulation in skeletal muscle of patients with type 2 diabetes (T2D) is inconclusive. This may relate to inadequate segregation of trimeric complexes in the investigation of AMPK activity. We examined the regulation of AMPK and downstream targets ACC-ß, TBC1D1, and TBC1D4 in muscle biopsy specimens obtained from 13 overweight/obese patients with T2D and 14 weight-matched male control subjects before, immediately after, and 3 h after exercise. Exercise increased AMPK α2ß2γ3 activity and phosphorylation of ACCß Ser(221), TBC1D1 Ser(237)/Thr(596), and TBC1D4 Ser(704) Conversely, exercise decreased AMPK α1ß2γ1 activity and TBC1D4 Ser(318)/Thr(642) phosphorylation. Interestingly, compared with preexercise, 3 h into exercise recovery, AMPK α2ß2γ1 and α1ß2γ1 activity were increased concomitant with increased TBC1D4 Ser(318)/Ser(341)/Ser(704) phosphorylation. No differences in these responses were observed between patients with T2D and control subjects. Subjects were also studied by euglycemic-hyperinsulinemic clamps performed at rest and 3 h after exercise. We found no evidence for insulin to regulate AMPK activity. Thus, AMPK signaling is not compromised in muscle of patients with T2D during exercise and insulin stimulation. Our results reveal a hitherto unrecognized activation of specific AMPK complexes in exercise recovery. We hypothesize that the differential regulation of AMPK complexes plays an important role for muscle metabolism and adaptations to exercise.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ejercicio Físico , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Acetil-CoA Carboxilasa/metabolismo , Ciclismo , Biopsia , Índice de Masa Corporal , Estudios de Cohortes , Terapia Combinada , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Fatiga/etiología , Fatiga/prevención & control , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Obesidad/complicaciones , Obesidad/terapia , Sobrepeso/complicaciones , Sobrepeso/terapia , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Músculo Cuádriceps
17.
Nucleic Acids Res ; 43(9): 4627-39, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25878036

RESUMEN

The prevalent c.903+469T>C mutation in MTRR causes the cblE type of homocystinuria by strengthening an SRSF1 binding site in an ESE leading to activation of a pseudoexon. We hypothesized that other splicing regulatory elements (SREs) are also critical for MTRR pseudoexon inclusion. We demonstrate that the MTRR pseudoexon is on the verge of being recognized and is therefore vulnerable to several point mutations that disrupt a fine-tuned balance between the different SREs. Normally, pseudoexon inclusion is suppressed by a hnRNP A1 binding exonic splicing silencer (ESS). When the c.903+469T>C mutation is present two ESEs abrogate the activity of the ESS and promote pseudoexon inclusion. Blocking the 3'splice site or the ESEs by SSOs is effective in restoring normal splicing of minigenes and endogenous MTRR transcripts in patient cells. By employing an SSO complementary to both ESEs, we were able to rescue MTRR enzymatic activity in patient cells to approximately 50% of that in controls. We show that several point mutations, individually, can activate a pseudoexon, illustrating that this mechanism can occur more frequently than previously expected. Moreover, we demonstrate that SSO blocking of critical ESEs is a promising strategy to treat the increasing number of activated pseudoexons.


Asunto(s)
Anemia Megaloblástica/genética , Exones , Ferredoxina-NADP Reductasa/genética , Homocistinuria/genética , Mutación , Oligonucleótidos , Empalme del ARN , Secuencias Reguladoras de Ácido Ribonucleico , Anemia Megaloblástica/enzimología , Línea Celular , Células Cultivadas , Ferredoxina-NADP Reductasa/metabolismo , Células HEK293 , Homocistinuria/enzimología , Humanos , Sitios de Empalme de ARN
18.
Hum Mutat ; 35(1): 86-95, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24123825

RESUMEN

Multiple acyl-CoA dehydrogenation deficiency is a disorder of fatty acid and amino acid oxidation caused by defects of electron transfer flavoprotein (ETF) or its dehydrogenase (ETFDH). A clear relationship between genotype and phenotype makes genotyping of patients important not only diagnostically but also for prognosis and for assessment of treatment. In the present study, we show that a predicted benign ETFDH missense variation (c.158A>G/p.Lys53Arg) in exon 2 causes exon skipping and degradation of ETFDH protein in patient samples. Using splicing reporter minigenes and RNA pull-down of nuclear proteins, we show that the c.158A>G variation increases the strength of a preexisting exonic splicing silencer (ESS) motif UAGGGA. This ESS motif binds splice inhibitory hnRNP A1, hnRNP A2/B1, and hnRNP H proteins. Binding of these inhibitory proteins prevents binding of the positive splicing regulatory SRSF1 and SRSF5 proteins to nearby and overlapping exonic splicing enhancer elements and this causes exon skipping. We further suggest that binding of hnRNP proteins to UAGGGA is increased by triggering synergistic hnRNP H binding to GGG triplets located upstream and downsteam of the UAGGGA motif. A number of disease-causing exonic elements that induce exon skipping in other genes have a similar architecture as the one in ETFDH exon 2.


Asunto(s)
Adenosina/metabolismo , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Guanina/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Empalme del ARN , Secuencias de Aminoácidos , Cadáver , Elementos de Facilitación Genéticos , Exones , Regulación de la Expresión Génica , Variación Genética , Células HEK293 , Células HeLa , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Humanos , Recién Nacido , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/diagnóstico , Mutación Missense , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ADN , Factores de Empalme Serina-Arginina , Elementos Silenciadores Transcripcionales , Repeticiones de Trinucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...