Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(2): 384-99, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23063127

RESUMEN

Vascular endothelial growth factor and its receptors, FLK1/KDR and FLT1, are key regulators of angiogenesis. Unlike FLK1/KDR, the role of FLT1 has remained elusive. FLT1 is produced as soluble (sFLT1) and full-length isoforms. Here, we show that pericytes from multiple tissues produce sFLT1. To define the biologic role of sFLT1, we chose the glomerular microvasculature as a model system. Deletion of Flt1 from specialized glomerular pericytes, known as podocytes, causes reorganization of their cytoskeleton with massive proteinuria and kidney failure, characteristic features of nephrotic syndrome in humans. The kinase-deficient allele of Flt1 rescues this phenotype, demonstrating dispensability of the full-length isoform. Using cell imaging, proteomics, and lipidomics, we show that sFLT1 binds to the glycosphingolipid GM3 in lipid rafts on the surface of podocytes, promoting adhesion and rapid actin reorganization. sFLT1 also regulates pericyte function in vessels outside of the kidney. Our findings demonstrate an autocrine function for sFLT1 to control pericyte behavior.


Asunto(s)
Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Podocitos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Gangliósidos/metabolismo , Humanos , Técnicas In Vitro , Metabolismo de los Lípidos , Lípidos/química , Ratones , Ratones Transgénicos , Pericitos/metabolismo , Proteinuria/metabolismo , Transducción de Señal , Sindecanos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
2.
Cell ; 147(1): 132-46, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21924763

RESUMEN

Alternative splicing (AS) is a key process underlying the expansion of proteomic diversity and the regulation of gene expression. Here, we identify an evolutionarily conserved embryonic stem cell (ESC)-specific AS event that changes the DNA-binding preference of the forkhead family transcription factor FOXP1. We show that the ESC-specific isoform of FOXP1 stimulates the expression of transcription factor genes required for pluripotency, including OCT4, NANOG, NR5A2, and GDF3, while concomitantly repressing genes required for ESC differentiation. This isoform also promotes the maintenance of ESC pluripotency and contributes to efficient reprogramming of somatic cells into induced pluripotent stem cells. These results reveal a pivotal role for an AS event in the regulation of pluripotency through the control of critical ESC-specific transcriptional programs.


Asunto(s)
Empalme Alternativo , Reprogramación Celular , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Animales , ADN/metabolismo , Células Madre Embrionarias/citología , Genes Homeobox , Humanos , Ratones , Células Madre Pluripotentes/citología , Isoformas de Proteínas/metabolismo
3.
Brain Behav Immun ; 119: 637-647, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38663773

RESUMEN

Obesity is a major modifiable risk factor for Alzheimer's disease (AD), characterized by progressive atrophy of the cerebral cortex. The neurobiology of obesity contributions to AD is poorly understood. Here we show with in vivo MRI that diet-induced obesity decreases cortical volume in mice, and that higher body adiposity associates with lower cortical volume in humans. Single-nuclei transcriptomics of the mouse cortex reveals that dietary obesity promotes an array of neuron-adverse transcriptional dysregulations, which are mediated by an interplay of excitatory neurons and glial cells, and which involve microglial activation and lowered neuronal capacity for neuritogenesis and maintenance of membrane potential. The transcriptional dysregulations of microglia, more than of other cell types, are like those in AD, as assessed with single-nuclei cortical transcriptomics in a mouse model of AD and two sets of human donors with the disease. Serial two-photon tomography of microglia demonstrates microgliosis throughout the mouse cortex. The spatial pattern of adiposity-cortical volume associations in human cohorts interrogated together with in silico bulk and single-nucleus transcriptomic data from the human cortex implicated microglia (along with other glial cells and subtypes of excitatory neurons), and it correlated positively with the spatial profile of cortical atrophy in patients with mild cognitive impairment and AD. Thus, multi-cell neuron-adverse dysregulations likely contribute to the loss of cortical tissue in obesity. The dysregulations of microglia may be pivotal to the obesity-related risk of AD.


Asunto(s)
Enfermedad de Alzheimer , Corteza Cerebral , Obesidad , Animales , Obesidad/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Masculino , Microglía/metabolismo , Neuronas/metabolismo , Femenino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Imagen por Resonancia Magnética , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/etiología , Atrofia , Dieta Alta en Grasa/efectos adversos , Anciano , Adiposidad , Transcriptoma
4.
Nature ; 563(7733): 701-704, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30429614

RESUMEN

Human pluripotent cell lines hold enormous promise for the development of cell-based therapies. Safety, however, is a crucial prerequisite condition for clinical applications. Numerous groups have attempted to eliminate potentially harmful cells through the use of suicide genes1, but none has quantitatively defined the safety level of transplant therapies. Here, using genome-engineering strategies, we demonstrate the protection of a suicide system from inactivation in dividing cells. We created a transcriptional link between the suicide gene herpes simplex virus thymidine kinase (HSV-TK) and a cell-division gene (CDK1); this combination is designated the safe-cell system. Furthermore, we used a mathematical model to quantify the safety level of the cell therapy as a function of the number of cells that is needed for the therapy and the type of genome editing that is performed. Even with the highly conservative estimates described here, we anticipate that our solution will rapidly accelerate the entry of cell-based medicine into the clinic.


Asunto(s)
Proteína Quinasa CDC2/genética , División Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Genes Transgénicos Suicidas/genética , Seguridad del Paciente , Animales , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/normas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Ganciclovir/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
5.
Physiology (Bethesda) ; 35(3): 185-195, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32293230

RESUMEN

Intermittent fasting (IF) is a widely practiced dietary method that encompasses periodic restriction of food consumption. Due to its protective benefits against metabolic diseases, aging, and cardiovascular and neurodegenerative diseases, IF continues to gain attention as a preventative and therapeutic intervention to counteract these chronic diseases. Although numerous animal studies have reported positive health benefits of IF, its feasibility and efficacy in clinical settings remain controversial. Importantly, since dietary interventions such as IF have systemic effects, thoroughly investigating the tissue-specific changes in animal models is crucial to identify IF's mechanism and evaluate its potential adverse effects in humans. As such, we will review and compare the outcomes and underlying mechanisms of IF in both animal and human studies. Moreover, the limitations of IF and inconsistencies between preclinical and clinical studies will be discussed to provide insight into the gaps between translating research from bench to bedside.


Asunto(s)
Ayuno , Ayuno Intermitente , Animales , Humanos , Ayuno/efectos adversos , Ayuno/metabolismo , Obesidad/metabolismo , Restricción Calórica/efectos adversos , Dieta
6.
Nature ; 507(7492): 371-5, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24646999

RESUMEN

Genome-wide association studies (GWAS) have reproducibly associated variants within introns of FTO with increased risk for obesity and type 2 diabetes (T2D). Although the molecular mechanisms linking these noncoding variants with obesity are not immediately obvious, subsequent studies in mice demonstrated that FTO expression levels influence body mass and composition phenotypes. However, no direct connection between the obesity-associated variants and FTO expression or function has been made. Here we show that the obesity-associated noncoding sequences within FTO are functionally connected, at megabase distances, with the homeobox gene IRX3. The obesity-associated FTO region directly interacts with the promoters of IRX3 as well as FTO in the human, mouse and zebrafish genomes. Furthermore, long-range enhancers within this region recapitulate aspects of IRX3 expression, suggesting that the obesity-associated interval belongs to the regulatory landscape of IRX3. Consistent with this, obesity-associated single nucleotide polymorphisms are associated with expression of IRX3, but not FTO, in human brains. A direct link between IRX3 expression and regulation of body mass and composition is demonstrated by a reduction in body weight of 25 to 30% in Irx3-deficient mice, primarily through the loss of fat mass and increase in basal metabolic rate with browning of white adipose tissue. Finally, hypothalamic expression of a dominant-negative form of Irx3 reproduces the metabolic phenotypes of Irx3-deficient mice. Our data suggest that IRX3 is a functional long-range target of obesity-associated variants within FTO and represents a novel determinant of body mass and composition.


Asunto(s)
Proteínas de Homeodominio/genética , Intrones/genética , Oxigenasas de Función Mixta/genética , Obesidad/genética , Oxo-Ácido-Liasas/genética , Proteínas/genética , Factores de Transcripción/genética , Tejido Adiposo/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Metabolismo Basal/genética , Índice de Masa Corporal , Peso Corporal/genética , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/genética , Dieta , Genes Dominantes/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Delgadez/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
7.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L740-L750, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30702342

RESUMEN

In healthy blood vessels, albumin crosses the endothelium to leave the circulation by transcytosis. However, little is known about the regulation of albumin transcytosis or how it differs in different tissues; its physiological purpose is also unclear. Using total internal reflection fluorescence microscopy, we quantified transcytosis of albumin across primary human microvascular endothelial cells from both lung and skin. We then validated our in vitro findings using a tissue-specific knockout mouse model. We observed that albumin transcytosis was saturable in the skin but not the lung microvascular endothelial cells, implicating a receptor-mediated process. We identified the scavenger receptor CD36 as being both necessary and sufficient for albumin transcytosis across dermal microvascular endothelium, in contrast to the lung where macropinocytosis dominated. Mutations in the apical helical bundle of CD36 prevented albumin internalization by cells. Mice deficient in CD36 specifically in endothelial cells exhibited lower basal permeability to albumin and less basal tissue edema in the skin but not in the lung. Finally, these mice also exhibited a smaller subcutaneous fat layer despite having identical total body weights and circulating fatty acid levels as wild-type animals. In conclusion, CD36 mediates albumin transcytosis in the skin but not the lung. Albumin transcytosis may serve to regulate fatty acid delivery from the circulation to tissues.


Asunto(s)
Albúminas/metabolismo , Antígenos CD36/metabolismo , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Animales , Antígenos CD36/química , Antígenos CD36/deficiencia , Antígenos CD36/genética , Células Cultivadas , Células Endoteliales/citología , Humanos , Pulmón/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/citología , Microvasos/metabolismo , Mutagénesis Sitio-Dirigida , Pinocitosis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Piel/irrigación sanguínea , Grasa Subcutánea/anatomía & histología , Grasa Subcutánea/metabolismo , Distribución Tisular , Transcitosis
8.
Nature ; 498(7453): 241-5, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23739326

RESUMEN

Previous investigations of the core gene regulatory circuitry that controls the pluripotency of embryonic stem (ES) cells have largely focused on the roles of transcription, chromatin and non-coding RNA regulators. Alternative splicing represents a widely acting mode of gene regulation, yet its role in regulating ES-cell pluripotency and differentiation is poorly understood. Here we identify the muscleblind-like RNA binding proteins, MBNL1 and MBNL2, as conserved and direct negative regulators of a large program of cassette exon alternative splicing events that are differentially regulated between ES cells and other cell types. Knockdown of MBNL proteins in differentiated cells causes switching to an ES-cell-like alternative splicing pattern for approximately half of these events, whereas overexpression of MBNL proteins in ES cells promotes differentiated-cell-like alternative splicing patterns. Among the MBNL-regulated events is an ES-cell-specific alternative splicing switch in the forkhead family transcription factor FOXP1 that controls pluripotency. Consistent with a central and negative regulatory role for MBNL proteins in pluripotency, their knockdown significantly enhances the expression of key pluripotency genes and the formation of induced pluripotent stem cells during somatic cell reprogramming.


Asunto(s)
Empalme Alternativo , Reprogramación Celular , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Unión al ARN/metabolismo , Empalme Alternativo/genética , Secuencias de Aminoácidos , Animales , Diferenciación Celular/genética , Línea Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Ratones , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas Represoras/metabolismo
9.
Curr Hypertens Rep ; 20(11): 96, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30229358

RESUMEN

PURPOSE OF REVIEW: Obesity is a major risk factor for the development of hypertension (HTN), a leading cause of cardiovascular morbidity and mortality. Growing body of research suggests that adipose tissue function is directly associated with the pathogenesis of obesity-related HTN. In this review, we will discuss recent research on the role of adipose tissue in blood pressure (BP) regulation and activation of brown adipose tissue (BAT) as a potentially new therapeutic means for obesity-related HTN. RECENT FINDINGS: Adipose tissue provides mechanical protection of the blood vessels and plays a role in regulation of vascular tone. Exercise and fasting activate BAT and induce browning of white adipose tissue (WAT). BAT-secreted FGF21 lowers BP and protects against HTN. Browning of perivascular WAT improves HTN. New insights on WAT browning and BAT activation can open new avenues of potential therapeutic interventions to treat obesity-related HTN.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Hipertensión/metabolismo , Obesidad/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Metabolismo Energético/fisiología , Ejercicio Físico/fisiología , Ayuno/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Metabolismo de los Lípidos , Termogénesis/fisiología , Hormonas Tiroideas/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
10.
FASEB J ; 30(3): 1276-86, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26675707

RESUMEN

Cellular senescence, an irreversible state of growth arrest, underlies organismal aging and age-related diseases. Recent evidence suggests that aging intervention based on inhibition of cellular senescence might be a promising strategy for treatment of aging and age-related diseases. Embryonic stem cells (ESCs) and ESC conditioned medium (CM) have been suggested as a desirable source for regenerative medicine. However, effects of ESC-CM on cellular senescence remain to be determined. We found that treatment of senescent human dermal fibroblasts with CM from mouse ESCs (mESCs) decreases senescence phenotypes. We found that platelet-derived growth factor BB in mESC-CM plays a critical role in antisenescence effect of mESC-CM through up-regulation of fibroblast growth factor 2. We confirmed that mESC-CM treatment accelerates the wound-healing process by down-regulating senescence-associated p53 expression in in vivo models. Taken together, our results suggest that mESC-CM has the ability to suppress cellular senescence and maintain proliferative capacity. Therefore, this strategy might emerge as a novel therapeutic strategy for aging and age-related diseases.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Becaplermina , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ratones , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Nature ; 458(7239): 766-70, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19252478

RESUMEN

Transgenic expression of just four defined transcription factors (c-Myc, Klf4, Oct4 and Sox2) is sufficient to reprogram somatic cells to a pluripotent state. The resulting induced pluripotent stem (iPS) cells resemble embryonic stem cells in their properties and potential to differentiate into a spectrum of adult cell types. Current reprogramming strategies involve retroviral, lentiviral, adenoviral and plasmid transfection to deliver reprogramming factor transgenes. Although the latter two methods are transient and minimize the potential for insertion mutagenesis, they are currently limited by diminished reprogramming efficiencies. piggyBac (PB) transposition is host-factor independent, and has recently been demonstrated to be functional in various human and mouse cell lines. The PB transposon/transposase system requires only the inverted terminal repeats flanking a transgene and transient expression of the transposase enzyme to catalyse insertion or excision events. Here we demonstrate successful and efficient reprogramming of murine and human embryonic fibroblasts using doxycycline-inducible transcription factors delivered by PB transposition. Stable iPS cells thus generated express characteristic pluripotency markers and succeed in a series of rigorous differentiation assays. By taking advantage of the natural propensity of the PB system for seamless excision, we show that the individual PB insertions can be removed from established iPS cell lines, providing an invaluable tool for discovery. In addition, we have demonstrated the traceless removal of reprogramming factors joined with viral 2A sequences delivered by a single transposon from murine iPS lines. We anticipate that the unique properties of this virus-independent simplification of iPS cell production will accelerate this field further towards full exploration of the reprogramming process and future cell-based therapies.


Asunto(s)
Diferenciación Celular , Reprogramación Celular/genética , Fibroblastos/citología , Fibroblastos/fisiología , Vectores Genéticos/genética , Células Madre Pluripotentes/fisiología , Animales , Línea Celular , Células Cultivadas , Elementos Transponibles de ADN , Fibroblastos/virología , Orden Génico , Técnicas de Transferencia de Gen , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones Desnudos , Alineación de Secuencia , Factores de Transcripción/genética , Transgenes/genética
12.
Biol Reprod ; 91(4): 87, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25122061

RESUMEN

Given the angiogenic function of vascular endothelial growth factor A (VEGFA), the function of its expression by trophoblast in the avascular placental junctional zone is unknown. In mice, cells from the trophoblast-specific protein alpha (Tpbpa) lineage populate this zone and, in late gestation, some of these cells invade the decidual layer. To diminish Vegfa expression in Tpbpa cells, we crossed Vegfa(flox/flox) females with males carrying Tpbpa-Cre. For single deletion (sd) of Vegfa in Tpbpa cells in 100% of conceptuses (SD100 pregnancies, sd conceptuses) we crossed homozygous lines. For double deletion (dd) of both Vegfa alleles in 50% of the conceptuses (DD50 pregnancies, 50% dd conceptuses and 50% no deletion [nd]), we crossed homozygous Vegfa(flox/flox) females with males heterozygous for Tpbpa-Cre and homozygous for Vegfa(flox/flox). Controls were Vegfa(flox/flox) females bred to wild-type males (V-CTRL pregnancies). In SD100 pregnancies, maternal plasma immunoreactive VEGFA significantly increased and arterial blood pressure decreased, whereas fetal body weight and placental Flt1, sFlt1, and Prl3b1 mRNA were unchanged. In DD50, maternal immunoreactive VEGFA and arterial pressures were unaltered, but both dd and nd conceptuses exhibited significantly increased embryonic lethality, altered expression of Flt1, sFlt1, and Prl3b1 mRNA in the decidual layer, and decreased fetal body weight relative to V-CTRL. Maternal cardiac output significantly increased in proportion to dd conceptuses in the pregnancy. In DD50, results are consistent with altered maternal function beginning in early gestation and adversely impacting both conceptus genotypes. We conclude that maternal function is influenced by Vegfa expression in trophoblast cells at the maternal-fetal interface, likely via an endocrine mechanism.


Asunto(s)
Placenta/metabolismo , Trofoblastos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Linaje de la Célula , Femenino , Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Integrasas/genética , Integrasas/metabolismo , Intercambio Materno-Fetal , Ratones , Ratones Transgénicos , Placenta/citología , Circulación Placentaria/fisiología , Embarazo , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
13.
Geroscience ; 46(3): 3457-3470, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38379117

RESUMEN

The aging of white adipose tissue (WAT) involves senescence of adipose stem and progenitor cells (ASPCs) and dysregulation of immune cell populations, serving as a major driver of age-associated adipose dysfunction and metabolic diseases. Conversely, the elimination of senescent ASPCs is associated with improvements in overall health. Intermittent fasting (IF), a dietary intervention that incorporates periodic cycles of fasting and refeeding, has been reported to promote weight loss and fat mass reduction and improve glucose and insulin homeostasis in both murine and human studies. While previous studies have assessed the effects of IF on obesity-associated metabolic dysfunction, few studies have examined the aging-specific changes to ASPCs and immune cell populations in WAT. Here, we show that IF in 18-20-month-old mice reduced senescent phenotypes of ASPCs and restored their adipogenic potential. Intriguingly, IF-treated mice exhibited an increase in adipose eosinophils, which has been reported to be associated with improved WAT homeostasis and immunological fitness in aged mice. The observed cellular and metabolic changes suggest that IF may be a feasible lifestyle regimen to reduce cellular senescence which could result in attenuation of downstream aging-induced WAT dysfunction and metabolic diseases.


Asunto(s)
Inmunosenescencia , Enfermedades Metabólicas , Ratones , Humanos , Animales , Anciano , Ayuno Intermitente , Rejuvenecimiento , Tejido Adiposo Blanco/metabolismo , Obesidad/metabolismo , Enfermedades Metabólicas/metabolismo
14.
Cell Rep ; 43(1): 113584, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38117653

RESUMEN

Severe burns induce a chronic hypermetabolic state that persists well past wound closure, indicating that additional internal mechanisms must be involved. Adipose tissue is suggested to be a central regulator in perpetuating hypermetabolism, although this has not been directly tested. Here, we show that thermogenic adipose tissues are activated in parallel to increases in hypermetabolism independent of cold stress. Using an adipose tissue transplantation model, we discover that burn-derived subcutaneous white adipose tissue alone is sufficient to invoke a hypermetabolic response in a healthy recipient mouse. Concomitantly, transplantation of healthy adipose tissue alleviates metabolic dysfunction in a burn recipient. We further show that the nicotinic acetylcholine receptor signaling pathway may mediate an immune-adipose crosstalk to regulate adipose tissue remodeling post-injury. Targeting this pathway could lead to innovative therapeutic interventions to counteract hypermetabolic pathologies.


Asunto(s)
Quemaduras , Grasa Subcutánea , Animales , Ratones , Grasa Subcutánea/metabolismo , Tejido Adiposo Blanco/metabolismo , Obesidad/metabolismo , Metabolismo Energético/fisiología , Quemaduras/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo/metabolismo
15.
Diabetes ; 73(7): 1084-1098, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38656552

RESUMEN

Forkhead box O1 (FOXO1) regulates muscle growth, but the metabolic role of FOXO1 in skeletal muscle and its mechanisms remain unclear. To explore the metabolic role of FOXO1 in skeletal muscle, we generated skeletal muscle-specific Foxo1 inducible knockout (mFOXO1 iKO) mice and fed them a high-fat diet to induce obesity. We measured insulin sensitivity, fatty acid oxidation, mitochondrial function, and exercise capacity in obese mFOXO1 iKO mice and assessed the correlation between FOXO1 and mitochondria-related protein in the skeletal muscle of patients with diabetes. Obese mFOXO1 iKO mice exhibited improved mitochondrial respiratory capacity, which was followed by attenuated insulin resistance, enhanced fatty acid oxidation, and improved skeletal muscle exercise capacity. Transcriptional inhibition of FOXO1 in peroxisome proliferator-activated receptor δ (PPARδ) expression was confirmed in skeletal muscle, and deletion of PPARδ abolished the beneficial effects of FOXO1 deficiency. FOXO1 protein levels were higher in the skeletal muscle of patients with diabetes and negatively correlated with PPARδ and electron transport chain protein levels. These findings highlight FOXO1 as a new repressor in PPARδ gene expression in skeletal muscle and suggest that FOXO1 links insulin resistance and mitochondrial dysfunction in skeletal muscle via PPARδ.


Asunto(s)
Proteína Forkhead Box O1 , Resistencia a la Insulina , Ratones Noqueados , Músculo Esquelético , PPAR delta , Animales , Resistencia a la Insulina/fisiología , Resistencia a la Insulina/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , PPAR delta/genética , PPAR delta/metabolismo , Ratones , Músculo Esquelético/metabolismo , Humanos , Masculino , Mitocondrias Musculares/metabolismo , Dieta Alta en Grasa , Obesidad/metabolismo , Obesidad/genética , Mitocondrias/metabolismo
16.
Biomolecules ; 13(5)2023 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-37238732

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that affects motor neurons, leading to muscle weakness, paralysis, and eventual death. Research from the past few decades has appreciated that ALS is not only a disease of the motor neurons but also a disease that involves systemic metabolic dysfunction. This review will examine the foundational research of understanding metabolic dysfunction in ALS and provide an overview of past and current studies in ALS patients and animal models, spanning from full systems to various metabolic organs. While ALS-affected muscle tissue exhibits elevated energy demand and a fuel preference switch from glycolysis to fatty acid oxidation, adipose tissue in ALS undergoes increased lipolysis. Dysfunctions in the liver and pancreas contribute to impaired glucose homeostasis and insulin secretion. The central nervous system (CNS) displays abnormal glucose regulation, mitochondrial dysfunction, and increased oxidative stress. Importantly, the hypothalamus, a brain region that controls whole-body metabolism, undergoes atrophy associated with pathological aggregates of TDP-43. This review will also cover past and present treatment options that target metabolic dysfunction in ALS and provide insights into the future of metabolism research in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Animales , Esclerosis Amiotrófica Lateral/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas Motoras/metabolismo , Modelos Animales , Glucosa/metabolismo
17.
Semin Nephrol ; 43(2): 151405, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37542985

RESUMEN

Polycystic kidney disease (PKD) is a chronic, progressive hereditary condition characterized by abnormal development and growth of cysts in the kidneys and other organs. There is increasing interest in exploring whether dietary modifications may prevent or slow the disease course in people with PKD. Although vasopressin-receptor agonists have emerged as a novel drug treatment in advancing care for people with PKD, several recent landmark trials and clinical discoveries also have provided new insights into potential dietary-related therapeutic strategies. In this review, we summarize the current evidence pertaining to nutrients, foods, dietary patterns, cyst growth, and progression of PKD. We also describe existing evidence-based dietary care for people with PKD and outline the potential implications for advancing evidence-based dietary interventions. Semin Nephrol 43:x-xx © 2023 Elsevier Inc. All rights reserved.


Asunto(s)
Enfermedades Renales Poliquísticas , Humanos , Riñón , Dieta , Nutrientes
18.
Obes Surg ; 33(8): 2443-2451, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37380880

RESUMEN

PURPOSE: Obesity can be associated with chronic inflammation and dysregulated expression of inflammatory adipokines that contribute to insulin resistance and type 2 diabetes. This may also affect the clinical response to bariatric surgery. Our objective was whether baseline visceral adipose tissue features and plasma adipokine are associated with HbA1c ≥0.06 at the time of Roux-en-Y gastric bypass (RYGB) surgery and with persistently elevated HbA1c at 12 months post-RYGB. METHODS: During the surgery, adipose biopsies and plasma were collected for adipokine/cytokine profile. Clinical and biochemical measurements were also collected at the time of RYGB and, in those with baseline elevated HbA1c, at 12 months post-RYGB. RESULTS: In the cross-sectional study, 109 patients (82.6% female; age 49 years; BMI 46.98 kg/m2) participated. Of those with elevated HbA1c at baseline (n = 61), 47 patients had repeated measurements at 12 months post-RYGB (23% drop-out). Using a multivariate logistic regression model, older age (adjusted odds ratio (aOR), 1.14; 95% confidence interval (CI), 1.06-1.22) and higher plasma resistin (aOR, 5.30; 95% CI, 1.25-22.44) were associated with higher odds of HbA1c ≥ 0.06, whereas higher plasma adiponectin (aOR, 0.993; 95% CI, 0.99-0.996) was associated with lower odds of HbA1c ≥0.06. In addition, baseline higher average adipose cell area (aOR, 1.0017; 95% CI, 1.0002-1.0032) and plasma resistin (aOR, 1.0004; 95% CI, 1.0000-1.0009) were associated with higher odds of having persistently elevated HbA1c at 12 months post-RYGB. CONCLUSION: Our study suggests that baseline plasma adipokine dysregulation, specifically high resistin, and adipocyte hypertrophy may affect the clinical response to RYGB.


Asunto(s)
Cirugía Bariátrica , Diabetes Mellitus Tipo 2 , Derivación Gástrica , Obesidad Mórbida , Humanos , Femenino , Persona de Mediana Edad , Masculino , Estudios Transversales , Diabetes Mellitus Tipo 2/complicaciones , Obesidad Mórbida/cirugía , Hemoglobina Glucada , Resistina/metabolismo , Estudios de Cohortes , Obesidad/cirugía , Tejido Adiposo/metabolismo , Adipoquinas
19.
Nat Biomed Eng ; 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996616

RESUMEN

The immunogenicity of transplanted allogeneic cells and tissues is a major hurdle to the advancement of cell therapies. Here we show that the overexpression of eight immunomodulatory transgenes (Pdl1, Cd200, Cd47, H2-M3, Fasl, Serpinb9, Ccl21 and Mfge8) in mouse embryonic stem cells (mESCs) is sufficient to immunologically 'cloak' the cells as well as tissues derived from them, allowing their survival for months in outbred and allogeneic inbred recipients. Overexpression of the human orthologues of these genes in human ESCs abolished the activation of allogeneic human peripheral blood mononuclear cells and their inflammatory responses. Moreover, by using the previously reported FailSafe transgene system, which transcriptionally links a gene essential for cell division with an inducible and cell-proliferation-dependent kill switch, we generated cloaked tissues from mESCs that served as immune-privileged subcutaneous sites that protected uncloaked allogeneic and xenogeneic cells from rejection in immune-competent hosts. The combination of cloaking and FailSafe technologies may allow for the generation of safe and allogeneically accepted cell lines and off-the-shelf cell products.

20.
Cell Rep Med ; 4(5): 101051, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37196633

RESUMEN

Alterations in the microbiome correlate with improved metabolism in patients following bariatric surgery. While fecal microbiota transplantation (FMT) from obese patients into germ-free (GF) mice has suggested a significant role of the gut microbiome in metabolic improvements following bariatric surgery, causality remains to be confirmed. Here, we perform paired FMT from the same obese patients (BMI > 40; four patients), pre- and 1 or 6 months post-Roux-en-Y gastric bypass (RYGB) surgery, into Western diet-fed GF mice. Mice colonized by FMT from patients' post-surgery stool exhibit significant changes in microbiota composition and metabolomic profiles and, most importantly, improved insulin sensitivity compared with pre-RYGB FMT mice. Mechanistically, mice harboring the post-RYGB microbiome show increased brown fat mass and activity and exhibit increased energy expenditure. Moreover, improvements in immune homeostasis within the white adipose tissue are also observed. Altogether, these findings point to a direct role for the gut microbiome in mediating improved metabolic health post-RYGB surgery.


Asunto(s)
Cirugía Bariátrica , Microbioma Gastrointestinal , Resistencia a la Insulina , Ratones , Animales , Tejido Adiposo Pardo , Obesidad/cirugía , Metabolismo Energético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA