Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33.491
Filtrar
1.
Am J Physiol Cell Physiol ; 326(4): C1248-C1261, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38581663

RESUMEN

Adipose-derived stem cells (ADSCs) play an important role in the differential capacity for excess energy storage between upper body abdominal (ABD) adipose tissue (AT) and lower body gluteofemoral (GF) AT. We cultured ADSCs from subcutaneous ABD AT and GF AT isolated from eight women with differential body fat distribution and performed single-cell RNA sequencing. Six populations of ADSCs were identified and segregated according to their anatomical origin. The three ADSC subpopulations in GF AT were characterized by strong cholesterol/fatty acid (FA) storage and proliferation signatures. The two ABD subpopulations, differentiated by higher expression of committed preadipocyte marker genes, were set apart by differential expression of extracellular matrix and ribosomal genes. The last population, identified in both depots, was similar to smooth muscle cells and when individually isolated and cultured in vitro they differentiated less than the other subpopulations. This work provides important insight into the use of ADSC as an in vitro model of adipogenesis and suggests that specific subpopulations of GF-ADSCs contribute to the more robust capacity for GF-AT to expand and grow compared with ABD-AT in women.NEW & NOTEWORTHY Identification of distinct subpopulations of adipose-derived stem cells (ADSCs) in upper body abdominal subcutaneous (ABD) and lower body gluteofemoral subcutaneous (GF) adipose tissue depots. In ABD-ADSCs, subpopulations are more committed to adipocyte lineage. GF-ADSC subpopulations are enriched for genes involved in lipids and cholesterol metabolism. Similar depot differences were found in stem cell population identified in freshly isolated stoma vascular fraction. The repertoire of ADSCs subpopulations was different in apple-shaped versus pear-shaped women.


Asunto(s)
Tejido Adiposo , Grasa Subcutánea , Humanos , Femenino , Tejido Adiposo/metabolismo , Adipocitos/metabolismo , Análisis de Secuencia de ARN , Colesterol/metabolismo
2.
Front Immunol ; 15: 1360065, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38558823

RESUMEN

Mounting evidence progressively appreciates the vital interplay between immunity and metabolism in a wide array of immunometabolic chronic disorders, both autoimmune and non-autoimmune mediated. The immune system regulates the functioning of cellular metabolism within organs like the brain, pancreas and/or adipose tissue by sensing and adapting to fluctuations in the microenvironment's nutrients, thereby reshaping metabolic pathways that greatly impact a pro- or anti-inflammatory immunophenotype. While it is agreed that the immune system relies on an adequate nutritional status to function properly, we are only just starting to understand how the supply of single or combined nutrients, all of them termed immunonutrients, can steer immune cells towards a less inflamed, tolerogenic immunophenotype. Polyphenols, a class of secondary metabolites abundant in Mediterranean foods, are pharmacologically active natural products with outstanding immunomodulatory actions. Upon binding to a range of receptors highly expressed in immune cells (e.g. AhR, RAR, RLR), they act in immunometabolic pathways through a mitochondria-centered multi-modal approach. First, polyphenols activate nutrient sensing via stress-response pathways, essential for immune responses. Second, they regulate mammalian target of rapamycin (mTOR)/AMP-activated protein kinase (AMPK) balance in immune cells and are well-tolerated caloric restriction mimetics. Third, polyphenols interfere with the assembly of NLR family pyrin domain containing 3 (NLRP3) in endoplasmic reticulum-mitochondria contact sites, inhibiting its activation while improving mitochondrial biogenesis and autophagosome-lysosome fusion. Finally, polyphenols impact chromatin remodeling and coordinates both epigenetic and metabolic reprogramming. This work moves beyond the well-documented antioxidant properties of polyphenols, offering new insights into the multifaceted nature of these compounds. It proposes a mechanistical appraisal on the regulatory pathways through which polyphenols modulate the immune response, thereby alleviating chronic low-grade inflammation. Furthermore, it draws parallels between pharmacological interventions and polyphenol-based immunonutrition in their modes of immunomodulation across a wide spectrum of socioeconomically impactful immunometabolic diseases such as Multiple Sclerosis, Diabetes (type 1 and 2) or even Alzheimer's disease. Lastly, it discusses the existing challenges that thwart the translation of polyphenols-based immunonutritional interventions into long-term clinical studies. Overcoming these limitations will undoubtedly pave the way for improving precision nutrition protocols and provide personalized guidance on tailored polyphenol-based immunonutrition plans.


Asunto(s)
Mitocondrias , Polifenoles , Humanos , Polifenoles/farmacología , Mitocondrias/metabolismo , Sistema Inmunológico/metabolismo , Inflamación/metabolismo , Tejido Adiposo/metabolismo
3.
Gen Comp Endocrinol ; 352: 114516, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38593942

RESUMEN

Cortisol is secreted from the adrenal cortex in response to stress, and its circulating levels are used as robust physiological indicators of stress intensity in various animals. Cortisol is also produced locally in adipose tissue by the conversion of steroid hormones such as cortisone, which is related to fat accumulation. Circulating cortisol levels, probably induced by cold stress, increase in cetaceans under cold conditions. However, whether cortisol production in subcutaneous adipose tissue is enhanced when fat accumulation is renewed during the cold season remains unclear. Therefore, in this study, we examine the effect of environmental temperature on the expression of cortisol synthesis-related enzymes and a glucocorticoid receptor in the subcutaneous fat (blubber) and explore the association between these expressions and fluctuations in circulating cortisol levels in common bottlenose dolphins (Tursiops truncatus). Skin biopsies were obtained seasonally from eight female dolphins, and seasonal differences in the expression of target genes in the blubber were analyzed. Blood samples were collected throughout the year, and cortisol levels were measured. We found that the expressions of cytochrome P450 family 21 subfamily A member 2 (CYP21A2) and nuclear receptor subfamily 3 group C member 1 (NR3C1), a glucocorticoid receptor, were increased in the cold season, and 11 beta-hydroxysteroid dehydrogenase type 1 (HSD11B1) showed a similar trend. Blood cortisol levels increased when the water temperature decreased. These results suggest that the conversion of 17-hydroxyprogesterone to cortisol via 11-deoxycortisol and/or of cortisone to cortisol is enhanced under cold conditions, and the physiological effects of cortisol in subcutaneous adipose tissue may contribute to on-site lipid accumulation and increase the circulating cortisol concentrations. The results obtained in this study highlight the role of cortisol in the regulation of the blubber that has developed to adapt to aquatic life.


Asunto(s)
Delfín Mular , Cortisona , Animales , Femenino , Hidrocortisona/metabolismo , Estaciones del Año , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Cortisona/metabolismo , Tejido Adiposo/metabolismo
4.
Physiol Rep ; 12(7): e15995, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38561245

RESUMEN

Exercise has different effects on different tissues in the body, the sum of which may determine the response to exercise and the health benefits. In the present study, we aimed to investigate whether physical training regulates transcriptional network communites common to both skeletal muscle (SM) and subcutaneous adipose tissue (SAT). Eight such shared transcriptional communities were found in both tissues. Eighteen young overweight adults voluntarily participated in 7 weeks of combined strength and endurance training (five training sessions per week). Biopsies were taken from SM and SAT before and after training. Five of the network communities were regulated by training in SM but showed no change in SAT. One community involved in insulin- AMPK signaling and glucose utilization was upregulated in SM but downregulated in SAT. This diverging exercise regulation was confirmed in two independent studies and was also associated with BMI and diabetes in an independent cohort. Thus, the current finding is consistent with the differential responses of different tissues and suggests that body composition may influence the observed individual whole-body metabolic response to exercise training and help explain the observed attenuated whole-body insulin sensitivity after exercise training, even if it has significant effects on the exercising muscle.


Asunto(s)
Resistencia a la Insulina , Obesidad , Adulto , Humanos , Obesidad/metabolismo , Músculo Esquelético/metabolismo , Ejercicio Físico/fisiología , Grasa Subcutánea/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Expresión Génica , Tejido Adiposo/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-38581357

RESUMEN

Fat is the main component of an adult bone marrow and constitutes the so-called bone marrow adipose tissue (BMAT). Marrow adipocytes, which are the fat cells in the bone marrow, become more abundant with age, and may influence the whole-body metabolism. In osteoporotic patients, the amount of BMAT has an inverse correlation with the amount of bone mass. In people with anorexia nervosa that lose weight after the reduction of peripheral adipose tissues, BMAT expands. Although bone marrow adipocytes are increasingly recognized as a target for therapy, there is still much to learn about their role in skeletal homeostasis, metabolism, cancer, and regenerative treatments. The Bone Marrow Adiposity Society (BMAS), established in 2017, aims to enhance the understanding of how BMAT relates to bone health, cancer, and systemic metabolism. BMAS is committed to training young scientists and organized the second edition of the BMAS Summer School, held on September 4-6, 2023, as a virtual event.


Asunto(s)
Médula Ósea , Neoplasias , Humanos , Médula Ósea/metabolismo , Adiposidad , Tejido Adiposo/metabolismo , Instituciones Académicas , Neoplasias/metabolismo
6.
Mol Med Rep ; 29(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38606791

RESUMEN

Obesity reaches up to epidemic proportions globally and increases the risk for a wide spectrum of co­morbidities, including type­2 diabetes mellitus (T2DM), hypertension, dyslipidemia, cardiovascular diseases, non­alcoholic fatty liver disease, kidney diseases, respiratory disorders, sleep apnea, musculoskeletal disorders and osteoarthritis, subfertility, psychosocial problems and certain types of cancers. The underlying inflammatory mechanisms interconnecting obesity with metabolic dysfunction are not completely understood. Increased adiposity promotes pro­inflammatory polarization of macrophages toward the M1 phenotype, in adipose tissue (AT), with subsequent increased production of pro­inflammatory cytokines and adipokines, inducing therefore an overall, systemic, low­grade inflammation, which contributes to metabolic syndrome (MetS), insulin resistance (IR) and T2DM. Targeting inflammatory mediators could be alternative therapies to treat obesity, but their safety and efficacy remains to be studied further and confirmed in future clinical trials. The present review highlights the molecular and pathophysiological mechanisms by which the chronic low­grade inflammation in AT and the production of reactive oxygen species lead to MetS, IR and T2DM. In addition, focus is given on the role of anti­inflammatory agents, in the resolution of chronic inflammation, through the blockade of chemotactic factors, such as monocytes chemotractant protein­1, and/or the blockade of pro­inflammatory mediators, such as IL­1ß, TNF­α, visfatin, and plasminogen activator inhibitor­1, and/or the increased synthesis of adipokines, such as adiponectin and apelin, in obesity­associated metabolic dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Síndrome Metabólico , Humanos , Obesidad/metabolismo , Síndrome Metabólico/complicaciones , Síndrome Metabólico/metabolismo , Inflamación/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Mediadores de Inflamación/metabolismo
7.
Front Immunol ; 15: 1381227, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38638434

RESUMEN

Obesity presents a significant global health challenge, increasing the susceptibility to chronic conditions such as diabetes, cardiovascular disease, and hypertension. Within the context of obesity, lipid metabolism, adipose tissue formation, and inflammation are intricately linked to endoplasmic reticulum stress (ERS). ERS modulates metabolism, insulin signaling, inflammation, as well as cell proliferation and death through the unfolded protein response (UPR) pathway. Serving as a crucial nexus, ERS bridges the functionality of adipose tissue and the inflammatory response. In this review, we comprehensively elucidate the mechanisms by which ERS impacts adipose tissue function and inflammation in obesity, aiming to offer insights into targeting ERS for ameliorating metabolic dysregulation in obesity-associated chronic diseases such as hyperlipidemia, hypertension, fatty liver, and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hipertensión , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Inflamación/metabolismo
8.
Sci Rep ; 14(1): 9465, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658613

RESUMEN

A poor nutritional status is associated with worse pulmonary function and survival in people with cystic fibrosis (pwCF). CF transmembrane conductance regulator modulators can improve pulmonary function and body weight, but more data is needed to evaluate its effects on body composition. In this retrospective study, a pre-trained deep-learning network was used to perform a fully automated body composition analysis on chest CTs from 66 adult pwCF before and after receiving elexacaftor/tezacaftor/ivacaftor (ETI) therapy. Muscle and adipose tissues were quantified and divided by bone volume to obtain body size-adjusted ratios. After receiving ETI therapy, marked increases were observed in all adipose tissue ratios among pwCF, including the total adipose tissue ratio (+ 46.21%, p < 0.001). In contrast, only small, but statistically significant increases of the muscle ratio were measured in the overall study population (+ 1.63%, p = 0.008). Study participants who were initially categorized as underweight experienced more pronounced effects on total adipose tissue ratio (p = 0.002), while gains in muscle ratio were equally distributed across BMI categories (p = 0.832). Our findings suggest that ETI therapy primarily affects adipose tissues, not muscle tissue, in adults with CF. These effects are primarily observed among pwCF who were initially underweight. Our findings may have implications for the future nutritional management of pwCF.


Asunto(s)
Aminofenoles , Benzodioxoles , Composición Corporal , Fibrosis Quística , Combinación de Medicamentos , Indoles , Quinolinas , Quinolonas , Humanos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/fisiopatología , Masculino , Adulto , Femenino , Composición Corporal/efectos de los fármacos , Aminofenoles/uso terapéutico , Quinolonas/uso terapéutico , Benzodioxoles/uso terapéutico , Estudios Retrospectivos , Indoles/uso terapéutico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Tomografía Computarizada por Rayos X , Adulto Joven , Pirrolidinas/uso terapéutico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Tejido Adiposo/diagnóstico por imagen , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Estado Nutricional
9.
Stem Cell Res Ther ; 15(1): 119, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38659070

RESUMEN

BACKGROUND: Adipose stromal cells (ASC) are a form of mesenchymal stromal cells that elicit effects primarily via secreted factors, which may have advantages for the treatment of injury or disease. Several previous studies have demonstrated a protective role for MSC/ASC on mitigating acute kidney injury but whether ASC derived factors could hasten recovery from established injury has not been evaluated. METHODS: We generated a concentrated secretome (CS) of human ASC under well-defined conditions and evaluated its ability to improve the recovery of renal function in a preclinical model of acute kidney injury (AKI) in rats. 24 h following bilateral ischemia/reperfusion (I/R), rats were randomized following determination of plasma creatinine into groups receiving vehicle -control or ASC-CS treatment by subcutaneous injection (2 mg protein/kg) and monitored for evaluation of renal function, structure and inflammation. RESULTS: Renal function, assessed by plasma creatinine levels, recovered faster in ASC-CS treated rats vs vehicle. The most prominent difference between the ASC-CS treated vs vehicle was observed in rats with the most severe degree of initial injury (Pcr > 3.0 mg/dl 24 h post I/R), whereas rats with less severe injury (Pcr < 2.9 mg/dl) recovered quickly regardless of treatment. The quicker recovery of ASC-treated rats with severe injury was associated with less tissue damage, inflammation, and lower plasma angiopoietin 2. In vitro, ASC-CS attenuated the activation of the Th17 phenotype in lymphocytes isolated from injured kidneys. CONCLUSIONS: Taken together, these data suggest that ASC-CS represents a potent therapeutic option to improve established AKI.


Asunto(s)
Lesión Renal Aguda , Inflamación , Lesión Renal Aguda/terapia , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Ratas , Humanos , Inflamación/patología , Inflamación/metabolismo , Masculino , Secretoma/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Ratas Sprague-Dawley , Inyecciones Subcutáneas , Riñón/metabolismo , Riñón/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/terapia , Células del Estroma/metabolismo
10.
Immun Inflamm Dis ; 12(4): e1241, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38629728

RESUMEN

BACKGROUND: Inflammation in adipose tissue, resulting from imbalanced caloric intake and energy expenditure, contributes to the metabolic dysregulation observed in obesity. The production of inflammatory cytokines, such as IL-1ß and IL-18, plays a key role in this process. While IL-1ß promotes insulin resistance and diabetes, IL-18 regulates energy expenditure and food intake. Previous studies have suggested that caspase-1, activated by the Nlrp3 inflammasome in response to lipid excess, mediates IL-1ß production, whereas activated by the Nlrp1b inflammasome in response to energy excess, mediates IL-18 production. However, this has not been formally tested. METHODS: Wild-type and caspase-1-deficient Balb/c mice, carrying the Nlrp1b1 allele, were fed with regular chow or a high-fat diet for twelve weeks. Food intake and mass gain were recorded weekly. At the end of the twelve weeks, glucose tolerance and insulin resistance were evaluated. Mature IL-18 protein levels and the inflammatory process in the adipose tissue were determined. Fasting lipid and cytokine levels were quantified in the sera of the different experimental groups. RESULTS: We found that IL-18 production in adipose tissue is independent of caspase-1 activity, regardless of the metabolic state, while Nlrp3-mediated IL-1ß production remains caspase-1 dependent. Additionally, caspase-1 null Balb/c mice did not develop metabolic abnormalities in response to energy excess from the high-fat diet. CONCLUSION: Our findings suggest that IL-18 production in the adipose tissue is independent of Nlrp3 inflammasome and caspase-1 activation, regardless of caloric food intake. In contrast, Nlrp3-mediated IL-1ß production is caspase-1 dependent. These results provide new insights into the mechanisms underlying cytokine production in the adipose tissue during both homeostatic conditions and metabolic stress, highlighting the distinct roles of caspase-1 and the Nlrp inflammasomes in regulating inflammatory responses.


Asunto(s)
Resistencia a la Insulina , Proteína con Dominio Pirina 3 de la Familia NLR , Ratones , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Interleucina-18 , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasas/metabolismo , Tejido Adiposo/metabolismo , Citocinas/metabolismo , Lípidos
11.
FASEB J ; 38(8): e23613, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38661048

RESUMEN

The unpredictable survival rate of autologous fat grafting (AFG) seriously affects its clinical application. Improving the survival rate of AFG has become an unresolved issue in plastic surgery. Peroxisome proliferator-activated receptor-γ (PPAR-γ) regulates the adipogenic differentiation of adipocytes, but the functional mechanism in AFG remains unclear. In this study, we established an animal model of AFG and demonstrated the superior therapeutic effect of PPAR-γ regulation in the process of AFG. From day 3 after fat grafting, the PPAR-γ agonist rosiglitazone group consistently showed better adipose integrity, fewer oil cysts, and fibrosis. Massive macrophage infiltration was observed after 7 days. At the same time, M2 macrophages begin to appear. At day 14, M2 macrophages gradually became the dominant cell population, which suppressed inflammation and promoted revascularization and fat regeneration. In addition, transcriptome sequencing showed that the differentially expressed genes in the Rosiglitazone group were associated with the pathways of adipose regeneration, differentiation, and angiogenesis; these results provide new ideas for clinical treatment.


Asunto(s)
Tejido Adiposo , Macrófagos , PPAR gamma , Rosiglitazona , Trasplante Autólogo , Animales , PPAR gamma/metabolismo , PPAR gamma/genética , Macrófagos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/citología , Rosiglitazona/farmacología , Masculino , Diferenciación Celular , Adipogénesis , Adipocitos/metabolismo , Ratones , Ratas
12.
Sci Rep ; 14(1): 7195, 2024 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-38532033

RESUMEN

Patients with type 1 diabetes (T1D) have a greater risk of cardiovascular disease. Proconvertase subtilisin-kexin 9 (PCSK9) is involved in the atherosclerosis process. This study aimed to determine the relationship between PCSK9 levels and epicardial adipose tissue (EAT) volume and cardiometabolic variables in patients with T1D. This was an observational cross-sectional study including 73 patients with T1D. Clinical, biochemical and imaging data were collected. We divided the patients into two groups according to their glycemic control and the EAT index (iEAT) percentile. We performed a correlation analysis between the collected variables and PCSK9 levels; subsequently, we performed a multiple regression analysis with the significant parameters. The mean age was 47.6 ± 8.5 years, 58.9% were men, and the BMI was 26.9 ± 4.6 kg/m2. A total of 31.5%, 49.3% and 34.2% of patients had hypertension, dyslipidemia and smoking habit, respectively. The PCSK9 concentration was 0.37 ± 0.12 mg/L, which was greater in patients with worse glycemic control (HbA1c > 7.5%), dyslipidemia and high EAT volume (iEAT > 75th percentile). The PCSK9 concentration was positively correlated with age (r = 0.259; p = 0.027), HbA1c (r = 0.300; p = 0.011), insulin dose (r = 0.275; p = 0.020), VLDL-C level (r = 0.331; p = 0.004), TG level (r = 0.328; p = 0.005), and iEAT (r = 0.438; p < 0.001). Multiple regression analysis revealed that 25% of the PCSK9 variability was explained by iEAT and HbA1c (p < 0.05). The PCSK9 concentration is associated with metabolic syndrome parameters, poor glycemic control and increased EAT volume in patients with T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Dislipidemias , Masculino , Humanos , Adulto , Persona de Mediana Edad , Femenino , Diabetes Mellitus Tipo 1/metabolismo , Proproteína Convertasa 9/metabolismo , 60428 , Hemoglobina Glucada , Subtilisina , Estudios Transversales , Tejido Adiposo/metabolismo
13.
Cells ; 13(6)2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38534357

RESUMEN

The development of obesity is associated with substantial modulation of adipose tissue (AT) structure. The plasticity of the AT is reflected by its remarkable ability to expand or reduce in size throughout the adult lifespan, which is linked to the development of its vasculature. This increase in AT vasculature could be mediated by the differentiation of adipose tissue-derived stem cells (ASCs) into endothelial cells (ECs) and form new microvasculature. We have already shown that microRNA (miRNA)-145 regulates the differentiation of ASCs into EC-like (ECL) cells. Here, we investigated whether ASCs-differentiation into ECs is governed by a miRNAs signature that depends on fat depot location and /or the metabolic condition produced by obesity. Human ASCs, which were obtained from white AT by surgical procedures from lean and obese patients, were induced to differentiate into ECL cells. We have identified that miRNA-29b-3p in both subcutaneous (s)ASCs and visceral ASCs and miRNA-424-5p and miRNA-378a-3p in subcutaneous (s)ASCs are involved in differentiation into EC-like cells. These miRNAs modulate their pro-angiogenic effects on ASCs by targeting FGFR1, NRP2, MAPK1, and TGF-ß2, and the MAPK signaling pathway. We show for the first time that miRNA-29b-3p upregulation contributes to ASCs' differentiation into ECL cells by directly targeting TGFB2 in both sASCs and visceral ASCs. Moreover, our results reveal that, independent of sASCs' origin (obese/lean), the upregulation of miRNA-378a-3p and the downregulation of miRNA-424-5p inhibit MAPK1 and overexpress FGFR1 and NRP2, respectively. In summary, both the adipose depot location and obesity affect the differentiation of resident ASCs through the expression of specific miRNAs.


Asunto(s)
Células Madre Mesenquimatosas , MicroARNs , Adulto , Humanos , MicroARNs/genética , Células Endoteliales/metabolismo , Tejido Adiposo/metabolismo , Células Madre Mesenquimatosas/metabolismo , Obesidad/metabolismo
14.
Mol Biol Rep ; 51(1): 451, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38536507

RESUMEN

BACKGROUND: Mitochondrial organelles play a crucial role in cellular metabolism so different cell types exhibit diverse metabolic and energy demands. Therefore, alternations in the intracellular distribution, quantity, function, and structure of mitochondria are required for stem cell differentiation. Finding an effective inducer capable of modulating mitochondrial activity is critical for the differentiation of specific stem cells into osteo-like cells for addressing issues related to osteogenic disorders. This study aimed to investigate the effect of oxaloacetate (OAA) on the osteogenic differentiation of human adipose-derived mesenchymal stem cells (hADSCs) in vitro. METHODS AND RESULTS: First, the most favorable OAA concentration was measured through MTT assay and subsequently confirmed using acridine orange staining. Human ADSCs were cultured in osteogenic medium supplemented with OAA and analyzed on days 7 and 14 of differentiation. Various assays including alkaline phosphatase assay (ALP), cellular calcium content assay, mineralized matrix staining with alizarin red, catalase (CAT) and superoxide dismutase (SOD) activity, and real-time RT-PCR analysis of three bone-specific markers (ALP, osteocalcin, and collagen type I) were conducted to characterize the differentiated cells. Following viability assessment, OAA at a concentration of 1 µM was considered the optimal dosage for further studies. The results of osteogenic differentiation assays showed that OAA at a concentration of 1 × 10- 6 M significantly increased ALP enzyme activity, mineralization, CAT and SOD activity and the expression of bone-specific genes in differentiated cells compared to control groups in vitro. CONCLUSIONS: In conclusion, the fundings from this study suggest that OAA possesses favorable properties that make it a potential candidate for application in medical bone regeneration.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Humanos , Tejido Adiposo/metabolismo , Ácido Oxaloacético/metabolismo , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Superóxido Dismutasa/metabolismo , Células Cultivadas
15.
Physiol Rep ; 12(6): e15957, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38546216

RESUMEN

Epicardial adipose tissue (EAT) is an active endocrine organ that is closely associated with occurrence of atrial fibrillation (AF). However, the role of EAT in the development of postoperative AF (POAF) remains unclear. We aimed to investigate the association between EAT profile and POAF occurrence in patients who underwent cardiovascular surgery. We obtained EAT samples from 53 patients to evaluate gene expression, histological changes, mitochondrial oxidative phosphorylation (OXPHOS) capacity in the EAT, and protein secretion in EAT-conditioned medium. EAT volume was measured using computed tomography scan. Eighteen patients (34%) experienced POAF within 7 days after surgery. Although no significant difference was observed in EAT profile between patients with and without POAF, logistic regression analysis identified that the mRNA expression levels of tumor necrosis factor-alpha (TNF-α) were positively correlated and adipocyte size in the EAT was inversely correlated with onset of POAF, respectively. Mitochondrial OXPHOS capacity in the EAT was not associated with POAF occurrence; however, it showed an inverse correlation with adipocyte size and a positive correlation with adiponectin secretion. In conclusion, changes in the secretory profile and adipocyte morphology of the EAT, which represent qualitative aspects of the adipose tissue, were present before the onset of AF.


Asunto(s)
Fibrilación Atrial , Humanos , Fibrilación Atrial/metabolismo , 60428 , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Inflamación/metabolismo , Pericardio/metabolismo
16.
BMC Endocr Disord ; 24(1): 39, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38481206

RESUMEN

BACKGROUND: A better understanding of adipose tissue (AT) dysfunction, which includes morphological and functional changes such as adipocyte hypertrophy as well as impaired adipogenesis, lipid storage/mobilization, endocrine and inflammatory responses, is needed in the context of obesity. One dimension of AT dysfunction, secretory adiposopathy, often assessed as a low plasma adiponectin (A)/leptin (L) ratio, is commonly observed in obesity. The aim of this study was to examine markers of AT development and metabolism in 67 women of varying age and adiposity (age: 40-62 years; body mass index, BMI: 17-41 kg/m2) according to levels of adiponectinemia, leptinemia or the plasma A/L ratio. METHODS: Body composition, regional AT distribution and circulating adipokines were determined. Lipolysis was measured from glycerol release in subcutaneous abdominal (SCABD) and omental (OME) adipocytes under basal, isoproterenol-, forskolin (FSK)- and dibutyryl-cyclic AMP (DcAMP)-stimulated conditions. Adipogenesis (C/EBP-α/ß/δ, PPAR-γ2 and SREBP-1c) and lipid metabolism (ß2-ARs, HSL, FABP4, LPL and GLUT4) gene expression (RT-qPCR) was assessed in both fat depots. Participants in the upper versus lower tertile of adiponectin, leptin or the A/L ratio were compared. RESULTS: Basal lipolysis was similar between groups. Women with a low plasma A/L ratio were characterized by higher adiposity and larger SCABD and OME adipocytes (p<0.01) compared to those with a high ratio. In OME adipocytes, women in the low adiponectinemia tertile showed higher isoproterenol-stimulated lipolysis (0.01

Asunto(s)
Adiponectina , Leptina , Femenino , Humanos , Adulto , Persona de Mediana Edad , Adiponectina/metabolismo , Leptina/metabolismo , Isoproterenol/metabolismo , Tejido Adiposo/metabolismo , Obesidad/metabolismo
17.
J Oleo Sci ; 73(4): 411-418, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38556276

RESUMEN

In 2021, we published three papers related to the anti-inflammatory effects of food ingredients. The present paper reports the effects of vitamin E homologs and sweet basil powder. In these papers, we investigated whether inflammation occurs in the adipose tissue of mice fed a high-fat and high-sucrose diet for 16 weeks. Inflammatory cytokine gene expression was significantly higher in the epididymal fat of the high-fat and high-sucrose diet group than in that of the control diet group. However, the addition of α-tocopherol or δ-tocopherol to the diet could not restrain the inflammation of mice epididymal fats. Thereafter, we investigated the anti-inflammatory effects of α- and δ-tocopherols using the co-cultured cells. Consequently, we clarified that δ-tocopherol inhibited the increase in the gene expressions of inflammatory cytokines. We also examined the effect of sweet basil powder on a similar obese mice model. The final body weight in the high-fat and high-sucrose group that received sweet basil powder was significantly lower than that in the high-fat and high-sucrose diet group. Liver weights were also significantly lower in the high-fat and high-sucrose diet group that received sweet basil powder than in the high-fat and high-sucrose diet group, although adipose tissue weights were unchanged in both groups. Furthermore, sweet basil powder tended to inhibit in lipid synthesis in the mice livers. Therefore, we suggested that sweet basil powder inhibited fatty acid synthesis in mice livers, thereby suppressing liver enlargement, and resulting in body weight loss. Moreover, the gene expression of MCP-1 in the adipose tissue of mice fed a high-fat and high-sucrose diet added with sweet basil powder was significantly lower than that of mice fed a high-fat and high-sucrose diet for 12 weeks. Therefore, sweet basil powder inhibited inflammation onset in the adipose tissue of mice. Taken together, the results suggested that food ingredients, especially vitamin E homologs and sweet basil powder, have anti-inflammatory effects on mice adipose tissue and mice adipocyte-induced inflammation.


Asunto(s)
Ingredientes Alimentarios , Ratones , Animales , Polvos , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Inflamación/genética , Citocinas/metabolismo , Sacarosa , Vitamina E/farmacología , Vitamina E/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL
18.
Biomolecules ; 14(3)2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38540669

RESUMEN

Browning of white adipose tissue (WAT) is a focus of research in type 2 diabetes mellitus (T2DM) and metabolism, which may be a potential molecular mechanism for high-intensity interval training (HIIT) to improve T2DM. In this study, male C57BL/6J wild-type mice were subjected to an 8-week HIIT regimen following T2DM induction through a high-fat diet (HFD) combined with streptozotocin (STZ) injection. We found that HIIT improved glucose metabolism, body weight, and fat mass in T2DM mice. HIIT also decreased adipocyte size and induced browning of WAT. Our data revealed a decrease in TNFα and an increase in IL-10 with HIIT, although the expression of chemokines MCP-1 and CXCL14 was increased. We observed increased pan-macrophage infiltration induced by HIIT, along with a simultaneous decrease in the expression of M1 macrophage markers (iNOS and CD11c) and an increase in M2 macrophage markers (Arg1 and CD206), suggesting that HIIT promotes M2 macrophage polarization. Additionally, HIIT upregulated the expression of Slit3 and neurotrophic factors (BDNF and NGF). The expression of the sympathetic marker tyrosine hydroxylase (TH) and the nerve growth marker GAP43 was also increased, demonstrating the promotion of sympathetic nerve growth and density by HIIT. Notably, we observed macrophages co-localizing with TH, and HIIT induced the accumulation of M2 macrophages around sympathetic nerves, suggesting a potential association between M2 macrophages and increased density of sympathetic nerves. In conclusion, HIIT induces adipose tissue browning and improves glucose metabolism in T2DM mice by enhancing M2 macrophage polarization and promoting sympathetic nerve growth and density.


Asunto(s)
Diabetes Mellitus Tipo 2 , Entrenamiento de Intervalos de Alta Intensidad , Masculino , Animales , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Ratones Endogámicos C57BL , Tejido Adiposo/metabolismo , Macrófagos/metabolismo , Tejido Adiposo Blanco/metabolismo , Glucosa/metabolismo , Proteínas de la Membrana/metabolismo
19.
Cell Rep ; 43(3): 113955, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38507414

RESUMEN

Epicardial adipose tissue (eAT) is a metabolically active fat depot that has been associated with a wide array of cardiac homeostatic functions and cardiometabolic diseases. A full understanding of its diverse physiological and pathological roles is hindered by the dearth of animal models. Here, we show, in the heart of an ectothermic teleost, the zebrafish, the existence of a fat depot localized underneath the epicardium, originating from the epicardium and exhibiting the molecular signature of beige adipocytes. Moreover, a subset of adipocytes within this cardiac fat tissue exhibits primitive thermogenic potential. Transcriptomic profiling and cross-species analysis revealed elevated glycolytic and cardiac homeostatic gene expression with downregulated obesity and inflammatory hallmarks in the teleost eAT compared to that of lean aged humans. Our findings unveil epicardium-derived beige fat in the heart of an ectotherm considered to possess solely white adipocytes for energy storage and identify pathways that may underlie age-driven remodeling of human eAT.


Asunto(s)
Tejido Adiposo Beige , Pez Cebra , Animales , Humanos , Anciano , Tejido Adiposo Beige/metabolismo , 60428 , Tejido Adiposo/metabolismo , Pericardio/metabolismo , Termogénesis , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo
20.
FASEB J ; 38(7): e23574, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38551804

RESUMEN

Aldo-keto reductase 1C3 (AKR1C3) is a key enzyme in the activation of both classic and 11-oxygenated androgens. In adipose tissue, AKR1C3 is co-expressed with 11ß-hydroxysteroid dehydrogenase type 1 (HSD11B1), which catalyzes not only the local activation of glucocorticoids but also the inactivation of 11-oxygenated androgens, and thus has the potential to counteract AKR1C3. Using a combination of in vitro assays and in silico modeling we show that HSD11B1 attenuates the biosynthesis of the potent 11-oxygenated androgen, 11-ketotestosterone (11KT), by AKR1C3. Employing ex vivo incubations of human female adipose tissue samples we show that inhibition of HSD11B1 results in the increased peripheral biosynthesis of 11KT. Moreover, circulating 11KT increased 2-3 fold in individuals with type 2 diabetes after receiving the selective oral HSD11B1 inhibitor AZD4017 for 35 days, thus confirming that HSD11B1 inhibition results in systemic increases in 11KT concentrations. Our findings show that HSD11B1 protects against excess 11KT production by adipose tissue, a finding of particular significance when considering the evidence for adverse metabolic effects of androgens in women. Therefore, when targeting glucocorticoid activation by HSD11B1 inhibitor treatment in women, the consequently increased generation of 11KT may offset beneficial effects of decreased glucocorticoid activation.


Asunto(s)
Andrógenos , Diabetes Mellitus Tipo 2 , Humanos , Femenino , Andrógenos/metabolismo , Glucocorticoides , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1 , Tejido Adiposo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA