Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 365
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Physiol Plant ; 176(2): e14262, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38522857

RESUMEN

Soybean (Glycine max) is economically significant, but the mechanisms underlying its adaptation to simultaneous low phosphorus and salt stresses are unclear. We employed the Shennong 94-1-8 soybean germplasm to conduct a comprehensive analysis, integrating both physiochemical and transcriptomic approaches, to unravel the response mechanisms of soybean when subjected to simultaneous low phosphorus and salt stresses. Remarkably, the combined stress exhibited the most pronounced impact on the soybean root system, which led to a substantial reduction in total soluble sugar (TSS) and total soluble protein (TSP) within the plants under this treatment. A total of 20,953 differentially expressed genes were identified through pairwise comparisons. Heatmap analysis of genes related to energy metabolism pathways demonstrated a significant down-regulation in expression under salt and low phosphorus + salt treatments, while low phosphorus treatment did not exhibit similar expression trends. Furthermore, the weighted gene co-expression network analysis (WGCNA) indicated that the blue module had a strong positive correlation with TSS and TSP. Notably, 2,3-bisphosphoglycerate-dependent phosphoglycerate mutase 1, FCS-Like Zinc finger 8, auxin response factor 18 isoform X2, and NADP-dependent malic enzyme emerged as hub genes associated with energy metabolism. In summary, our findings indicate that soybean roots are more adversely affected by salt and combined stress than by low phosphorus alone due to reduced activity in energy metabolism-related pathways and hub genes. These results offer novel insights into the adaptive mechanisms of soybeans when facing the combined stress of low phosphorus and salinity.


Asunto(s)
Glycine max , Estrés Fisiológico , Glycine max/genética , Estrés Fisiológico/genética , Cloruro de Sodio/farmacología , Cloruro de Sodio/metabolismo , Perfilación de la Expresión Génica , Metabolismo Energético/genética , Fósforo/metabolismo , Regulación de la Expresión Génica de las Plantas
2.
J Neurosci ; 44(14)2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38395612

RESUMEN

ß-Catenin is a bifunctional molecule that is an effector of the wingless-related integration site (Wnt) signaling to control gene expression and contributes to the regulation of cytoskeleton and neurotransmitter vesicle trafficking. In its former role, ß-catenin binds transcription factor 7-like 2 (TCF7L2), which shows strong genetic associations with the pathogenesis of obesity and type-2 diabetes. Here, we sought to determine whether ß-catenin plays a role in the neuroendocrine regulation of body weight and glucose homeostasis. Bilateral injections of adeno-associated virus type-2 (AAV2)-mCherry-Cre were placed into the arcuate nucleus of adult male and female ß-catenin flox mice, to specifically delete ß-catenin expression in the mediobasal hypothalamus (MBH-ß-cat KO). Metabolic parameters were then monitored under conditions of low-fat (LFD) and high-fat diet (HFD). On LFD, MBH-ß-cat KO mice showed minimal metabolic disturbances, but on HFD, despite having only a small difference in weekly caloric intake, the MBH-ß-cat KO mice were significantly heavier than the control mice in both sexes (p < 0.05). This deficit seemed to be due to a failure to show an adaptive increase in energy expenditure seen in controls, which served to offset the increased calories by HFD. Both male and female MBH-ß-cat KO mice were highly glucose intolerant when on HFD and displayed a significant reduction in both leptin and insulin sensitivity compared with controls. This study highlights a critical role for ß-catenin in the hypothalamic circuits regulating body weight and glucose homeostasis and reveals potential mechanisms by which genetic variation in this pathway could impact on development of metabolic disease.


Asunto(s)
Diabetes Mellitus Tipo 2 , Dieta Alta en Grasa , Animales , Femenino , Masculino , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Peso Corporal/genética , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Glucosa/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo
3.
Glia ; 72(2): 433-451, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37870193

RESUMEN

Mitochondria support the energetic demands of the cells. Autophagic turnover of mitochondria serves as a critical pathway for mitochondrial homeostasis. It is unclear how bioenergetics and autophagy are functionally connected. Here, we identify an endolysosomal membrane protein that facilitates autophagy to regulate ATP production in glia. We determined that Drosophila tweety (tty) is highly expressed in glia and localized to endolysosomes. Diminished fusion between autophagosomes and endolysosomes in tty-deficient glia was rescued by expressing the human Tweety Homolog 1 (TTYH1). Loss of tty in glia attenuated mitochondrial turnover, elevated mitochondrial oxidative stress, and impaired locomotor functions. The cellular and organismal defects were partially reversed by antioxidant treatment. We performed live-cell imaging of genetically encoded metabolite sensors to determine the impact of tty and autophagy deficiencies on glial bioenergetics. We found that tty-deficient glia exhibited reduced mitochondrial pyruvate consumption accompanied by a shift toward glycolysis for ATP production. Likewise, genetic inhibition of autophagy in glia resulted in a similar glycolytic shift in bioenergetics. Furthermore, the survival of mutant flies became more sensitive to starvation, underlining the significance of tty in the crosstalk between autophagy and bioenergetics. Together, our findings uncover the role for tty in mitochondrial homeostasis via facilitating autophagy, which determines bioenergetic balance in glia.


Asunto(s)
Autofagia , Drosophila , Metabolismo Energético , Mitocondrias , Animales , Humanos , Adenosina Trifosfato/metabolismo , Autofagia/genética , Drosophila/genética , Drosophila/metabolismo , Metabolismo Energético/genética , Homeostasis , Mitocondrias/metabolismo , Neuroglía/metabolismo
4.
Commun Biol ; 6(1): 857, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37591947

RESUMEN

The body temperature of mice is higher at night than during the day. We show here that global deletion of acid-sensing ion channel 1a (ASIC1a) results in lower body temperature during a part of the night. ASICs are pH sensors that modulate neuronal activity. The deletion of ASIC1a decreased the voluntary activity at night of mice that had access to a running wheel but did not affect their spontaneous activity. Daily rhythms of thyrotropin-releasing hormone mRNA in the hypothalamus and of thyroid-stimulating hormone ß mRNA in the pituitary, and of prolactin mRNA in the hypothalamus and pituitary were suppressed in ASIC1a-/- mice. The serum thyroid hormone levels were however not significantly changed by ASIC1a deletion. Our findings indicate that ASIC1a regulates activity and signaling in the hypothalamus and pituitary. This likely leads to the observed changes in body temperature by affecting the metabolism or energy expenditure.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Temperatura Corporal , Animales , Ratones , Canales Iónicos Sensibles al Ácido/genética , Metabolismo Energético/genética , Hipotálamo , ARN Mensajero
5.
Cell Rep ; 42(5): 112415, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37116488

RESUMEN

Crosstalk among organs/tissues is important for regulating systemic metabolism. Here, we demonstrate inter-organ crosstalk between hepatic insulin and hypothalamic leptin actions, which maintains survival during food shortages. In inducible liver insulin receptor knockout mice, body weight is increased with hyperphagia and decreased energy expenditure, accompanied by increased circulating leptin receptor (LepR) and decreased hypothalamic leptin actions. Additional hepatic LepR deficiency reverses these metabolic phenotypes. Thus, decreased hepatic insulin action suppresses hypothalamic leptin action with increased liver-derived soluble LepR. Human hepatic and circulating LepR levels also correlate negatively with hepatic insulin action indices. In mice, food restriction decreases hepatic insulin action and energy expenditure with increased circulating LepR. Hepatic LepR deficiency increases mortality with enhanced energy expenditure during food restriction. The liver translates metabolic cues regarding energy-deficient status, which is reflected by decreased hepatic insulin action, into soluble LepR, thereby suppressing energy dissipation and assuring survival during food shortages.


Asunto(s)
Insulina , Leptina , Animales , Ratones , Humanos , Leptina/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Peso Corporal , Hipotálamo/metabolismo , Ratones Noqueados , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Metabolismo Energético/genética
6.
Glia ; 71(7): 1626-1647, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36919670

RESUMEN

Hypothalamic circuits compute systemic information to control metabolism. Astrocytes residing within the hypothalamus directly sense nutrients and hormones, integrating metabolic information, and modulating neuronal responses. Nevertheless, the role of the astrocytic circadian clock on the control of energy balance remains unclear. We used mice with a targeted ablation of the core-clock gene Bmal1 within Gfap-expressing astrocytes to gain insight on the role played by this transcription factor in astrocytes. While this mutation does not substantially affect the phenotype in mice fed normo-caloric diet, under high-fat diet we unmasked a thermogenic phenotype consisting of increased energy expenditure, and catabolism in brown adipose and overall metabolic improvement consisting of better glycemia control, and body composition. Transcriptomic analysis in the ventromedial hypothalamus revealed an enhanced response to moderate cellular stress, including ER-stress response, unfolded protein response and autophagy. We identified Xbp1 and Atf1 as two key transcription factors enhancing cellular stress responses. Therefore, we unveiled a previously unknown role of the astrocytic circadian clock modulating energy balance through the regulation of cellular stress responses within the VMH.


Asunto(s)
Relojes Circadianos , Ratones , Animales , Relojes Circadianos/genética , Astrocitos/metabolismo , Hipotálamo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Metabolismo Energético/genética
7.
Plant Commun ; 4(4): 100560, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-36774536

RESUMEN

Heterosis refers to the superior performance of a hybrid compared with its parental lines. Although several genetic and molecular models have been proposed to explain heterosis, it remains unclear how hybrid cells integrate complementary gene expression or activity to drive heterotic growth. In this work, we show that accumulation of growth-promoting and energy metabolism proteins, enhanced energy metabolism activities, and increased protein lysine acetylation were associated with superior growth of the panicle meristem in the elite hybrid rice Shanyou 63 relative to its parental varieties. Metabolism of nuclear/cytosolic acetyl-coenzyme A was also enhanced in the hybrid, which paralleled increases in histone H3 acetylation to selectively target the expression of growth-promoting and metabolic genes. Lysine acetylation of cellular proteins, including TARGET OF RAPAMYCIN complex 1, ribosomal proteins, and energy metabolism enzymes, was also augmented and/or remodeled to modulate their activities. The data indicate that an enhanced network of energy-producing metabolic activity and growth-promoting histone acetylation/gene expression in the hybrid could contribute to its superior growth rate and may constitute a model to explain heterosis.


Asunto(s)
Vigor Híbrido , Oryza , Vigor Híbrido/genética , Lisina/genética , Oryza/genética , Acetilación , Metabolismo Energético/genética
8.
Am J Physiol Endocrinol Metab ; 323(5): E448-E466, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36342228

RESUMEN

Maternal obesity is exceedingly common and strongly linked to offspring obesity and metabolic disease. Hypothalamic function is critical to obesity development. Hypothalamic mechanisms causing obesity following exposure to maternal obesity have not been elucidated. Therefore, we studied a cohort of C57BL/6J dams, treated with a control or high-fat-high-sugar diet, and their adult offspring to explore potential hypothalamic mechanisms to explain the link between maternal and offspring obesity. Dams treated with obesogenic diet were heavier with mild insulin resistance, which is reflective of the most common metabolic disease in pregnancy. Adult offspring exposed to maternal obesogenic diet had no change in body weight but significant increase in fat mass, decreased glucose tolerance, decreased insulin sensitivity, elevated plasma leptin, and elevated plasma thyroid-stimulating hormone. In addition, offspring exposed to maternal obesity had decreased energy intake and activity without change in basal metabolic rate. Hypothalamic neurochemical profile and transcriptome demonstrated decreased neuronal activity and inhibition of oxidative phosphorylation. Collectively, these results indicate that maternal obesity without diabetes is associated with adiposity and decreased hypothalamic energy production in offspring. We hypothesize that altered hypothalamic function significantly contributes to obesity development. Future studies focused on neuroprotective strategies aimed to improve hypothalamic function may decrease obesity development.NEW & NOTEWORTHY Offspring exposed to maternal diet-induced obesity demonstrate a phenotype consistent with energy excess. Contrary to previous studies, the observed energy phenotype was not associated with hyperphagia or decreased basal metabolic rate but rather decreased hypothalamic neuronal activity and energy production. This was supported by neurochemical changes in the hypothalamus as well as inhibition of hypothalamic oxidative phosphorylation pathway. These results highlight the potential for neuroprotective interventions in the prevention of obesity with fetal origins.


Asunto(s)
Resistencia a la Insulina , Enfermedades Metabólicas , Obesidad Materna , Efectos Tardíos de la Exposición Prenatal , Humanos , Animales , Ratones , Femenino , Masculino , Embarazo , Hipotálamo/metabolismo , Obesidad/metabolismo , Metabolismo Energético/genética , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Enfermedades Metabólicas/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos
9.
Int J Obes (Lond) ; 46(11): 2029-2039, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36115924

RESUMEN

OBJECTIVE: Obesity, a leading cause of several metabolic abnormalities, is mainly caused by imbalanced energy homeostasis. IRX3 and IRX5 have been suggested as genetic determinants of obesity in connection with the intronic variants of the FTO gene, the strongest genetic risk factor of polygenic obesity in humans. Although the causal effects of Irx3 and its cooperation with Irx5 in obesity and associated metabolic abnormalities have been demonstrated in vivo, the function of Irx5 in energy homeostasis remains unclear. Here we aim to decipher the actions of Irx5 in the regulation of obesity and metabolic abnormalities. METHODS: We employed a mouse model homozygous for an Irx5-knockout (Irx5KO) allele and determined its metabolic phenotype in the presence or absence of a high-fat diet challenge. To investigate the function of Irx5 in the regulation of energy homeostasis, adipose thermogenesis and hypothalamic leptin response were assessed, and single-cell RNA sequencing (scRNA-seq) in the hypothalamic arcuate-median eminence (ARC-ME) was conducted. RESULTS: Irx5KO mice were leaner and resistant to diet-induced obesity as well as associated metabolic abnormalities, primarily through loss of adiposity. Assessments of energy expenditure and long-term dietary intake revealed that an increase in basal metabolic rate with adipose thermogenesis and a reduction of food intake with improved hypothalamic leptin response in Irx5KO mice may contribute to the anti-obesity effects. Utilizing scRNA-seq and marker gene analyses, we demonstrated the number of ARC-ME neurons was elevated in Irx5KO mice, suggesting a direct role for Irx5 in hypothalamic feeding control. CONCLUSIONS: Our study demonstrates that Irx5 is a genetic factor determining body mass/composition and obesity and regulates both energy expenditure and intake.


Asunto(s)
Leptina , Obesidad , Humanos , Animales , Ratones , Leptina/metabolismo , Obesidad/genética , Obesidad/metabolismo , Dieta Alta en Grasa , Hipotálamo/metabolismo , Metabolismo Energético/genética , Ratones Noqueados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo
10.
Sci Rep ; 12(1): 13374, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35927440

RESUMEN

Leptin exerts its effects on energy balance by inhibiting food intake and increasing energy expenditure via leptin receptors in the hypothalamus. While LepR neurons in the arcuate nucleus of the hypothalamus, the primary target of leptin, have been extensively studied, LepR neurons in other hypothalamic nuclei remain understudied. LepR neurons in the lateral hypothalamus contribute to leptin's effects on food intake and reward, but due to the low abundance of this population it has been difficult to study their molecular profile and responses to energy deficit. We here explore the transcriptome of LepR neurons in the LH and their response to energy deficit. Male LepR-Cre mice were injected in the LH with an AAV carrying Cre-dependent L10:GFP. Few weeks later the hypothalami from fed and food-restricted (24-h) mice were dissected and the TRAP protocol was performed, for the isolation of translating mRNAs from LepR cells in the LH, followed by RNA sequencing. After mapping and normalization, differential expression analysis was performed with DESeq2. We confirm that the isolated mRNA is enriched in LepR transcripts and other known neuropeptide markers of LepRLH neurons, of which we investigate the localization patterns in the LH. We identified novel markers of LepRLH neurons with association to energy balance and metabolic disease, such as Acvr1c, Npy1r, Itgb1, and genes that are differentially regulated by food deprivation, such as Fam46a and Rrad. Our dataset provides a reliable and extensive resource of the molecular makeup of LH LepR neurons and their response to food deprivation.


Asunto(s)
Área Hipotalámica Lateral , Receptores de Leptina , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/genética , Área Hipotalámica Lateral/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
11.
Diabetes ; 71(10): 2106-2122, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35856496

RESUMEN

The neuropeptide AgRP is essential for maintaining systemic energy homeostasis. In the current study, we show that hypothalamic Foxi2, as a novel regulator of nutrient sensing, controls systemic energy metabolism by specifically stimulating AgRP expression. Foxi2 was highly expressed in the hypothalamus, and its expression was induced by fasting. Immunofluorescence assays demonstrated that Foxi2 was localized in AgRP neurons. We stereotactically injected adeno-associated virus to selectively overexpress Foxi2 in AgRP-IRES-Cre mice and found that Foxi2 overexpression in AgRP neurons specifically increased AgRP expression, thereby increasing food intake and reducing energy expenditure, subsequently leading to obesity and insulin resistance. Mechanistically, Foxi2 stimulated AgRP expression by directly binding to it and activating its transcription. Furthermore, Foxi2 overexpression activated AgRP neuron activity, as revealed by whole-cell patch-clamp experiments. Conversely, global Foxi2-mutant mice became leaner with age and were resistant to high-fat diet-induced obesity and metabolic disturbances. Collectively, our data suggest that Foxi2 plays an important role in controlling energy metabolism by regulating AgRP expression.


Asunto(s)
Factores de Transcripción Forkhead , Neuropéptidos , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Metabolismo Energético/genética , Factores de Transcripción Forkhead/metabolismo , Hipotálamo/metabolismo , Ratones , Neuropéptidos/genética , Neuropéptidos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Factores de Transcripción
12.
Matrix Biol ; 111: 53-75, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35671866

RESUMEN

Pulmonary hypertension (PH) comprises a diverse group of disorders that share a common pathway of pulmonary vascular remodeling leading to right ventricular failure. Development of anti-remodeling strategies is an emerging frontier in PH therapeutics that requires a greater understanding of the interactions between vascular wall cells and their extracellular matrices. The ubiquitous matrix glycan, hyaluronan (HA), is markedly elevated in lungs from patients and experimental models with PH. Herein, we identified HA synthase-2 (HAS2) in the pulmonary artery smooth muscle cell (PASMC) layer as a predominant locus of HA dysregulation. HA upregulation involves depletion of NUDT21, a master regulator of alternative polyadenylation, resulting in 3'UTR shortening and hyper-expression of HAS2. The ensuing increase of HAS2 and hyper-synthesis of HA promoted bioenergetic dysfunction of PASMC characterized by impaired mitochondrial oxidative capacity and a glycolytic shift. The resulting HA accumulation stimulated pro-remodeling phenotypes such as cell proliferation, migration, apoptosis-resistance, and stimulated pulmonary artery contractility. Transgenic mice, mimicking HAS2 hyper-synthesis in smooth muscle cells, developed spontaneous PH, whereas targeted deletion of HAS2 prevented experimental PH. Pharmacological blockade of HAS2 restored normal bioenergetics in PASMC, ameliorated cell remodeling phenotypes, and reversed experimental PH in vivo. In summary, our results uncover a novel mechanism of HA hyper-synthesis and downstream effects on pulmonary vascular cell metabolism and remodeling.


Asunto(s)
Metabolismo Energético , Hialuronano Sintasas , Ácido Hialurónico , Hipertensión Pulmonar , Regiones no Traducidas 3'/genética , Animales , Proliferación Celular , Metabolismo Energético/genética , Humanos , Hialuronano Sintasas/genética , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/biosíntesis , Hipertensión Pulmonar/enzimología , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/enzimología
13.
Stem Cell Res Ther ; 13(1): 260, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715869

RESUMEN

BACKGROUND: Mitochondrial dysfunction and mitochondrial DNA (mtDNA) damage in the retinal pigment epithelium (RPE) have been implicated in the pathogenesis of age-related macular degeneration (AMD). However, a deeper understanding is required to determine the contribution of mitochondrial dysfunction and impaired mitochondrial autophagy (mitophagy) to RPE damage and AMD pathobiology. In this study, we model the impact of a prototypical systemic mitochondrial defect, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), in RPE health and homeostasis as an in vitro model for impaired mitochondrial bioenergetics. METHODS: We used induced pluripotent stem cells (iPSCs) derived from skin biopsies of MELAS patients (m.3243A > G tRNA leu mutation) with different levels of mtDNA heteroplasmy and differentiated them into RPE cells. Mitochondrial depletion of ARPE-19 cells (p0 cells) was also performed using 50 ng/mL ethidium bromide (EtBr) and 50 mg/ml uridine. Cell fusion of the human platelets with the p0 cells performed using polyethylene glycol (PEG)/suspension essential medium (SMEM) mixture to generate platelet/RPE "cybrids." Confocal microscopy, FLowSight Imaging cytometry, and Seahorse XF Mito Stress test were used to analyze mitochondrial function. Western Blotting was used to analyze expression of autophagy and mitophagy proteins. RESULTS: We found that MELAS iPSC-derived RPE cells exhibited key characteristics of native RPE. We observed heteroplasmy-dependent impairment of mitochondrial bioenergetics and reliance on glycolysis for generating energy in the MELAS iPSC-derived RPE. The degree of heteroplasmy was directly associated with increased activation of signal transducer and activator of transcription 3 (STAT3), reduced adenosine monophosphate-activated protein kinase α (AMPKα) activation, and decreased autophagic activity. In addition, impaired autophagy was associated with aberrant lysosomal function, and failure of mitochondrial recycling. The mitochondria-depleted p0 cells replicated the effects on autophagy impairment and aberrant STAT3/AMPKα signaling and showed reduced mitochondrial respiration, demonstrating phenotypic similarities between p0 and MELAS iPSC-derived RPE cells. CONCLUSIONS: Our studies demonstrate that the MELAS iPSC-derived disease models are powerful tools for dissecting the molecular mechanisms by which mitochondrial DNA alterations influence RPE function in aging and macular degeneration, and for testing novel therapeutics in patients harboring the MELAS genotype.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome MELAS , Degeneración Macular , Autofagia/genética , ADN Mitocondrial/genética , Metabolismo Energético/genética , Células Epiteliales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome MELAS/genética , Síndrome MELAS/metabolismo , Síndrome MELAS/patología , Degeneración Macular/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Pigmentos Retinianos/metabolismo
14.
Neuroendocrinology ; 112(6): 606-620, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34384081

RESUMEN

INTRODUCTION: The mechanisms underlying obesity are not fully understood, necessitating the creation of novel animal models for the investigation of metabolic disorders. We have previously found that neurosecretory protein GL (NPGL), a newly identified hypothalamic neuropeptide, is involved in feeding behavior and fat accumulation in rats. However, the impact of NPGL on obesity remains unclear in any animal model. The present investigation sought to elucidate whether NPGL causes obesity in the obesity-prone mouse strain C57BL/6J. METHODS: We overexpressed the NPGL-precursor gene (Npgl) in the hypothalamus using adeno-associated virus in male C57BL/6J mice fed normal chow (NC) or a high-calorie diet (HCD). After 9 weeks of Npgl overexpression, we measured adipose tissues, muscle, and several organ masses in addition to food intake and body mass. To assess the effects of Npgl overexpression on peripheral tissues, we analyzed mRNA expression of lipid metabolism-related genes by quantitative RT-PCR. Whole body energy consumption was assessed using an O2/CO2 metabolism measurement before an apparent increase in body mass. RESULTS: Npgl overexpression increased food intake, body mass, adipose tissues and liver masses, and food efficiency under both NC and HCD, resulting in obesity observable within 8 weeks. Furthermore, we observed fat accumulation in adipose tissues and liver. Additionally, mRNA expression of lipid metabolism-related factors was increased in white adipose tissue and the liver after Npgl overexpression. Npgl overexpression inhibited energy expenditure during a dark period. CONCLUSION: Taken together, the present study suggests that NPGL can act as an obesogenic factor that acts within a short period of time in mice. As a result, this Npgl overexpression-induced obesity can be widely applied to study the etiology of obesity from genes to behavior.


Asunto(s)
Hipotálamo , Proteínas del Tejido Nervioso , Animales , Dieta Alta en Grasa , Metabolismo Energético/genética , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Obesidad/genética , Obesidad/metabolismo , ARN Mensajero/metabolismo , Ratas
15.
FEBS J ; 289(22): 6969-6993, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34510753

RESUMEN

Mitochondrial dysfunction is increasingly appreciated as a central contributor to human disease. Oxidative metabolism at the mitochondrial respiratory chain produces ATP and is intricately tied to redox homeostasis and biosynthetic pathways. Metabolic stress arising from genetic mutations in mitochondrial genes and environmental factors such as malnutrition or overnutrition is perceived by the cell and leads to adaptive and maladaptive responses that can underlie pathology. Here, we will outline cellular sensors that react to alterations in energy production, organellar redox, and metabolites stemming from mitochondrial disease (MD) mutations. MD is a heterogeneous group of disorders primarily defined by defects in mitochondrial oxidative phosphorylation from nuclear or mitochondrial-encoded gene mutations. Preclinical therapies that improve fitness of MD mouse models have been recently identified. Targeting metabolic/energetic deficiencies, maladaptive signaling processes, and hyper-oxygenation of tissues are all strategies aside from direct genetic approaches that hold therapeutic promise. A further mechanistic understanding of these curative processes as well as the identification of novel targets will significantly impact mitochondrial biology and disease research.


Asunto(s)
Enfermedades Mitocondriales , Estrés Oxidativo , Humanos , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/metabolismo , Metabolismo Energético/genética , Oxidación-Reducción , Enfermedades Mitocondriales/metabolismo
16.
Am J Hum Biol ; 34(2): e23629, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34146380

RESUMEN

OBJECTIVES: Mitochondria are critical for the survival of eukaryotic organisms due to their ability to produce cellular energy, which drives virtually all aspects of host biology. However, the effects of mitochondrial DNA (mtDNA) variation in relation to disease etiology and adaptation within contemporary global human populations remains incompletely understood. METHODS: To develop a more holistic understanding of the role of mtDNA diversity in human adaptation, health, and disease, we investigated mitochondrial biology and bioenergetics. More specifically, we synthesized details from studies of mitochondrial function and variation in the context of haplogroup background, climatic adaptation, and oxidative disease. RESULTS: The majority of studies show that mtDNA variation arose during modern human dispersal around the world. Some of these variants appear to have been positively selected for their adaptiveness in colder climates, with these sequence changes having implications for tissue-specific function and thermogenic capacity. In addition, many variants modulating energy production are also associated with damaging metabolic byproducts and mitochondrial dysfunction, which, in turn, are implicated in the onset and severity of several different adult mitochondrial diseases. Thus, mtDNA variation that governs bioenergetics, metabolism, and thermoregulation may potentially have adverse consequences for human health, depending on the genetic background and context in which it occurs. CONCLUSIONS: Our review suggests that the mitochondrial research field would benefit from independently replicating mtDNA haplogroup-phenotype associations across global populations, incorporating potentially confounding environmental, demographic, and disease covariates into studies of mtDNA variation, and extending association-based studies to include analyses of complete mitogenomes and assays of mitochondrial function.


Asunto(s)
Adaptación Fisiológica , ADN Mitocondrial , ADN Mitocondrial/genética , Metabolismo Energético/genética , Variación Genética , Haplotipos , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo
17.
Biol Trace Elem Res ; 200(8): 3829-3840, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34750741

RESUMEN

The nutritional selenium (Se) has been demonstrated to have health-boosting effects on fish. However, its effect on fish energy metabolism remains unclear. This study explores the effect and underlying mechanism of the action of nutritional Se on energy metabolism in fish. Rainbow trout (Oncorhynchus mykiss) were fed a basal diet (0 mg Se/kg diet) and a diet containing an already established nutritional Se level (2 mg Se/kg diet, based on Se-yeast) for 30 days. After the feeding experiment, the plasma and liver biochemical profiles and liver transcriptome were analyzed. The results showed that the present nutritional level of Se significantly increased liver triglyceride, total cholesterol, and plasma total cholesterol contents (P < 0.05) compared with the control. Transcriptome analysis showed that 336 and 219 genes were significantly upregulated and downregulated, respectively. Gene enrichment analysis showed that many differentially expressed genes (DEGs) were associated with lipid metabolism pathways (fatty acid biosynthesis, fatty acid elongation, and unsaturated fatty acid biosynthesis), carbohydrate metabolism pathways (glycolysis, the pentose phosphate pathway, and the citrate cycle), and the oxidative phosphorylation pathway. Real-time quantitative PCR (Q-PCR) validation results showed that the expression profiles of 15 genes exhibited similar trends both in RNA sequencing (RNA-seq) and Q-PCR analysis. These results reveal that optimum dietary Se activates glucose catabolic processes, fatty acid biosynthetic processes, and energy production and hence produces higher liver lipid content. This study concludes that the previously established level of nutritional Se (Se-yeast) (2 mg/kg diet, fed basis) for rainbow trout promotes energy storage in the liver, which may benefit fish growth to some extent.


Asunto(s)
Oncorhynchus mykiss , Selenio , Animales , Colesterol , Dieta , Metabolismo Energético/genética , Ácidos Grasos/metabolismo , Hígado/metabolismo , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo , Saccharomyces cerevisiae/metabolismo , Selenio/metabolismo , Transcriptoma
18.
Food Funct ; 12(19): 9238-9247, 2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34606536

RESUMEN

Solanum anguivi Lam. fruits (SALF) are traditionally consumed as a remedy for type 2 diabetes mellitus (T2DM). However, data regarding the potential anti-diabetic effect of SALF and its underlying mechanisms are scarce. As the fruit fly's energy metabolism has been suggested to be comparable with mammals including the secretion of insulin-like peptides, we fed Drosophila melanogaster a high-sugar diet (HSD) to induce a T2DM-like phenotype and subsequently exposed them to a HSD supplemented with SALF. Following, flies were analyzed for various biomarkers in relation to energy metabolism. The HSD-induced glucose levels were significantly down-regulated in flies exposed to a HSD supplemented with SALF. In addition, flies exposed to SALF-supplemented HSD exhibited a better survival in comparison to HSD-fed counterparts. Other parameters of the energy metabolism such as triglyceride levels, weights, and fitness were not affected by SALF supplementation. This was also true for the expression levels of the insulin-like-peptides 3 and 6 as well as for spargel, the Drosophila homolog of PPARγ-co-activator 1α, a central player in mitochondrial biogenesis. Overall, the present study shows that SALF significantly lowered the HSD-induced glucose levels and increased the survival while the biomarkers of the energy metabolism were not affected.


Asunto(s)
Sacarosa en la Dieta/administración & dosificación , Suplementos Dietéticos , Drosophila melanogaster/metabolismo , Frutas , Glucosa/metabolismo , Solanum , Animales , Antioxidantes/análisis , Diabetes Mellitus Tipo 2/metabolismo , Dieta , Drosophila melanogaster/fisiología , Ingestión de Alimentos , Metabolismo Energético/genética , Femenino , Expresión Génica , Modelos Animales , Fenoles/análisis , Fitoquímicos/análisis , Extractos Vegetales/química , Extractos Vegetales/farmacología , Triglicéridos/metabolismo
19.
Nutrients ; 13(10)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34684608

RESUMEN

Anti-obesity activity has been reported for beta-carotene (BC) supplementation at high doses and metformin (MET). We studied whether BC treatment at a closer to dietary dose and MET treatment at a lower than therapeutic dose are effective in ameliorating unwanted effects of an obesogenic diet and whether their combination is advantageous. Obesity-prone mice were challenged with a high-fat diet (HFD, 45% energy as fat) for 4 weeks while receiving a placebo or being treated orally with BC (3 mg/kg/day), MET (100 mg/kg/day), or their combination (BC+MET); a fifth group received a placebo and was kept on a normal-fat diet (10% energy as fat). HFD-induced increases in body weight gain and inguinal white adipose tissue (WAT) adipocyte size were attenuated maximally or selectively in the BC+MET group, in which a redistribution towards smaller adipocytes was noted. Cumulative energy intake was unaffected, yet results suggested increased systemic energy expenditure and brown adipose tissue activation in the treated groups. Unwanted effects of HFD on glucose control and insulin sensitivity were attenuated in the treated groups, especially BC and BC+MET, in which hepatic lipid content was also decreased. Transcriptional analyses suggested effects on skeletal muscle and WAT metabolism could contribute to better responses to the HFD, especially in the MET and BC+MET groups. The results support the benefits of the BC+MET cotreatment.


Asunto(s)
Dieta Alta en Grasa , Metformina/farmacología , Sustancias Protectoras/farmacología , beta Caroteno/farmacología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adiposidad , Animales , Glucemia/metabolismo , Tamaño de la Célula , Metabolismo Energético/genética , Ácidos Grasos/sangre , Regulación de la Expresión Génica , Insulina/sangre , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Aumento de Peso
20.
Sci Rep ; 11(1): 18015, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504172

RESUMEN

Obesity and high-fat diet (HFD) consumption result in hypothalamic inflammation and metabolic dysfunction. While the TLR4 activation by dietary fats is a well-characterized pathway involved in the neuronal and glial inflammation, the role of its accessory proteins in diet-induced hypothalamic inflammation remains unknown. Here, we demonstrate that the knockdown of TLR4-interactor with leucine-rich repeats (Tril), a functional component of TLR4, resulted in reduced hypothalamic inflammation, increased whole-body energy expenditure, improved the systemic glucose tolerance and protection from diet-induced obesity. The POMC-specific knockdown of Tril resulted in decreased body fat, decreased white adipose tissue inflammation and a trend toward increased leptin signaling in POMC neurons. Thus, Tril was identified as a new component of the complex mechanisms that promote hypothalamic dysfunction in experimental obesity and its inhibition in the hypothalamus may represent a novel target for obesity treatment.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Neuronas/metabolismo , Obesidad/etiología , Proopiomelanocortina/genética , Receptor Toll-Like 4/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hipotálamo/patología , Inflamación , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Obesidad/metabolismo , Obesidad/patología , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA