Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Am J Physiol Endocrinol Metab ; 327(1): E13-E26, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38717362

RESUMEN

Adipose tissue metabolism is actively involved in the regulation of energy balance. Adipose-derived stem cells (ASCs) play a critical role in maintaining adipose tissue function through their differentiation into mature adipocytes (Ad). This study aimed to investigate the impact of an obesogenic environment on the epigenetic landscape of ASCs and its impact on adipocyte differentiation and its metabolic consequences. Our results showed that ASCs from rats on a high-fat sucrose (HFS) diet displayed reduced adipogenic capacity, increased fat accumulation, and formed larger adipocytes than the control (C) group. Mitochondrial analysis revealed heightened activity in undifferentiated ASC-HFS but decreased respiratory and glycolytic capacity in mature adipocytes. The HFS diet significantly altered the H3K4me3 profile in ASCs on genes related to adipogenesis, mitochondrial function, inflammation, and immunomodulation. After differentiation, adipocytes retained H3K4me3 alterations, confirming the upregulation of genes associated with inflammatory and immunomodulatory pathways. RNA-seq confirmed the upregulation of genes associated with inflammatory and immunomodulatory pathways in adipocytes. Overall, the HFS diet induced significant epigenetic and transcriptomic changes in ASCs, impairing differentiation and causing dysfunctional adipocyte formation.NEW & NOTEWORTHY Obesity is associated with the development of chronic diseases like metabolic syndrome and type 2 diabetes, and adipose tissue plays a crucial role. In a rat model, our study reveals how an obesogenic environment primes adipocyte precursor cells, leading to epigenetic changes that affect inflammation, adipogenesis, and mitochondrial activity after differentiation. We highlight the importance of histone modifications, especially the trimethylation of histone H3 to lysine 4 (H3K4me3), showing its influence on adipocyte expression profiles.


Asunto(s)
Adipocitos , Adipogénesis , Tejido Adiposo , Dieta Alta en Grasa , Epigénesis Genética , Histonas , Transcriptoma , Animales , Ratas , Adipocitos/metabolismo , Dieta Alta en Grasa/efectos adversos , Histonas/metabolismo , Masculino , Adipogénesis/genética , Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Diferenciación Celular/genética , Células Madre/metabolismo , Obesidad/metabolismo , Obesidad/genética , Reprogramación Celular/fisiología , Células Cultivadas , Ratas Wistar , Ratas Sprague-Dawley
2.
Pediatr Obes ; 19(6): e13120, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38590200

RESUMEN

Maternal obesity is a well-known risk factor for developing premature obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes in the progeny. The development of white adipose tissue is a dynamic process that starts during prenatal life: fat depots laid down in utero are associated with the proportion of fat in children later on. How early this programming takes place is still unknown. However, recent evidence shows that mesenchymal stem cells (MSC), the embryonic adipocyte precursor cells, show signatures of the early setting of an adipogenic committed phenotype when exposed to maternal obesity. This review aims to present current findings on the cellular adaptations of MSCs from the offspring of women with obesity and how the metabolic environment of MSCs could affect the early commitment towards adipocytes. In conclusion, maternal obesity can induce early programming of fetal adipose tissue by conditioning MSCs. These cells have higher expression of adipogenic markers, altered insulin signalling and mitochondrial performance, compared to MSCs of neonates from lean pregnancies. Fetal MSCs imprinting by maternal obesity could help explain the increased risk of childhood obesity and development of further noncommunicable diseases.


Asunto(s)
Células Madre Mesenquimatosas , Obesidad Materna , Efectos Tardíos de la Exposición Prenatal , Humanos , Femenino , Embarazo , Obesidad Materna/metabolismo , Tejido Adiposo , Obesidad Infantil , Adipogénesis/fisiología , Recién Nacido , Adipocitos
3.
Rev. bras. zootec ; 51: e20220061, 2022. ilus, tab
Artículo en Inglés | VETINDEX | ID: biblio-1442979

RESUMEN

The productive traits of beef cattle are orchestrated by their genetics, postnatal environmental conditions, and also by the intrauterine background. Both under- or overnutrition, as specific dietary components, are able to promote persistent effects on the offspring. This occurs because dietary factors act not only affecting the availability of substrates for fetal anabolism and oxidative metabolism, but also as signals that regulate several events toward fetal development. Therefore, this study aimed to summarize the gestational nutrition effects on the offspring performance and meat quality in a long term. Overall, studies have shown that many of these alterations are under the control of epigenetic mechanisms, as DNA methylation, histones modification, and non-coding RNA. The current knowledge has indicated that the fetal programming responses are dependent on the window of fetal development in which the dietary treatment is applied, the intensity of maternal nutritional stimuli, and the treatment application length. Collectively, studies demonstrated that muscle cell hyperplasia is impaired when maternal requirements were not achieved in the second third of gestation, which limits the formation of a greater number of muscle fibers and the offspring growth potential in a long term. Changes in muscle fibers metabolism and in collagen content were also reported as consequence of a dietary perturbation during pregnancy. In contrast, a maternal overnutrition during the late pregnancy has been associated with beneficial responses on meat quality. In summary, ensuring an adequate maternal environment during the fetal development is crucial to enhance the productive responses in beef cattle operations.(AU)


Asunto(s)
Animales , Femenino , Embarazo , Bovinos/fisiología , Adipogénesis/fisiología , Carne/análisis , Atención Prenatal/métodos
4.
Life Sci ; 287: 120066, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34678264

RESUMEN

AIM: An adverse endogenous environment during early life predisposes to metabolic disorder development. We previously reported adverse metabolic and adipose tissue effects in adult male rats born to dams fed with a fructose-rich diet (FRD). The aim of this work was to determine the effect of a FRD consumed by the pregnant mother on the white adipose tissue (WAT) browning capacity of male offspring at adulthood. MAIN METHODS: Adult SD male offspring from control (C) and FRD-fed mothers were exposed during one week to a cold stimulus. WAT browning capacity was studied through in vivo and in vitro approaches. KEY FINDINGS: After cold exposure, WAT browning was higher in fructose-programmed animals as evidenced by an increase in ucp-1 gene expression, protein levels, and higher UCP-1 positive foci. Moreover, pgc1-α gene expression was increased. In vitro studies showed a lower adipogenic capacity in cells of prenatally fructose-exposed animals differentiated with a white differentiation cocktail, while a higher ucp-1 expression was noted when their cells were treated with a pro-beige differentiation cocktail. SIGNIFICANCE: For the first time we demonstrate that pre-natal fructose exposure predisposes programmed male rats to a higher WAT browning-induced response, under stimulated conditions, despite an apparent lower basal thermogenic capacity. These results should be considered in future studies to generate new therapeutic approaches to deal with adverse programming malnutrition effects.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Frío/efectos adversos , Azúcares de la Dieta/toxicidad , Fructosa/toxicidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Termogénesis/fisiología , Adipogénesis/efectos de los fármacos , Adipogénesis/fisiología , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Azúcares de la Dieta/administración & dosificación , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Femenino , Fructosa/administración & dosificación , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Ratas Sprague-Dawley , Termogénesis/efectos de los fármacos
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(9): 166167, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33989739

RESUMEN

BACKGROUND: Adipocytes from lipodystrophic Agpat2-/- mice have impaired adipogenesis and fewer caveolae. Herein, we examined whether these defects are associated with changes in lipid composition or abnormal levels of caveolae-associated proteins. Lipidome changes were quantified in differentiated Agpat2-/- adipocytes to identify lipids with potential adipogenic roles. METHODS: Agpat2-/- and wild type brown preadipocytes were differentiated in vitro. Plasma membrane was purified by ultracentrifugation. Number of caveolae and caveolae-associated proteins, as well as sterol, sphingolipid, and phospholipid lipidome were determined across differentiation. RESULTS: Differentiated Agpat2-/- adipocytes had decreased caveolae number but conserved insulin signaling. Caveolin-1 and cavin-1 levels were equivalent between Agpat2-/- and wild type adipocytes. No differences in PM cholesterol and sphingolipids abundance were detected between genotypes. Levels of phosphatidylserine at day 10 of differentiation were increased in Agpat2-/- adipocytes. Wild type adipocytes had increased whole cell triglyceride, diacylglycerol, phosphatidylglycerol, phosphatidic acid, lysophosphatidylcholine, lysophosphatidylethanolamine, and trihexosyl ceramide, and decreased 24,25-dihydrolanosterol and sitosterol, as a result of adipogenic differentiation. By contrast, adipogenesis did not modify whole cell neutral lipids but increased lysophosphatidylcholine, sphingomyelin, and trihexosyl ceramide levels in Agpat2-/- adipocytes. Unexpectedly, adipogenesis decreased PM levels of main phospholipids in both genotypes. CONCLUSION: In Agpat2-/- adipocytes, decreased caveolae is not associated with changes in PM cholesterol nor sphingolipid levels; however, increased PM phosphatidylserine content may be implicated. Abnormal lipid composition is associated with the adipogenic abnormalities of Agpat2 -/- adipocytes but does not prevent insulin signaling.


Asunto(s)
Aciltransferasas/metabolismo , Adipocitos/metabolismo , Adipogénesis/fisiología , Caveolas/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Esfingolípidos/metabolismo , Animales , Insulina/metabolismo , Metabolismo de los Lípidos/fisiología , Lipidómica/métodos , Lípidos/fisiología , Ratones , Transducción de Señal/fisiología
6.
J Bone Miner Metab ; 39(2): 163-173, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32889573

RESUMEN

INTRODUCTION: The jabuticaba peel extract (JPE) contains bioactive compounds that regulate fat metabolism. Because the negative correlation between fat accumulation and bone formation in bone marrow, we hypothesized that JPE inhibits adipocyte as well as favors osteoblast differentiation of mesenchymal stromal cells (MSCs) under healthy and osteoporotic conditions, a disease that display an imbalance between adipocyte and osteoblast differentiation resulting in reduced bone mass. MATERIAL AND METHODS: To test these hypotheses, bone marrow MSCs were harvested from healthy and osteoporotic rats and cultured in adipogenic and osteogenic media with three concentrations of JPE, 0.25, 5 and 10 µg/ml, and vehicle (control). After selecting the most efficient concentrations of JPE, we used them to evaluate adipocyte and osteoblast differentiation of MSCs from both sources. RESULTS: We observed that, in general, JPE inhibited adipocyte differentiation of MSCs with more pronounced effects in cells from healthy than osteoporotic rats. In addition, JPE increased osteoblast differentiation, exhibiting a slightly higher osteogenic potential on MSCs from osteoporotic compared to healthy condition. CONCLUSION: Our results demonstrated that JPE drives MSCs to inhibit adipocyte differentiation and toward osteoblast differentiation under healthy and osteoporotic conditions. These findings pave the way for further translational studies to investigate the therapeutic possibilities of JPE in both prevention and treatment of osteoporosis.


Asunto(s)
Adipocitos/citología , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteoporosis/patología , Extractos Vegetales/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/fisiología , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Huesos/efectos de los fármacos , Huesos/patología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteoporosis/metabolismo , Ovariectomía , Ratas Wistar
7.
Domest Anim Endocrinol ; 74: 106572, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33039930

RESUMEN

The aim of this study was to differentiate canine adipose-derived mesenchymal stem cells (ADMSCs) into insulin-producing cells by using culture media with different compositions to determine the most efficient media. Stem cells isolated from the fat tissues close to the bitch uterus were distributed into 6 groups: (1) Dulbecco's modified Eagle medium (DMEM)-high glucose (HG), ß-mercaptoethanol, and nicotinamide; (2) DMEM-HG, ß-mercaptoethanol, nicotinamide, and exendin-4; (3) DMEM-HG, ß-mercaptoethanol, nicotinamide, exendin-4, B27, nonessential amino acids, and l-glutamine; (4) DMEM-HG, ß-mercaptoethanol, and nicotinamide (for the initial 8-d period), and DMEM-HG, ß-mercaptoethanol, nicotinamide, exendin-4, B27, nonessential amino acids, l-glutamine, and basic fibroblast growth factor (for the remaining 8-d period); (5) DMEM-HG and fetal bovine serum; and (6) DMEM-low glucose and fetal bovine serum (standard control group). Adipose-derived mesenchymal stem cells from groups 1 to 5 gradually became round in shape and gathered in clusters. These changes differed between the groups. In group 3, the cell clusters were apparently more in numbers and gathered as bigger aggregates. Dithizone staining showed that groups 3 and 4 were similar in terms of the mean area of each aggregate stained for insulin. However, only in group 4, the number of insulin aggregates and the total area of aggregates stained were significantly bigger than in the other groups. The mRNA expression of PDX1, BETA2, MafA, and Insulin were also confirmed in all the groups. We conclude that by manipulating the composition of the culture medium it is possible to induce canine ADMSCs into insulin-producing cells, and the 2-staged protocol that was used promoted the best differentiation.


Asunto(s)
Diferenciación Celular , Medios de Cultivo/farmacología , Insulina/metabolismo , Células Madre Mesenquimatosas/fisiología , Adipogénesis/efectos de los fármacos , Adipogénesis/fisiología , Animales , Carbazoles/química , Carbazoles/farmacología , Condrogénesis/efectos de los fármacos , Condrogénesis/fisiología , Medios de Cultivo/química , Perros , Inmunofenotipificación , Mercaptoetanol/farmacología , Niacinamida/química , Niacinamida/farmacología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología
8.
Front Endocrinol (Lausanne) ; 11: 563816, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123088

RESUMEN

Obesity is a multifactorial and complex condition that is characterized by abnormal and excessive white adipose tissue accumulation, which can lead to the development of metabolic diseases, such as type 2 diabetes mellitus, nonalcoholic fatty liver disease, cardiovascular diseases, and several types of cancer. Obesity is characterized by excessive adipose tissue accumulation and associated with alterations in immunity, displaying a chronic low-grade inflammation profile. Adipose tissue is a dynamic and complex endocrine organ composed not only by adipocytes, but several immunological cells, which can secrete hormones, cytokines and many other factors capable of regulating metabolic homeostasis and several critical biological pathways. Remarkably, adipose tissue is a major source of circulating microRNAs (miRNAs), recently described as a novel form of adipokines. Several adipose tissue-derived miRNAs are deeply associated with adipocytes differentiation and have been identified with an essential role in obesity-associated inflammation, insulin resistance, and tumor microenvironment. During obesity, adipose tissue can completely change the profile of the secreted miRNAs, influencing circulating miRNAs and impacting the development of different pathological conditions, such as obesity, metabolic syndrome, and cancer. In this review, we discuss how miRNAs can act as epigenetic regulators affecting adipogenesis, adipocyte differentiation, lipid metabolism, browning of the white adipose tissue, glucose homeostasis, and insulin resistance, impacting deeply obesity and metabolic diseases. Moreover, we characterize how miRNAs can often act as oncogenic and tumor suppressor molecules, significantly modulating cancer establishment and progression. Furthermore, we highlight in this manuscript how adipose tissue-derived miRNAs can function as important new therapeutic targets.


Asunto(s)
Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Síndrome Metabólico/metabolismo , MicroARNs/metabolismo , Neoplasias/metabolismo , Obesidad/metabolismo , Animales , Humanos , Resistencia a la Insulina/fisiología , Síndrome Metabólico/genética , MicroARNs/genética , Neoplasias/genética , Obesidad/genética , Microambiente Tumoral/fisiología
9.
Biochem Pharmacol ; 182: 114214, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32905795

RESUMEN

Obesity is a worldwide health problem which have reached pandemic proportions, now also including low and middle-income countries. Excessive or abnormal fat deposition in the abdomen especially in the visceral compartment is tightly associated with a high metabolic risk for arterial hypertension, type II diabetes, cardiovascular diseases, musculoskeletal disorders (especially articular degeneration) and some cancers. Contrariwise, accumulation of fat in the subcutaneous compartment has been associated with a neutral metabolic impact, favoring a lower risk of insulin resistance. Obesity results more often from an avoidable imbalance between food consumption and energy expenditure. There are several recommended strategies for dealing with obesity, including pharmacological therapies, but their success remains incomplete and may not compensate the associated adverse effects. Purinergic signaling operated by ATP and its metabolite, adenosine, has attracted increasing attention in obesity. The extracellular levels of purines often reflect the energy status of a given cell population. Adenine nucleotides and nucleosides fine tuning control adipogenesis and mature adipocytes function via the activation of P2 and P1 purinoceptors, respectively. These features make the purinergic signaling cascade a putative target for therapeutic intervention in obesity and related metabolic syndromes. There are, however, gaps in our knowledge regarding the role of purines in adipocyte precursors differentiation and mature adipocytes functions, as well as their impact among distinct adipose tissue deposits (e.g. white vs. brown, visceral vs. subcutaneous), which warrants further investigations before translation to clinical trials can be made.


Asunto(s)
Adipogénesis/fisiología , Obesidad/metabolismo , Purinas/metabolismo , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos/metabolismo , Transducción de Señal/fisiología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Humanos , Obesidad/patología
10.
Biomed Res Int ; 2020: 7453786, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32149131

RESUMEN

Endocrine disruptors (EDs) are defined as environmental pollutants capable of interfering with the functioning of the hormonal system. They are environmentally distributed as synthetic fertilizers, electronic waste, and several food additives that are part of the food chain. They can be considered as obesogenic compounds since they have the capacity to influence cellular events related to adipose tissue, altering lipid metabolism and adipogenesis processes. This review will present the latest scientific evidence of different EDs such as persistent organic pollutants (POPs), heavy metals, "nonpersistent" phenolic compounds, triclosan, polybrominated diphenyl ethers (PBDEs), and smoke-derived compounds (benzo -alpha-pyrene) and their influence on the differentiation processes towards adipocytes in both in vitro and in vivo models.


Asunto(s)
Adipogénesis/fisiología , Disruptores Endocrinos/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Benzoatos/metabolismo , Contaminantes Ambientales/metabolismo , Éteres Difenilos Halogenados/metabolismo , Humanos , Metabolismo de los Lípidos , Fenoles/farmacología , Hidrocarburos Policíclicos Aromáticos/metabolismo , Pirenos/metabolismo , Factores de Transcripción , Triclosán/metabolismo
11.
Int J Mol Sci ; 20(15)2019 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-31357412

RESUMEN

Obesity is considered to significantly increase the risk of the development of a vast range of metabolic diseases. However, adipogenesis is a complex physiological process, necessary to sequester lipids effectively to avoid lipotoxicity in other tissues, like the liver, heart, muscle, essential for maintaining metabolic homeostasis and has a crucial role as a component of the innate immune system, far beyond than only being an inert mass of energy storage. In pathophysiological conditions, adipogenesis promotes a pro-inflammatory state, angiogenesis and the release of adipokines, which become dangerous to health. It results in a hypoxic state, causing oxidative stress and the synthesis and release of harmful free fatty acids. In this review, we try to explain the mechanisms occurring at the breaking point, at which adipogenesis leads to an uncontrolled lipotoxicity. This review highlights the types of adipose tissue and their functions, their way of storing lipids until a critical point, which is associated with hypoxia, inflammation, insulin resistance as well as lipodystrophy and adipogenesis modulation by Krüppel-like factors and miRNAs.


Asunto(s)
Adipogénesis , Tejido Adiposo/metabolismo , Adipocitos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo/citología , Tejido Adiposo/inmunología , Animales , Susceptibilidad a Enfermedades , Metabolismo Energético , Humanos , Lipogénesis , Paniculitis/etiología , Paniculitis/metabolismo , Paniculitis/patología
12.
J Mol Endocrinol ; 63(1): 93-102, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067509

RESUMEN

Neuromedin B, a bombesin-like peptide, and its receptor, are expressed in white adipose tissue with undefined roles. Female mice with disruption of neuromedin B receptor (NB-R) exhibited partial resistance to diet-induced obesity leading to our hypothesis that NB-R is involved in adipogenesis. Here, we showed that adipose stem/stromal cells (ASC) from perigonadal fat of female NB-R-knockout mice, exposed to a differentiation protocol in vitro, accumulated less lipid (45%) than wild type, suggesting reduced capacity to differentiate under adipogenic input. To further explore mechanisms, preadipocytes 3T3-L1 cells were incubated in the presence of NB-R antagonist (PD168368) during the first 3 days in culture. Cells were analyzed in the end of the treatment (Day 3) and later when fully differentiated (Day 21). NB-R antagonist induced lower number of cells at day 3 and 21 (33-39%), reduced cell proliferation at day 3 (-53%) and reduced lipid accumulation at day 21 (-86%). The mRNA expressions of several adipocyte differentiation markers were importantly reduced at both days: Cebpb and Pparg and Fabp4, Plin-1 and Adipoq, and additionally Lep mRNA at day 21. The antagonist had no effect when incubated with mature 3T3-L1 adipocytes. Therefore, genetically disruption of NB-R in mice ASC or pharmacological antagonism of NB-R in 3T3-L1 cells impairs adipogenesis. The mechanisms suggested by results in 3T3-L1 cells involve reduction of cell proliferation and of early gene expressions, leading to decreased number of mature adipocytes. We speculate that NB-R antagonism may be useful to limit the increase in adiposity due to pre-adipocyte differentiation.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/fisiología , Receptores de Bombesina/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipogénesis/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Indoles/farmacología , Ratones , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/metabolismo , Perilipina-1/genética , Perilipina-1/metabolismo , Piridinas/farmacología , Receptores de Bombesina/antagonistas & inhibidores , Receptores de Bombesina/genética
13.
Sci Rep ; 9(1): 5611, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948750

RESUMEN

The adipogenic process is characterized by the expression of adipocyte differentiation markers that lead to changes in cell metabolism and to the accumulation of lipid droplets. Moreover, during early adipogenesis, cells undergo a strong downregulation of translational activity with a decrease in cell size, proliferation and migration. In the present study, we identified that after 24 hours of adipogenic induction, human adipose tissue-derived stem cells (hASCs) undergo a G1-cell cycle arrest consistent with reduced proliferation, and this effect was correlated with a shift in polysome profile with an enrichment of the monosomal fraction and a reduction of the polysomal fraction. Polysome profiling analysis also revealed that this change in the monosomal/polysomal ratio was related to a strong downregulation of cell cycle and proliferation genes, such as cyclins and cyclin-dependent kinases (CDKs). Comparing total and polysome-associated mRNA sequencing, we also observed that this downregulation was mostly due to a reduction of cell cycle and proliferation transcripts via control of total mRNA abundance, rather than by translational control.


Asunto(s)
Adipogénesis/genética , Proteínas de Ciclo Celular/genética , Células Madre Mesenquimatosas/metabolismo , Adipocitos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Adolescente , Adulto , Ciclo Celular , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/genética , Humanos , Gotas Lipídicas/metabolismo , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , ARN Mensajero/genética , Células Madre/citología , Transcriptoma/genética
14.
Orphanet J Rare Dis ; 13(1): 98, 2018 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-29941005

RESUMEN

BACKGROUND: Neurofibromatosis 1 (NF1) presents a wide range of clinical manifestations, including bone alterations. Studies that seek to understand cellular and molecular mechanisms underlying NF1 orthopedic problems are of great importance to better understand the pathogenesis and the development of new therapies. Dental pulp stem cells (DPSCs) are being used as an in vitro model for several diseases and appear as a suitable model for NF1. The aim of this study was to evaluate in vitro chondrogenic differentiation of DPSCs from individuals with NF1 using two-dimensional (2D) and three-dimensional (3D) cultures. RESULTS: To fulfill the criteria of the International Society for Cellular Therapy, DPSCs were characterized by surface antigen expression and by their multipotentiality, being induced to differentiate towards adipogenic, osteogenic, and chondrogenic lineages in 2D cultures. Both DPSCs from individuals with NF1 (NF1 DPSCs) and control cultures were positive for CD90, CD105, CD146 and negative for CD13, CD14, CD45 and CD271, and successfully differentiated after the protocols. Chondrogenic differentiation was evaluated in 2D and in 3D (pellet) cultures, which were further evaluated by optical microscopy and transmission electron microscopy (TEM). 2D cultures showed greater extracellular matrix deposition in NF1 DPSCs comparing with controls during chondrogenic differentiation. In semithin sections, control pellets hadhomogenous-sized intra and extracelullar matrix vesicles, whereas NF1 cultures had matrix vesicles of different sizes. TEM analysis showed higher amount of collagen fibers in NF1 cultures compared with control cultures. CONCLUSION: NF1 DPSCs presented increased extracellular matrix deposition during chondrogenic differentiation, which could be related to skeletal changes in individuals with NF1.


Asunto(s)
Diferenciación Celular/fisiología , Condrogénesis/fisiología , Pulpa Dental/citología , Neurofibromatosis 1/metabolismo , Células Madre/citología , Adipogénesis/fisiología , Adulto , Diferenciación Celular/genética , Células Cultivadas , Condrogénesis/genética , Femenino , Humanos , Masculino , Células Madre/metabolismo , Adulto Joven
15.
Dev Biol ; 441(2): 297-304, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29883659

RESUMEN

Cavefish populations of Astyanax mexicanus have increased body fat compared to surface fish populations of the same species when fed ad libitum in the laboratory. We have previously shown that some cavefish populations display hyperphagia (elevated appetite) to increase food consumption, fat deposition and starvation resistance. However, not all cavefish populations display hyperphagia, yet all previously tested cavefish display elevated body fat levels. Here we have extended this analysis by focusing on visceral fat acquisition in three independently derived cavefish populations. We show that cavefish from two independently derived cavefish populations (Pachón and Tinaja) display increased amounts of visceral adipose tissue (VAT) due to hypertrophy of visceral adipocytes while Molino cavefish display hypertrophy but only slightly elevated VAT levels compared to surface fish. Furthermore, we show that Pachón and Tinaja cavefish develop increased VAT even when food intake is matched to surface fish, suggesting appetite independent mechanisms. We show that in the Pachón population, the differences in the visceral fat in adults correlates with changes in the timing of visceral development, making a developmental contribution likely. Visceral fat development in surface fish starts between 10 and 11 dpf, while in Pachón cavefish, visceral fat cells become visible as early as 8 dpf and develop significantly higher amounts of lipid droplets before surface fish start visceral fat accumulation. We further show that this developmental difference is unique to the Pachón cavefish population, while the Tinaja cavefish population - which displays hyperphagia - starts to develop visceral fat similar to surface fish. We suggest the differences in early adipogenesis in the Pachón population as an additional strategy of increased fat gain in cavefish to adapt to food scarcity.


Asunto(s)
Adaptación Fisiológica , Adipogénesis/fisiología , Characiformes/fisiología , Grasa Intraabdominal/fisiología , Animales , Cuevas
16.
J Cell Physiol ; 233(9): 6853-6865, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29319174

RESUMEN

We evaluated whether genetic predisposition is sufficient to induce changes due to chronic high glucose (HG; 25 mmol/L) in the presence or absence of insulin (HGI; 10 µg/ml) on osteogenic differentiation and markers in bone-marrow mesenchymal stem cells (BMSCs) from young Wistar (WBMSCs) and spontaneous hypertensive rats (SBMSCs) without hypertension. HG suppressed osteogenic differentiation in both the strains, observed by mineralization inhibition and decreased levels of the osteogenic markers Runx2, osterix, osteopontin, and bone sialoprotein, compared to osteogenic medium (OM) cells. In WBMSCs, the effects of HG were associated with the down regulation of ERK1/2 and up regulation of p38 activities; however, HGI did not revert the effects of HG on MAPK activities. Moreover, HG did not affect MAPK signaling in SBMSCs compared to that in OM. HGI increased mineralization in WBMSCs compared to that in OM, but not in SBMSCs. High expression of peroxisome proliferator-activated receptor-gamma and glucose transporter type 4 in OM could be related with the predisposition to adipogenic differentiation noted in SBMSCs and was confirmed by emergence of adipocyte-like cells by HGI treatment. Downregulation of p38 and upregulation of JNK activities were observed in both BMSCs treated with HGI compared to those treated by HG. Ma (osmotic control) also suppressed osteogenic differentiation in both the strains. In conclusion, we demonstrated that SBMSCs from young spontaneous hypertensive rats, without hypertension but with genetic and epigenetic predisposition, exhibited decreased osteoblastic differentiation under HG and HGI did not revert the effects of HG in SBMSCs but increased adipogenic differentiation.


Asunto(s)
Adipogénesis/fisiología , Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Glucosa/metabolismo , Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Regulación hacia Abajo/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Osteoblastos/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Regulación hacia Arriba/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Anim Physiol Anim Nutr (Berl) ; 102(2): 545-556, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29024128

RESUMEN

To determine the effects of maternal nutrition on modifications of foetal development of the skeletal muscle and possible increase in the potential of skeletal muscle growth in cattle, gestating cows were either fed 190% NRC recommendations (overnourished; ON) or 100% NRC recommendation (control; CO). Interaction between maternal nutrition (MN) and the foetal sex (FS) was also investigated. Foetuses were necropsied at four different time points throughout gestation (139, 199, 241 and 268 days of gestation) to assess the mRNA expression of myogenic, adipogenic and fibrogenic markers in skeletal muscle. Phenotypic indicators of the development of skeletal muscle fibres, intramuscular lipogenesis and collagen development were also evaluated. Modifications in mRNA expression of skeletal muscle of foetuses were observed in function of MN and FS despite the lack of effect of MN and FS on foetal weight at necropsy. Maternal ON increased the mRNA expression of the myogenic marker Cadherin-associated protein, beta 1 (CTNNB1) and adipogenic markers Peroxissome proliferator-activated receptor gamma (PPARG) and Zinc finger protein 423 (ZNF423) at midgestation. However, no differences on foetal skeletal muscle development were observed between treatments at late gestation indicating that a compensatory development may have occurred on CO foetuses making the effect of MN on skeletal muscle development not significant at late gestation. Moreover, our data have shown an evidence of sexual dimorphism during foetal stage with a greater skeletal muscle development in male than in female foetuses. In conclusion, providing a higher nutritional level to pregnant cows changes the trajectory of the development of skeletal muscle during midgestation, but apparently does not change the potential of post-natal growth of muscle mass of the offspring, as no differences in skeletal muscle development were observed in late gestation.


Asunto(s)
Alimentación Animal/análisis , Bovinos/fisiología , Desarrollo Fetal/fisiología , Edad Gestacional , Fenómenos Fisiologicos Nutricionales Maternos , Adipogénesis/efectos de los fármacos , Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Biomarcadores , Dieta/veterinaria , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Músculo Esquelético/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores Sexuales
18.
Rev. Bras. Zootec. (Online) ; 47: e20170019, 2018. tab, ilus, graf
Artículo en Inglés | VETINDEX | ID: biblio-1513106

RESUMEN

This study examined the effects of purmorphamine and cyclopamine, classical agonists and inhibitors of the Hedgehog (Hh) signaling pathway, in the adipogenic differentiation of porcine adipose-derived mesenchymal stem cells (AMSC) to investigate the roles underlying adipogenic differentiation in AMSC. Porcine-derived AMSC were established, and the Hh signaling pathway was activated or inhibited by treatment with purmorphamine or cyclopamine. The adipogenic differentiation of the porcine AMSC was then analyzed by Oil Red O staining. The expression levels of Hh signaling pathway factors and adipogenic transcription factors were determined using quantitative real-time polymerase chain reaction and western blot analysis. We verified that the expression levels of positive regulators of the Hh pathway (Smo, Gli1, Gli2, and Gli3) decreased during adipogenesis, whereas those of negative regulators (Ptc1 and Ptc2) increased. Purmorphamine can inhibit the adipogenic differentiation of porcine AMSC in vitro culture. In addition, both the expression of the CCAAT/enhancerbindingprotein-α and that of the peroxisome proliferator-activated receptor-γ decreased in the presence of purmorphamine. By contrast, cyclopamine had no significant effect on the adipogenic differentiation of porcine AMSC. The Hh signaling pathway inhibits the adipogenic differentiation potential of porcine AMSC.(AU)


Asunto(s)
Animales , Porcinos/genética , Alcaloides/efectos adversos , Adipogénesis/fisiología , Proteínas Hedgehog/efectos adversos , Células Madre Mesenquimatosas/fisiología , Tejido Adiposo
19.
Biol Res ; 50(1): 43, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29279050

RESUMEN

BACKGROUND: Understanding the molecular basis underlying the formation of bone-forming osteocytes and lipid-storing adipocytes will help provide insights into the cause of disorders originating in stem/progenitor cells and develop therapeutic treatments for bone- or adipose-related diseases. In this study, the role of RGS2 and RGS4, two members of the regulators of G protein signaling (RGS) family, was investigated during adipogenenic and osteogenenic differentiation of human mesenchymal stem cells (hMSCs). RESULTS: Expression of RGS2 and RGS4 were found to be inversely regulated during adipogenesis induced by dexamethasone (DEX) and 3-isobutyl-methylxanthine, regardless if insulin was present, with RGS2 up-regulated and RGS4 down-regulated in response to adipogenic induction. RGS2 expression was also up-regulated during osteogenesis at a level similar to that induced by treatment of DEX alone, a shared component of adipogenic and osteogenic differentiation inducing media, but significantly lower than the level induced by adipogenic inducing media. RGS4 expression was down-regulated during the first 48 h of osteogenesis but up-regulated afterwards, in both cases at levels similar to that induced by DEX alone. Expression knock-down using small interfering RNA against RGS2 resulted in decreased differentiation efficiency during both adipogenesis and osteogenesis. On the other hand, expression knock-down of RGS4 also resulted in decreased adipogenic differentiation but increased osteogenic differentiation. CONCLUSIONS: RGS2 and RGS4 are differentially regulated during adipogenic and osteogenic differentiation of hMSCs. In addition, both RGS2 and RGS4 play positive roles during adipogenesis but opposing roles during osteogenesis, with RGS2 as a positive regulator and RGS4 as a negative regulator. These results imply that members of RGS proteins may play multifaceted roles during human adipogenesis and osteogenesis to balance or counterbalance each other's function during those processes.


Asunto(s)
Adipogénesis/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Mesenquimatosas/citología , Osteocitos/citología , Osteogénesis/fisiología , Proteínas RGS/metabolismo , Adipogénesis/genética , Regulación de la Expresión Génica/genética , Humanos , Osteogénesis/genética , Proteínas RGS/genética , Factores de Tiempo
20.
Rev. argent. endocrinol. metab ; Rev. argent. endocrinol. metab;54(3): 109-123, set. 2017. ilus
Artículo en Español | LILACS | ID: biblio-957976

RESUMEN

Se describe la relación funcional del metabolismo de las grasas y los hidratos de carbono y su interdependencia, desde los tradicionales conceptos del ciclo glucosa-ácidos grasos (Randle) y la hipótesis portal de la insulinorresistencia hasta los nuevos sobre los adipocitos marrones y beiges, con énfasis en el normal funcionamiento de un patrón endocrino cuya disfunción es clave en la fisiopatología: el eje adipoinsular, vinculado funcionalmente incluso con el hipotálamo, la hipófisis y las adrenales, que involucra 2 hormonas adipogénicas (insulina y glucocorticoide) que facilitarían el desarrollo de la grasa omental perivisceral, con fuertes consecuencias metabólicas. Se discute la ectopia o asiento de grasa en tejido magro por incapacidad del tejido adiposo para seguir atesorando grasas y la actividad endocrina del adipocito, con la producción de moléculas que influyen sobre los mecanismos productores de insulinorresistencia (leptina, adiponectina, TNF-α, resistina, etc.) y disfunción insular. Se describe la disminución de la capacidad oxidativa en la cadena respiratoria mitocondrial y el renacer del concepto de lipogénesis de novo, ambas favorecedoras del atesoramiento de grasas intracelular. En tejidos magros existen pequeñas reservas intracelulares de grasas que mantienen una regulación de funciones esenciales, aunque si aparece una sobrecarga lipídica, el fenómeno conduciría a disfunción (lipotoxicidad) y muerte celular (lipoapoptosis). La tormentosa relación entre las grasas y el islote de Langerhans va más allá del esfuerzo funcional que impone la insulinorresistencia periférica sobre la célula β, por efectos directos de los lípidos o sus derivados sobre la función del islote pancreático. Sin déficit de insulina no hay diabetes.


A review is presented on a functional relationship between fat and carbohydrate metabolism and inter-dependence from the traditional concepts of glucose-fatty acids cycle (Randle), and from the insulin resistance portal hypothesis up to the new aspects on brown and beige adipocytes. Emphasis is placed on the normal function of an endocrine pattern, in which its malfunction is the key in the pathophysiology of these conditions: the adipoinsular axis, with a functional link with the hypothalamic-pituitary-adrenal axis, which involves 2 adipogenic hormones (insulin and glucocorticoid). This has an influence on the development of omental peri-visceral fat, with severe metabolic consequences. A discussion is also presented on the concept of ectopic fat on non-adipose tissues that results in the incapacity of fatty tissue for storing lipids and the considerations about the endocrine activity of adipocyte producing substances that influence several mechanisms that could result in insulin resistance (leptin, adiponectin, TNF-α, resistin, etc.). New aspects are considered regarding the decrease in the oxidative capacity in the mitochondrial respiratory chain, and the rebirth of the concept of de novo lipogenesis that increases the storing of intra-cellular fat. In non-adipose tissues there are small intra-cellular fat quantities for essential functions, but lipid overloading leads to cell dysfunction (lipo-toxicity) and death (lipo-apoptosis). The stormy relationship between fat and Langerhans' Islets goes beyond the functional effort as consequence of peripheral insulin-resistance and the pancreatic beta cell suffers a direct lipid (or derivatives) functional effect. Without insulin deficiency diabetes does not appear.


Asunto(s)
Humanos , Diabetes Mellitus Tipo 2/fisiopatología , Metabolismo Energético/fisiología , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Adipogénesis/fisiología , Metabolismo de los Lípidos , Adipoquinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA