Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Alzheimers Dement ; 19(11): 4872-4885, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37037474

RESUMEN

INTRODUCTION: Sporadic Alzheimer's disease (sAD) is the leading type of dementia. Brain glucose hypometabolism, along with decreased O-GlcNAcylation levels, occurs before the onset of symptoms and correlates with pathogenesis. Heretofore, the mechanisms involved and the roles of O-GlcNAcylation in sAD pathology largely remain unknown due to a lack of human models of sAD. METHODS: Human cortical neurons were generated from pluripotent stem cells (PSCs) and treated with glucose reduction media. RESULTS: We found a narrow window of glucose concentration that induces sAD-like phenotypes in PSC-derived neurons. With our model, we reveal that dysregulated O-GlcNAc, in part through mitochondrial dysfunction, causes the onset of sAD-like changes. We demonstrate the therapeutic potential of inhibiting O-GlcNAcase in alleviating AD-like biochemical changes. DISCUSSION: Our results suggest that dysregulated O-GlcNAc might be a direct molecular link between hypometabolism and sAD-like alternations. Moreover, this model can be exploited to explore molecular processes and for drug development. HIGHLIGHTS: Lowering glucose to a critical level causes AD-like changes in cortical neurons. Defective neuronal structure and function were also recapitulated in current model. Dysregulated O-GlcNAcylation links impaired glucose metabolism to AD-like changes. Mitochondrial abnormalities correlate with O-GlcNAcylation and precede AD-like phenotype. Our model provides a platform to study sAD as a metabolic disease in human neurons.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Humanos , Enfermedad de Alzheimer/patología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Glucosa/metabolismo , Acetilglucosamina/metabolismo
2.
Am J Physiol Cell Physiol ; 318(2): C242-C252, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31721614

RESUMEN

The objective of this study was to interrogate the link between mitochondrial dysfunction and mitochondrial-specific autophagy in skeletal muscle. C57BL/6J mice were used to establish a time course of mitochondrial function and autophagy induction after fatigue (n = 12), eccentric contraction-induced injury (n = 20), or traumatic freeze injury (FI, n = 28); only FI resulted in a combination of mitochondrial dysfunction, i.e., decreased mitochondrial respiration, and autophagy induction. Moving forward, we tested the hypothesis that mitochondrial-specific autophagy is important for the timely recovery of mitochondrial function after FI. Following FI, there is a significant increase in several mitochondrial-specific autophagy-related protein contents including dynamin-related protein 1 (Drp1), BCL1 interacting protein (BNIP3), Pink1, and Parkin (~2-fold, P < 0.02). Also, mitochondrial-enriched fractions from FI muscles showed microtubule-associated protein light chain B1 (LC3)II colocalization suggesting autophagosome assembly around the damaged mitochondrial. Unc-51 like autophagy activating kinase (Ulk1) is considered necessary for mitochondrial-specific autophagy and herein we utilized a mouse model with Ulk1 deficiency in adult skeletal muscle (myogenin-Cre). While Ulk1 knockouts had contractile weakness compared with littermate controls (-27%, P < 0.02), the recovery of mitochondrial function was not different, and this may be due in part to a partial rescue of Ulk1 protein content within the regenerating muscle tissue of knockouts from differentiated satellite cells in which Ulk1 was not genetically altered via myogenin-Cre. Lastly, autophagy flux was significantly less in injured versus uninjured muscles (-26%, P < 0.02) despite the increase in autophagy-related protein content. This suggests autophagy flux is not upregulated to match increases in autophagy machinery after injury and represents a potential bottleneck in the clearance of damaged mitochondria by autophagy.


Asunto(s)
Autofagia/fisiología , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Heridas y Lesiones/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Diferenciación Celular/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo
3.
FASEB J ; 33(8): 8822-8835, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31059287

RESUMEN

Brown adipose tissue (BAT) thermogenesis increases energy expenditure (EE). Expanding the volume of active BAT via transplantation holds promise as a therapeutic strategy for morbid obesity and diabetes. Brown adipose progenitor cells (BAPCs) can be isolated and expanded to generate autologous brown adipocyte implants. However, the transplantation of brown adipocytes is currently impeded by poor efficiency of BAT tissue formation in vivo and undesirably short engraftment time. In this study, we demonstrated that transplanting BAPCs into limb skeletal muscles consistently led to the ectopic formation of uncoupling protein 1 (UCP1)+pos adipose tissue with long-term engraftment (>4 mo). Combining VEGF with the BAPC transplant further improved BAT formation in muscle. Ectopic engraftment of BAPC-derived BAT in skeletal muscle augmented the EE of recipient mice. Although UCP1 expression declined in long-term BAT grafts, this deterioration can be reversed by swimming exercise because of sympathetic activation. This study suggests that intramuscular transplantation of BAPCs represents a promising approach to deriving functional BAT engraftment, which may be applied to therapeutic BAT transplantation and tissue engineering.-Liu, Y., Fu, W., Seese, K., Yin, A., Yin, H. Ectopic brown adipose tissue formation within skeletal muscle after brown adipose progenitor cell transplant augments energy expenditure.


Asunto(s)
Tejido Adiposo Pardo , Coristoma/metabolismo , Metabolismo Energético , Músculo Esquelético/metabolismo , Animales , Células Cultivadas , Coristoma/etiología , Femenino , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Ratones , Ratones Endogámicos C57BL , Esfuerzo Físico , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
4.
Prostate ; 79(8): 896-908, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30900312

RESUMEN

Numerous genetic alterations have been identified during prostate cancer progression. The influence of environmental factors, particularly the diet, on the acceleration of tumor progression is largely unknown. Expression levels and/or activity of Src kinase are highly elevated in numerous cancers including advanced stages of prostate cancer. In this study, we demonstrate that high-fat diets (HFDs) promoted pathological transformation mediated by the synergy of Src and androgen receptor in vivo. Additionally, a diet high in saturated fat significantly enhanced proliferation of Src-mediated xenograft tumors in comparison with a diet high in unsaturated fat. The saturated fatty acid palmitate, a major constituent in a HFD, significantly upregulated the biosynthesis of palmitoyl-CoA in cancer cells in vitro and in xenograft tumors in vivo. The exogenous palmitate enhanced Src-dependent mitochondrial ß-oxidation. Additionally, it elevated the amount of C16-ceramide and total saturated ceramides, increased the level of Src kinase localized in the cell membrane, and Src-mediated downstream signaling, such as the activation of mitogen-activated protein kinase and focal adhesion kinase. Our results uncover how the metabolism of dietary palmitate cooperates with elevated Src kinase in the acceleration of prostate tumor progression.


Asunto(s)
Palmitatos/administración & dosificación , Neoplasias de la Próstata/etiología , Familia-src Quinasas/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Dieta Alta en Grasa/efectos adversos , Progresión de la Enfermedad , Células HEK293 , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Células PC-3 , Palmitatos/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
5.
FASEB J ; 32(9): 4836-4847, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29579398

RESUMEN

Our previous studies have shown that response gene to complement (RGC)-32 deficiency (Rgc32-/-) protects mice from diet-induced obesity and increases thermogenic gene expression in adipose tissues. However, the underlying mechanisms by which RGC-32 regulates thermogenic gene expression remain to be determined. In the present study, RGC-32 expression in white adipose tissue (WAT) was suppressed during cold exposure-induced WAT browning. Rgc32-/- significantly increased thermogenic gene expression in the differentiated stromal vascular fraction (SVF) of inguinal (i)WAT and interscapular brown adipose tissue (BAT). Rgc32-/- and cold exposure regulated a common set of genes in iWAT, as shown by RNA sequencing data. Pathway enrichment analyses showed that Rgc32-/- down-regulated PI3K/Akt signaling-related genes. Akt phosphorylation was also consistently decreased in Rgc32-/- iWAT, which led to an increase in ß3-adrenergic receptor (ß3-AR) expression and subsequent activation of mammalian target of rapamycin complex (mTORC)-1. ß3-AR antagonist SR 59230A and mTORC1 inhibitor rapamycin blocked Rgc32-/--induced thermogenic gene expression in both iWAT and interscapular BAT. These results indicate that RGC-32 suppresses adipose tissue thermogenic gene expression through down-regulation of ß3-AR expression and mTORC1 activity via a PI3K/Akt-dependent mechanism.-Chen, S., Mei, X., Yin, A., Yin, H., Cui, X.-B., Chen, S.-Y. Response gene to complement 32 suppresses adipose tissue thermogenic genes through inhibiting ß3-adrenergic receptor/mTORC1 signaling.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Nucleares/deficiencia , Receptores Adrenérgicos beta 3/metabolismo , Termogénesis/genética , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular/genética , Proteínas del Sistema Complemento/metabolismo , Ratones Noqueados , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética
6.
J Lipid Res ; 58(9): 1777-1784, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28716822

RESUMEN

Obesity is the major risk factor for type 2 diabetes, cardiovascular disorders, and many other diseases. Adipose tissue inflammation is frequently associated with obesity and contributes to the morbidity and mortality. Dedicator of cytokinesis 2 (DOCK2) is involved in several inflammatory diseases, but its role in obesity remains unknown. To explore the function of DOCK2 in obesity and insulin resistance, WT and DOCK2-deficient (DOCK2-/-) mice were given chow or high-fat diet (HFD) for 12 weeks followed by metabolic, biochemical, and histologic analyses. DOCK2 was robustly induced in adipose tissues of WT mice given HFD. DOCK2-/- mice with HFD showed decreased body weight gain and improved metabolic homeostasis and insulin resistance compared with WT mice. DOCK2 deficiency also attenuated adipose tissue and systemic inflammation accompanied by reduced macrophage infiltration. Moreover, DOCK2-/- mice exhibited increased expression of metabolic genes in adipose tissues with greater energy expenditure. Mechanistically, DOCK2 appeared to regulate brown adipocyte differentiation because increased preadipocyte differentiation to brown adipocytes in interscapular and inguinal fat was observed in DOCK2-/- mice, as compared with WT. These data indicated that DOCK2 deficiency protects mice from HFD-induced obesity, at least in part, by stimulating brown adipocyte differentiation. Therefore, targeting DOCK2 may be a potential therapeutic strategy for treating obesity-associated diseases.


Asunto(s)
Tejido Adiposo/patología , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Obesidad/genética , Obesidad/patología , Adipocitos/patología , Tejido Adiposo/efectos de los fármacos , Animales , Diferenciación Celular/genética , Línea Celular , Metabolismo Energético/efectos de los fármacos , Técnicas de Inactivación de Genes , Factores de Intercambio de Guanina Nucleótido , Homeostasis/genética , Inflamación/patología , Resistencia a la Insulina/genética , Ratones Endogámicos C57BL , Obesidad/inducido químicamente , Obesidad/metabolismo
7.
J Cachexia Sarcopenia Muscle ; 15(2): 631-645, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38333911

RESUMEN

BACKGROUND: Chronic hypoxia and skeletal muscle atrophy commonly coexist in patients with COPD and CHF, yet the underlying physio-pathological mechanisms remain elusive. Muscle regeneration, driven by muscle stem cells (MuSCs), holds therapeutic potential for mitigating muscle atrophy. This study endeavours to investigate the influence of chronic hypoxia on muscle regeneration, unravel key molecular mechanisms, and explore potential therapeutic interventions. METHODS: Experimental mice were exposed to prolonged normobaric hypoxic air (15% pO2, 1 atm, 2 weeks) to establish a chronic hypoxia model. The impact of chronic hypoxia on body composition, muscle mass, muscle strength, and the expression levels of hypoxia-inducible factors HIF-1α and HIF-2α in MuSC was examined. The influence of chronic hypoxia on muscle regeneration, MuSC proliferation, and the recovery of muscle mass and strength following cardiotoxin-induced injury were assessed. The muscle regeneration capacities under chronic hypoxia were compared between wildtype mice, MuSC-specific HIF-2α knockout mice, and mice treated with HIF-2α inhibitor PT2385, and angiotensin converting enzyme (ACE) inhibitor lisinopril. Transcriptomic analysis was performed to identify hypoxia- and HIF-2α-dependent molecular mechanisms. Statistical significance was determined using analysis of variance (ANOVA) and Mann-Whitney U tests. RESULTS: Chronic hypoxia led to limb muscle atrophy (EDL: 17.7%, P < 0.001; Soleus: 11.5% reduction in weight, P < 0.001) and weakness (10.0% reduction in peak-isometric torque, P < 0.001), along with impaired muscle regeneration characterized by diminished myofibre cross-sectional areas, increased fibrosis (P < 0.001), and incomplete strength recovery (92.3% of pre-injury levels, P < 0.05). HIF-2α stabilization in MuSC under chronic hypoxia hindered MuSC proliferation (26.1% reduction of MuSC at 10 dpi, P < 0.01). HIF-2α ablation in MuSC mitigated the adverse effects of chronic hypoxia on muscle regeneration and MuSC proliferation (30.9% increase in MuSC numbers at 10 dpi, P < 0.01), while HIF-1α ablation did not have the same effect. HIF-2α stabilization under chronic hypoxia led to elevated local ACE, a novel direct target of HIF-2α. Notably, pharmacological interventions with PT2385 or lisinopril enhanced muscle regeneration under chronic hypoxia (PT2385: 81.3% increase, P < 0.001; lisinopril: 34.6% increase in MuSC numbers at 10 dpi, P < 0.05), suggesting their therapeutic potential for alleviating chronic hypoxia-associated muscle atrophy. CONCLUSIONS: Chronic hypoxia detrimentally affects skeletal muscle regeneration by stabilizing HIF-2α in MuSC and thereby diminishing MuSC proliferation. HIF-2α increases local ACE levels in skeletal muscle, contributing to hypoxia-induced regenerative deficits. Administration of HIF-2α or ACE inhibitors may prove beneficial to ameliorate chronic hypoxia-associated muscle atrophy and weakness by improving muscle regeneration under chronic hypoxia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Indanos , Lisinopril , Sulfonas , Animales , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia , Músculo Esquelético/metabolismo , Atrofia Muscular/etiología
8.
Oncogene ; 40(10): 1806-1820, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33564069

RESUMEN

Fatty acid metabolism is essential for the biogenesis of cellular components and ATP production to sustain proliferation of cancer cells. Long-chain fatty acyl-CoA synthetases (ACSLs), a group of rate-limiting enzymes in fatty acid metabolism, catalyze the bioconversion of exogenous or de novo synthesized fatty acids to their corresponding fatty acyl-CoAs. In this study, systematical analysis of ACSLs levels and the amount of fatty acyl-CoAs illustrated that ACSL1 were significantly associated with the levels of a broad spectrum of fatty acyl-CoAs, and were elevated in human prostate tumors. ACSL1 increased the biosynthesis of fatty acyl-CoAs including C16:0-, C18:0-, C18:1-, and C18:2-CoA, triglycerides and lipid accumulation in cancer cells. Mechanistically, ACSL1 modulated mitochondrial respiration, ß-oxidation, and ATP production through regulation of CPT1 activity. Knockdown of ACSL1 inhibited the cell cycle, and suppressed the proliferation and migration of prostate cancer cells in vitro, and growth of prostate xenograft tumors in vivo. Our study implicates ACSL1 as playing an important role in prostate tumor progression, and provides a therapeutic strategy of targeting fatty acid metabolism for the treatment of prostate cancer.


Asunto(s)
Coenzima A Ligasas/genética , Ácidos Grasos/metabolismo , Lipogénesis/genética , Neoplasias de la Próstata/genética , Adenosina Trifosfato/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Ácidos Grasos/genética , Xenoinjertos , Humanos , Masculino , Ratones , Oxidación-Reducción , Neoplasias de la Próstata/patología
9.
Nat Commun ; 11(1): 2758, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488069

RESUMEN

Human beige adipocytes (BAs) have potential utility for the development of therapeutics to treat diabetes and obesity-associated diseases. Although several reports have described the generation of beige adipocytes in vitro, their potential utility in cell therapy and drug discovery has not been reported. Here, we describe the generation of BAs from human adipose-derived stem/stromal cells (ADSCs) in serum-free medium with efficiencies >90%. Molecular profiling of beige adipocytes shows them to be similar to primary BAs isolated from human tissue. In vitro, beige adipocytes exhibit uncoupled mitochondrial respiration and cAMP-induced lipolytic activity. Following transplantation, BAs increase whole-body energy expenditure and oxygen consumption, while reducing body-weight in recipient mice. Finally, we show the therapeutic utility of BAs in a platform for high-throughput drug screening (HTS). These findings demonstrate the potential utility of BAs as a cell therapeutic and as a tool for the identification of drugs to treat metabolic diseases.


Asunto(s)
Adipocitos Beige/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Descubrimiento de Drogas/métodos , Enfermedades Metabólicas/metabolismo , Adipocitos Beige/citología , Animales , Peso Corporal , Evaluación Preclínica de Medicamentos , Metabolismo Energético , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Células Madre Mesenquimatosas , Enfermedades Metabólicas/tratamiento farmacológico , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/metabolismo , Consumo de Oxígeno , Células del Estroma , Trasplante
10.
Cell Stem Cell ; 27(5): 784-797.e11, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-32783886

RESUMEN

Brown adipocytes (BAs) are a potential cell source for the treatment of metabolic disease, including type 2 diabetes. In this report, human pluripotent stem cells (hPSCs) are subject to directed differentiation through a paraxial mesoderm progenitor state that generates BAs at high efficiency. Molecular analysis identifies potential regulatory networks for BA development, giving insight into development along this lineage. hPSC-derived BAs undergo elevated rates of glycolysis, uncoupled respiration, and lipolysis that are responsive to changes in cyclic AMP (cAMP)-dependent signaling, consistent with metabolic activity in BA tissue depots. Transplanted human BAs engraft into the inter-scapular region of recipient mice and exhibit thermogenic activity. Recipient animals have elevated metabolic activity, respiratory exchange ratios, and whole-body energy expenditure. Finally, transplanted BAs reduce circulating glucose levels in hyperglycemic animals. These data provide a roadmap for brown adipocyte development and indicate that BAs generated from hPSCs have potential as a tool for therapeutic development.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Madre Pluripotentes , Adipocitos Marrones , Animales , Diferenciación Celular , Humanos , Mesodermo , Ratones , Termogénesis
11.
Sci Rep ; 9(1): 4079, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30858541

RESUMEN

Volumetric muscle loss (VML) injury is characterized by a non-recoverable loss of muscle fibers due to ablative surgery or severe orthopaedic trauma, that results in chronic functional impairments of the soft tissue. Currently, the effects of VML on the oxidative capacity and adaptability of the remaining injured muscle are unclear. A better understanding of this pathophysiology could significantly shape how VML-injured patients and clinicians approach regenerative medicine and rehabilitation following injury. Herein, the data indicated that VML-injured muscle has diminished mitochondrial content and function (i.e., oxidative capacity), loss of mitochondrial network organization, and attenuated oxidative adaptations to exercise. However, forced PGC-1α over-expression rescued the deficits in oxidative capacity and muscle strength. This implicates physiological activation of PGC1-α as a limiting factor in VML-injured muscle's adaptive capacity to exercise and provides a mechanistic target for regenerative rehabilitation approaches to address the skeletal muscle dysfunction.


Asunto(s)
Músculo Esquelético/lesiones , Enfermedades Musculares/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Medicina Regenerativa , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/fisiopatología , Estrés Oxidativo/genética , Regeneración/genética
12.
Adv Mater ; 31(46): e1904058, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31553099

RESUMEN

Many inorganic nanoparticles are prepared and their behaviors in living systems are investigated. Yet, common electrolytes such as NaCl are left out of this campaign. The underlying assumption is that electrolyte nanoparticles will quickly dissolve in water and behave similarly as their constituent salts. Herein, this preconception is challenged. The study shows that NaCl nanoparticles (SCNPs) but not salts are highly toxic to cancer cells. This is because SCNPs enter cells through endocytosis, bypassing cell regulations on ion transport. When dissolved inside cancer cells, SCNPs cause a surge of osmolarity and rapid cell lysis. Interestingly, normal cells are much more resistant to the treatment due to their relatively low sodium levels. Unlike conventional chemotherapeutics, SCNPs cause immunogenic cell death or ICD. In vivo studies show that SCNPs not only kill cancer cells, but also boost an anticancer immunity. The discovery opens up a new perspective on nanoparticle-based therapeutics.


Asunto(s)
Nanopartículas , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Cloruro de Sodio/química , Cloruro de Sodio/uso terapéutico , Nanomedicina Teranóstica/métodos , Línea Celular Tumoral , Endocitosis , Humanos , Neoplasias/inmunología , Neoplasias/patología , Cloruro de Sodio/metabolismo
13.
Magn Reson Imaging ; 52: 84-93, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29928937

RESUMEN

PURPOSE: To explore, at a high field strength of 7T, the performance of various fat spectral models on the quantification of triglyceride composition and proton density fat fraction (PDFF) using chemical-shift encoded MRI (CSE-MRI). METHODS: MR data was acquired from CSE-MRI experiments for various fatty materials, including oil and butter samples and in vivo brown and white adipose mouse tissues. Triglyceride composition and PDFF were estimated using various a priori 6- or 9-peak fat spectral models. To serve as references, NMR spectroscopy experiments were conducted to obtain material specific fat spectral models and triglyceride composition estimates for the same fatty materials. Results obtained using the spectroscopy derived material specific models were compared to results obtained using various published fat spectral models. RESULTS: Using a 6-peak fat spectral model to quantify triglyceride composition may lead to large biases at high field strengths. When using a 9-peak model, triglyceride composition estimations vary greatly depending on the relative amplitudes of the chosen a priori spectral model, while PDFF estimations show small variations across spectral models. Material specific spectroscopy derived spectral models produce estimations that better correlate with NMR spectroscopy estimations in comparison to those obtained using non-material specific models. CONCLUSION: At a high field strength of 7T, a material specific 9-peak fat spectral model, opposed to a widely accepted or generic human liver model, is necessary to accurately quantify triglyceride composition when using CSE-MRI estimation methods that assume the spectral model to be known as a priori information. CSE-MRI allows for the quantification of the spatial distribution of triglyceride composition for certain in vivo applications. Additionally, PDFF quantification is shown to be independent of the chosen a priori spectral model, which agrees with previously reported results obtained at lower field strengths (e.g. 3T).


Asunto(s)
Tejido Adiposo/diagnóstico por imagen , Tejido Adiposo/metabolismo , Hígado/diagnóstico por imagen , Hígado/metabolismo , Imagen por Resonancia Magnética/métodos , Triglicéridos/análisis , Animales , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fantasmas de Imagen , Reproducibilidad de los Resultados , Triglicéridos/metabolismo
14.
J Clin Invest ; 128(6): 2339-2355, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29533927

RESUMEN

The remarkable regeneration capability of skeletal muscle depends on the coordinated proliferation and differentiation of satellite cells (SCs). The self-renewal of SCs is critical for long-term maintenance of muscle regeneration potential. Hypoxia profoundly affects the proliferation, differentiation, and self-renewal of cultured myoblasts. However, the physiological relevance of hypoxia and hypoxia signaling in SCs in vivo remains largely unknown. Here, we demonstrate that SCs are in an intrinsic hypoxic state in vivo and express hypoxia-inducible factor 2A (HIF2A). HIF2A promotes the stemness and long-term homeostatic maintenance of SCs by maintaining their quiescence, increasing their self-renewal, and blocking their myogenic differentiation. HIF2A stabilization in SCs cultured under normoxia augments their engraftment potential in regenerative muscle. Conversely, HIF2A ablation leads to the depletion of SCs and their consequent regenerative failure in the long-term. In contrast, transient pharmacological inhibition of HIF2A accelerates muscle regeneration by increasing SC proliferation and differentiation. Mechanistically, HIF2A induces the quiescence and self-renewal of SCs by binding the promoter of the Spry1 gene and activating Spry1 expression. These findings suggest that HIF2A is a pivotal mediator of hypoxia signaling in SCs and may be therapeutically targeted to improve muscle regeneration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Hipoxia de la Célula , Ratones , Ratones Transgénicos , Desarrollo de Músculos , Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/citología
15.
Cell Stem Cell ; 21(4): 502-516.e9, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28965765

RESUMEN

As human pluripotent stem cells (hPSCs) exit pluripotency, they are thought to switch from a glycolytic mode of energy generation to one more dependent on oxidative phosphorylation. Here we show that, although metabolic switching occurs during early mesoderm and endoderm differentiation, high glycolytic flux is maintained and, in fact, essential during early ectoderm specification. The elevated glycolysis observed in hPSCs requires elevated MYC/MYCN activity. Metabolic switching during endodermal and mesodermal differentiation coincides with a reduction in MYC/MYCN and can be reversed by ectopically restoring MYC activity. During early ectodermal differentiation, sustained MYCN activity maintains the transcription of "switch" genes that are rate-limiting for metabolic activity and lineage commitment. Our work, therefore, shows that metabolic switching is lineage-specific and not a required step for exit of pluripotency in hPSCs and identifies MYC and MYCN as developmental regulators that couple metabolism to pluripotency and cell fate determination.


Asunto(s)
Linaje de la Célula , Análisis de Flujos Metabólicos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ciclo Celular , Diferenciación Celular , Estratos Germinativos/citología , Glucólisis , Humanos , Espectroscopía de Resonancia Magnética , Modelos Biológicos , Proteína Proto-Oncogénica N-Myc/metabolismo
16.
PLoS One ; 10(9): e0138344, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26390217

RESUMEN

Beta-adrenergic activation stimulates uncoupling protein 1 (UCP1), enhancing metabolic rate. In vitro, most work has studied brown adipocytes, however, few have investigated more established adipocyte lines such as the murine 3T3-L1 line. To assess the effect of beta-adrenergic activation, mature 3T3-L1s were treated for 6 or 48 hours with or without isoproterenol (10 and 100 µM) following standard differentiation supplemented with thyroid hormone (T3; 1 nM). The highest dose of isoproterenol increased lipid content following 48 hours of treatment. This concentration enhanced UCP1 mRNA and protein expression. The increase in UCP1 following 48 hours of isoproterenol increased oxygen consumption rate. Further, coupling efficiency of the electron transport chain was disturbed and an enhancement of glycolytic rate was measured alongside this, indicating an attempt to meet the energy demands of the cell. Lastly, markers of beige adipocytes (protein content of CD137 and gene transcript of CITED1) were also found to be upregulated at 48 hours of isoproterenol treatment. This data indicates that mature 3T3-L1 adipocytes are responsive to isoproterenol and induce UCP1 expression and activity. Further, this finding provides a model for further pharmaceutical and nutraceutical investigation of UCP1 in 3T3-L1s.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Agonistas Adrenérgicos beta/farmacología , Glucólisis/efectos de los fármacos , Canales Iónicos/genética , Isoproterenol/farmacología , Proteínas Mitocondriales/genética , Células 3T3-L1 , Adipocitos/citología , Animales , Ratones , ARN Mensajero/genética , Proteína Desacopladora 1 , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA