Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.504
Filtrer
1.
Ageing Res Rev ; 99: 102402, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38977081

RÉSUMÉ

Alzheimer's disease (AD) is a leading cause of cognitive impairment worldwide. Overweight and obesity are strongly associated with comorbidities, such as hypertension, diabetes, and insulin resistance (IR), which contribute substantially to the development of AD and subsequent morbidity and mortality. Adipose tissue (AT) is a highly dynamic organ composed of a diverse array of cell types, which can be classified based on their anatomic localization or cellular composition. The expansion and remodeling of AT in the context of obesity involves immunometabolic and functional shifts steered by the intertwined actions of multiple immune cells and cytokine signaling within AT, which contribute to the development of metabolic disorders, IR, and systemic markers of chronic low-grade inflammation. Chronic low-grade inflammation, a prolonged, low-dose stimulation by specific immunogens that can progress from localized sites and affect multiple organs throughout the body, leads to neurodystrophy, increased apoptosis, and disruption of homeostasis, manifesting as brain atrophy and AD-related pathology. In this review, we sought to elucidate the mechanisms by which AT contributes to the onset and progression of AD in obesity through the mediation of chronic low-grade inflammation, particularly focusing on the roles of adipokines and AT-resident immune cells.


Sujet(s)
Tissu adipeux , Maladie d'Alzheimer , Inflammation , Obésité , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/étiologie , Obésité/métabolisme , Obésité/immunologie , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Tissu adipeux/immunologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Animaux , Adipokines/métabolisme
2.
Food Funct ; 15(14): 7658-7668, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38953736

RÉSUMÉ

Obesity is often accompanied by low-grade chronic inflammation and metabolic syndrome. It has been established that microbiota influences many physiological processes, including the development of obesity, and dysbiosis has been observed in obese individuals. In this study, we aimed to evaluate the impact of a new probiotic formulation, containing two probiotic strains and the bioactive compound octacosanol, on body weight, metabolic parameters, and concentrations of certain adipocytokines and appetite-regulating hormones in obese women. This double blind placebo-controlled supplementary intervention study included twenty-five women in the intervention group and twenty-three in the placebo group, and it lasted 12 weeks. Daily oral supplementation included 7 × 1010 CFU of Lactiplantibacillus plantarum 299v (DSM9843), 5 × 109 CFU of Saccharomyces cerevisiae var. boulardii (DBVPG6763), and 40 mg of octacosanol or placebo. Body weight, metabolic parameters, adipocytokines, and appetite-regulating hormones were assessed before (T0) and after the intervention (T1). After the intervention, significantly lower median concentrations of CRP (p = 0.005) and IL-6 (p = 0.012) were measured in the intervention group than the baseline, while the median concentrations of ghrelin (p = 0.026) and HDL-cholesterol (p = 0.03) were significantly increased. The intervention group had lower CRP levels (p = 0.023) and higher ghrelin levels (p = 0.006) than the placebo group. Significant changes in BMI between groups were not observed. In summary, although the new probiotic formulation showed beneficial effects on IL-6, CRP, HDL, and ghrelin levels, its potential effects on regulating triglyceride, insulin, and glucose levels require further studies before the novel dietary intervention could be considered a useful adjuvant therapy and an effective strategy for the management of obesity and obesity-associated comorbidities.


Sujet(s)
Adipokines , Obésité , Probiotiques , Humains , Femelle , Probiotiques/pharmacologie , Probiotiques/usage thérapeutique , Obésité/diétothérapie , Obésité/métabolisme , Méthode en double aveugle , Adulte , Adipokines/sang , Adipokines/métabolisme , Adulte d'âge moyen , Ghréline/sang , Appétit/effets des médicaments et des substances chimiques , Lactobacillus plantarum , Poids/effets des médicaments et des substances chimiques , Protéine C-réactive/métabolisme
3.
Expert Opin Ther Targets ; 28(7): 587-600, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38965991

RÉSUMÉ

BACKGROUND: Psoriasis is a chronic immune-mediated skin condition with several types of manifestation, including psoriatic arthritis. In recent years, studies have demonstrated multiple molecules and mechanisms that play important roles in the pathophysiology of psoriasis. Studies have been conducted to determine the role of adipokines, bioactive peptides secreted by the adipose tissue, in the pathogenesis of inflammatory diseases. These studies have shown that adipokines are dysregulated in psoriasis and their abnormal expression profile could contribute to the inflammatory mechanisms observed in psoriasis. AREAS COVERED: In this review, we discuss the immunomodulatory features of resistin, omentin-1, and vaspin, and discuss their potential involvement in the pathogenesis of psoriasis. EXPERT OPINION: The adipokines resistin, omentin, and vaspin appear to be promising therapeutic targets in psoriasis. It is important to seek to block the action of resistin, either by blocking its receptors or by blocking its systemic effects with antibodies. In the case of omentin and vaspin, substances that are receptor mimetics of these adipokines should be sought and studies conducted of their analogues for the treatment of psoriasis. To introduce these therapies into clinical practice, multicentre clinical trials are required to confirm their efficacy and safety after initial studies in animal models.


Sujet(s)
Cytokines , Protéines liées au GPI , Lectines , Psoriasis , Résistine , Serpines , Humains , Psoriasis/traitement médicamenteux , Protéines liées au GPI/métabolisme , Serpines/pharmacologie , Serpines/métabolisme , Animaux , Cytokines/métabolisme , Résistine/métabolisme , Adipokines/métabolisme
4.
Clin Interv Aging ; 19: 1259-1272, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011312

RÉSUMÉ

Postmenopausal osteoporosis (PMOP) is a major health problem affecting millions of women worldwide. PMOP patients are often accompanied by abnormal accumulation of bone marrow adipose tissue (BMAT). BMAT is a critical regulator of bone homeostasis, and an increasing BMAT volume is negatively associated with bone mass reduction or fracture. BMAT regulates bone metabolism via adipokines, cytokines and the immune system, but the specific mechanisms are largely unknown. This review emphasizes the impact of estrogen deficiency on bone homeostasis and BMAT expansion, and the mechanism by which BMAT regulates PMOP, providing a promising strategy for targeting BMAT in preventing and treating PMOP.


Sujet(s)
Tissu adipeux , Moelle osseuse , Ostéoporose post-ménopausique , Humains , Tissu adipeux/métabolisme , Femelle , Densité osseuse , Adipokines/métabolisme , Oestrogènes/métabolisme , Os et tissu osseux/métabolisme , Animaux , Cytokines/métabolisme , Homéostasie
5.
Reprod Fertil Dev ; 362024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39074236

RÉSUMÉ

Adipose tissues produce a variety of biologically active compounds, including cytokines, growth factors and adipokines. Adipokines are important as they function as endocrine hormones that are related to various metabolic and reproductive diseases. The goal of this review was to summarise the role of asprosin, a recently discovered adipokine, and compare its role in ovarian steroidogenesis with that of other adipokines including adiponectin, leptin, resistin, apelin, visfatin, chemerin, irisin, and gremlin 1. The summary of concentrations of these adipokines in humans, rats and other animals will help researchers identify appropriate doses to test in future studies. Review of the literature indicated that asprosin increases androstenedione production in theca cells (Tc), and when cotreated with FSH increases oestradiol production in granulosa cells (Gc). In comparison, other adipokines (1) stimulate Gc oestradiol production but inhibit Tc androgen production (adiponectin), (2) inhibit Gc oestradiol production and Tc androstenedione production (leptin and chemerin), (3) inhibit Gc steroidogenesis with no effect on Tc (resistin), (4) inhibit Gc oestradiol production but stimulate Tc androgen production (gremlin 1), and (5) increase steroid secretion by Gc, with unknown effects on Tc steroidogenesis (apelin and visfatin). Irisin has direct effects on Gc but its precise role (inhibitory or stimulatory) may be species dependent and its effects on Tc will require additional research. Thus, most adipokines have direct effects (either positive or negative) on steroid production in ovarian cells, but how they all work together to create a cumulative effect or disease will require further research.


Sujet(s)
Adipokines , Cellules de la granulosa , Cellules thécales , Femelle , Humains , Adipokines/métabolisme , Animaux , Cellules thécales/métabolisme , Cellules thécales/effets des médicaments et des substances chimiques , Cellules de la granulosa/métabolisme , Cellules de la granulosa/effets des médicaments et des substances chimiques
6.
Nutrients ; 16(14)2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39064727

RÉSUMÉ

Gymnema sylvestre (GS) and berberine (BBR) are natural products that have demonstrated therapeutic potential for the management of obesity and its comorbidities, as effective and safe alternatives to synthetic drugs. Although their anti-obesogenic and antidiabetic properties have been widely studied, comparative research on their impact on the gene expression of adipokines, such as resistin (Res), omentin (Ome), visfatin (Vis) and apelin (Ap), has not been reported. METHODOLOGY: We performed a comparative study in 50 adult Mexican patients with obesity treated with GS or BBR for 3 months. The baseline and final biochemical parameters, body composition, blood pressure, gene expression of Res, Ome, Vis, and Ap, and safety parameters were evaluated. RESULTS: BBR significantly decreased (p < 0.05) body weight, blood pressure and Vis and Ap gene expression and increased Ome, while GS decreased fasting glucose and Res gene expression (p < 0.05). A comparative analysis of the final measurements revealed a lower gene expression of Ap and Vis (p < 0.05) in patients treated with BBR than in those treated with GS. The most frequent adverse effects in both groups were gastrointestinal symptoms, which attenuated during the first month of treatment. CONCLUSION: In patients with obesity, BBR has a better effect on body composition, blood pressure, and the gene expression of adipokines related to metabolic risk, while GS has a better effect on fasting glucose and adipokines related to insulin resistance, with minimal side effects.


Sujet(s)
Adipokines , Berbérine , Composition corporelle , Gymnema sylvestre , Obésité , Résistine , Humains , Mâle , Femelle , Adulte , Obésité/traitement médicamenteux , Obésité/métabolisme , Adipokines/sang , Adipokines/métabolisme , Composition corporelle/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Berbérine/pharmacologie , Résistine/sang , Résistine/métabolisme , Apeline , Pression sanguine/effets des médicaments et des substances chimiques , Nicotinamide phosphoribosyltransferase/métabolisme , Cytokines/métabolisme , Cytokines/sang , Extraits de plantes/pharmacologie , Glycémie/effets des médicaments et des substances chimiques , Glycémie/métabolisme , Lectines , Protéines liées au GPI/métabolisme , Protéines liées au GPI/génétique , Agents antiobésité/pharmacologie , Agents antiobésité/usage thérapeutique
7.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892118

RÉSUMÉ

The connection between body weight alterations and Alzheimer's disease highlights the intricate relationship between the brain and adipose tissue in the context of neurological disorders. During midlife, weight gain increases the risk of cognitive decline and dementia, whereas in late life, weight gain becomes a protective factor. Despite their substantial impact on metabolism, the role of adipokines in the transition from healthy aging to neurological disorders remains largely unexplored. We aim to investigate how the adipose tissue milieu and the secreted adipokines are involved in the transition between biological and pathological aging, highlighting the bidirectional relationship between the brain and systemic metabolism. Understanding the function of these adipokines will allow us to identify biomarkers for early detection of Alzheimer's disease and uncover novel therapeutic options.


Sujet(s)
Adipokines , Tissu adipeux , Maladie d'Alzheimer , Encéphale , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Humains , Adipokines/métabolisme , Tissu adipeux/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Animaux , Marqueurs biologiques , Vieillissement/métabolisme
8.
Curr Med Sci ; 44(3): 463-474, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38900388

RÉSUMÉ

Excessive fat deposition in obese subjects promotes the occurrence of metabolic diseases, such as type 2 diabetes mellitus (T2DM), cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Adipose tissue is not only the main form of energy storage but also an endocrine organ that not only secretes adipocytokines but also releases many extracellular vesicles (EVs) that play a role in the regulation of whole-body metabolism. Exosomes are a subtype of EVs, and accumulating evidence indicates that adipose tissue exosomes (AT Exos) mediate crosstalk between adipose tissue and multiple organs by being transferred to targeted cells or tissues through paracrine or endocrine mechanisms. However, the roles of AT Exos in crosstalk with metabolic organs remain to be fully elucidated. In this review, we summarize the latest research progress on the role of AT Exos in the regulation of metabolic disorders. Moreover, we discuss the potential role of AT Exos as biomarkers in metabolic diseases and their clinical application.


Sujet(s)
Tissu adipeux , Exosomes , Maladies métaboliques , Exosomes/métabolisme , Humains , Tissu adipeux/métabolisme , Maladies métaboliques/métabolisme , Diabète de type 2/métabolisme , Marqueurs biologiques/métabolisme , Animaux , Stéatose hépatique non alcoolique/métabolisme , Obésité/métabolisme , Adipokines/métabolisme , Maladie chronique
10.
Life Sci ; 352: 122866, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38936605

RÉSUMÉ

Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.


Sujet(s)
Tissu adipeux , Évolution de la maladie , Insuffisance rénale chronique , Système rénine-angiotensine , Humains , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Système rénine-angiotensine/physiologie , Rein/métabolisme , Rein/anatomopathologie , Animaux , Adipokines/métabolisme
11.
Lipids Health Dis ; 23(1): 192, 2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38909257

RÉSUMÉ

BACKGROUND: Overweight, often known as obesity, is the abnormal and excessive accumulation of fat that exposes the health of a person at risk by increasing the likelihood that they may experience many chronic conditions. Consequently, obesity has become a global health threat, presenting serious health issues, and attracting a lot of attention in the healthcare profession and the scientific community. METHOD: This study aims to explore the anti-adipogenic properties of 7-MEGA™ in an attempt to address obesity, using both in vitro and in vivo research. The effects of 7MEGA™ at three distinct concentrations were investigated in obese mice who were given a high-fat diet (HFD) and 3T3-L1 adipocytes. RESULTS: 7MEGA™ decreased the total fat mass, overall body weight, and the perirenal and subcutaneous white adipose tissue (PWAT and SWAT) contents in HFD mice. Additionally, 7MEGA™ showed promise in improving the metabolic health of individuals with obesity and regulate the levels of insulin hormone, pro-inflammatory cytokines and adipokines. Furthermore, Peroxisome proliferator-activated receptors (PPAR) α and γ, Uncoupling Protein 1 (UCP-1), Sterol Regulatory Element-Binding Protein 1 (SREBP-1), Fatty Acid-Binding Protein 4 (FABP4), Fatty Acid Synthase (FAS), Acetyl-CoA Carboxylase (ACC), Stearoyl-CoA Desaturase-1 (SCD-1) and CCAAT/Enhancer-Binding Protein (C/EBPα) were among the adipogenic regulators that 7MEGA™ could regulate. CONCLUSION: In summary, this study uncovered that 7MEGA™ demonstrates anti-adipogenic and anti-obesity effects, suggesting its potential in combating obesity.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Alimentation riche en graisse , Souris de lignée C57BL , Obésité , Animaux , Alimentation riche en graisse/effets indésirables , Adipogenèse/effets des médicaments et des substances chimiques , Obésité/métabolisme , Souris , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Mâle , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Acyl-(acyl-carrier-protein)desaturase/métabolisme , Acyl-(acyl-carrier-protein)desaturase/génétique , Souris obèse , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Adipokines/métabolisme , Agents antiobésité/pharmacologie , Protéine-1 de découplage/métabolisme , Protéine-1 de découplage/génétique , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Protéines liant les séquences stimulatrices de type CCAAT
12.
Pharmacol Res ; 205: 107219, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38763327

RÉSUMÉ

Adipokines are a heterogeneous group of signalling molecules secreted prevalently by adipose tissue. Initially considered as regulators of energy metabolism and appetite, adipokines have been recognized for their substantial involvement in musculoskeletal disorders, including osteoarthritis, rheumatoid arthritis, and many others. Understanding the role of adipokines in rheumatic inflammatory and autoimmune diseases, as well as in other musculoskeletal diseases such as intervertebral disc degeneration, is crucial for the development of novel therapeutic strategies. Targeting adipokines, or their signalling pathways, may offer new opportunities for the treatment and management of these conditions. By modulating adipokines levels or activity, it may be possible to regulate inflammation, to maintain bone health, and preserve muscle mass, thereby improving the outcomes and quality of life for individuals affected by musculoskeletal diseases. The aim of this review article is to update the reader on the multifaceted role of adipokines in the main rheumatic diseases such as osteoarthritis and rheumatoid arthritis and to unravel the complex interplay among adipokines, cartilage metabolism, bone remodelling and muscles, which will pave the way for innovative therapeutic intervention in the future. For completeness, the role of adipokines in intervertebral disc degeneration will be also addressed.


Sujet(s)
Adipokines , Polyarthrite rhumatoïde , Dégénérescence de disque intervertébral , Arthrose , Humains , Adipokines/métabolisme , Adipokines/immunologie , Dégénérescence de disque intervertébral/traitement médicamenteux , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/immunologie , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Arthrose/immunologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/immunologie , Animaux , Rhumatismes/traitement médicamenteux , Rhumatismes/immunologie , Rhumatismes/métabolisme
13.
Life Sci ; 349: 122735, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38768776

RÉSUMÉ

AIMS: Imeglimin, a novel antidiabetic drug, has recently been reported to affect pancreatic ß-cells and hepatocytes. Adipose tissue plays a crucial role in systemic metabolism. However, its effect on adipocytes remains unexplored. Herein, we investigated the effects of imeglimin on adipocytes, particularly in the mitochondria. MAIN METHODS: The 3T3-L1 adipocytes were treated with imeglimin. Mitochondrial respiratory complex I activity and NAD+, NADH, and AMP levels were measured. Protein expression levels were determined by western blotting, mitochondrial DNA and mRNA expression levels were determined using quantitative polymerase chain reaction, and secreted adipocytokine and mitokine levels were determined using adipokine array and enzyme-linked immunosorbent assay. KEY FINDINGS: Imeglimin inhibited complex I activity, decreased the NAD+/NADH ratio, and increased AMP levels, which were associated with the enhanced phosphorylation of AMP-activated protein kinase. In addition, imeglimin increased the mitochondrial DNA content and levels of mitochondrial transcription factor A and peroxisome proliferator-activated receptor-γ coactivator 1-α mRNA, which were abolished by Ly294002, a phosphoinositide 3-kinase inhibitor. Furthermore, imeglimin facilitated the expression levels of markers of the mitochondrial unfolded protein response, and the gene expression and secretion of two mitokines, fibroblast growth factor 21 and growth differentiation factor 15. The production of both mitokines was transcriptionally regulated and abolished by phosphoinositide 3-kinase and Akt inhibitors. SIGNIFICANCE: Imeglimin modulates mitochondrial biology in adipocytes and may exert a mitohormetic effect through mitokine secretion.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Mitochondries , Animaux , Souris , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Adipokines/métabolisme , AMP-Activated Protein Kinases/métabolisme , Facteurs de croissance fibroblastique
14.
Mol Biol Rep ; 51(1): 631, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38722405

RÉSUMÉ

Adipokines are now well-known to regulate reproduction. Visfatin is an adipokine expressed in the hypothalamus, pituitary, ovary, uterus, and placenta of different species, and since it has been found to modulate the endocrine secretion of the hypothalamus, pituitary gland and ovary, it may be considered a novel regulator of female reproduction. Although the majority of the literature explored its role in ovarian regulation, visfatin has also been shown to regulate uterine remodeling, endometrial receptivity and embryo development, and its expression in the uterus is steroid dependent. Like other adipokines, visfatin expression and levels are deregulated in pathological conditions including polycystic ovary syndrome. Thus, the present mini-review focuses on the role of visfatin in female reproduction under both physiological and pathological conditions.


Sujet(s)
Nicotinamide phosphoribosyltransferase , Syndrome des ovaires polykystiques , Reproduction , Femelle , Humains , Nicotinamide phosphoribosyltransferase/métabolisme , Nicotinamide phosphoribosyltransferase/génétique , Reproduction/physiologie , Reproduction/génétique , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/physiopathologie , Animaux , Ovaire/métabolisme , Utérus/métabolisme , Cytokines/métabolisme , Grossesse , Adipokines/métabolisme
15.
Brain Behav Immun ; 120: 34-43, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38772428

RÉSUMÉ

BACKGROUND: Increased adiposity during pregnancy may be related to offspring risk for mental health disorders, although the biological mechanisms are poorly understood. One promising hypothesis is that factors secreted from adipocytes such as leptin and adiponectin may explain this association. The current study examined whether pregnancy or umbilical cord blood concentrations of leptin and/or adiponectin a) predict elevated infant negative affect at 6 months (an early life marker of risk for psychopathology); and b) help explain the association between pregnancy adiposity and increased infant negative affect. METHODS: Data came from a prospective cohort (N = 305) of pregnant individuals and their offspring. Second trimester adiposity was assessed using air displacement plethysmography. Concentrations of leptin and adiponectin were measured in second trimester plasma and umbilical cord plasma. Infant negative affect was assessed by standardized observation at 6 months. Second trimester inflammation was assessed using a comprehensive panel of cytokines. RESULTS: Lower second trimester adiponectin was associated with elevated infant negative affect, and mediated the effect of pregnancy adiposity on infant negative affect. This association was independent of the effect of second trimester inflammation. Umbilical cord leptin also predicted higher infant negative affect and mediated the association between pregnancy adiposity and infant negative affect. CONCLUSIONS: This is the first study to link pregnancy adiponectin or cord blood leptin to infant markers of risk for psychopathology, and the first to demonstrate that these adipokines mediate the association between pregnancy adiposity and offspring behavioral outcomes, suggesting novel markers of risk and potential mechanisms of effect.


Sujet(s)
Adipokines , Adiponectine , Adiposité , Affect , Sang foetal , Leptine , Deuxième trimestre de grossesse , Humains , Femelle , Grossesse , Sang foetal/métabolisme , Leptine/sang , Adulte , Adiponectine/sang , Deuxième trimestre de grossesse/sang , Adipokines/sang , Adipokines/métabolisme , Adiposité/physiologie , Études prospectives , Affect/physiologie , Nourrisson , Mâle , Nouveau-né , Marqueurs biologiques/sang , Inflammation/sang , Inflammation/métabolisme
16.
Int J Mol Sci ; 25(10)2024 May 11.
Article de Anglais | MEDLINE | ID: mdl-38791266

RÉSUMÉ

Sympathetic nervous system (SNS) hyperactivity is mediated by elevated catecholamine (CA) secretion from the adrenal medulla, as well as enhanced norepinephrine (NE) release from peripheral sympathetic nerve terminals. Adrenal CA production from chromaffin cells is tightly regulated by sympatho-inhibitory α2-adrenergic (auto)receptors (ARs), which inhibit both epinephrine (Epi) and NE secretion via coupling to Gi/o proteins. α2-AR function is, in turn, regulated by G protein-coupled receptor (GPCR)-kinases (GRKs), especially GRK2, which phosphorylate and desensitize them, i.e., uncouple them from G proteins. On the other hand, the short-chain free fatty acid (SCFA) receptor (FFAR)-3, also known as GPR41, promotes NE release from sympathetic neurons via the Gi/o-derived free Gßγ-activated phospholipase C (PLC)-ß/Ca2+ signaling pathway. However, whether it exerts a similar effect in adrenal chromaffin cells is not known at present. In the present study, we examined the interplay of the sympatho-inhibitory α2A-AR and the sympatho-stimulatory FFAR3 in the regulation of CA secretion from rat adrenal chromaffin (pheochromocytoma) PC12 cells. We show that FFAR3 promotes CA secretion, similarly to what GRK2-dependent α2A-AR desensitization does. In addition, FFAR3 activation enhances the effect of the physiologic stimulus (acetylcholine) on CA secretion. Importantly, GRK2 blockade to restore α2A-AR function or the ketone body beta-hydroxybutyrate (BHB or 3-hydroxybutyrate), via FFAR3 antagonism, partially suppress CA production, when applied individually. When combined, however, CA secretion from PC12 cells is profoundly suppressed. Finally, propionate-activated FFAR3 induces leptin and adiponectin secretion from PC12 cells, two important adipokines known to be involved in tissue inflammation, and this effect of FFAR3 is fully blocked by the ketone BHB. In conclusion, SCFAs can promote CA and adipokine secretion from adrenal chromaffin cells via FFAR3 activation, but the metabolite/ketone body BHB can effectively inhibit this action.


Sujet(s)
Catécholamines , Récepteurs alpha-2 adrénergiques , Récepteurs couplés aux protéines G , Animaux , Cellules PC12 , Rats , Récepteurs couplés aux protéines G/métabolisme , Catécholamines/métabolisme , Récepteurs alpha-2 adrénergiques/métabolisme , Adipokines/métabolisme , Cellules chromaffines/métabolisme , Transduction du signal , Norépinéphrine/métabolisme , Norépinéphrine/pharmacologie
17.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38731880

RÉSUMÉ

Adipose tissue is a multifunctional organ that regulates many physiological processes such as energy homeostasis, nutrition, the regulation of insulin sensitivity, body temperature, and immune response. In this review, we highlight the relevance of the different mediators that control adipose tissue activity through a systematic review of the main players present in white and brown adipose tissues. Among them, inflammatory mediators secreted by the adipose tissue, such as classical adipokines and more recent ones, elements of the immune system infiltrated into the adipose tissue (certain cell types and interleukins), as well as the role of intestinal microbiota and derived metabolites, have been reviewed. Furthermore, anti-obesity mediators that promote the activation of beige adipose tissue, e.g., myokines, thyroid hormones, amino acids, and both long and micro RNAs, are exhaustively examined. Finally, we also analyze therapeutic strategies based on those mediators that have been described to date. In conclusion, novel regulators of obesity, such as microRNAs or microbiota, are being characterized and are promising tools to treat obesity in the future.


Sujet(s)
Tissu adipeux , Obésité , Humains , Animaux , Obésité/métabolisme , Tissu adipeux/métabolisme , Adipokines/métabolisme , microARN/métabolisme , microARN/génétique , Microbiome gastro-intestinal , Tissu adipeux brun/métabolisme , Tissu adipeux blanc/métabolisme , Médiateurs de l'inflammation/métabolisme , Métabolisme énergétique
18.
Adv Clin Chem ; 121: 172-269, 2024.
Article de Anglais | MEDLINE | ID: mdl-38797542

RÉSUMÉ

Reproductive success consists of a sequential events chronology, starting with the ovum fertilization, implantation of the embryo, placentation, and cellular processes like proliferation, apoptosis, angiogenesis, endocrinology, or metabolic changes, which taken together finally conduct the birth of healthy offspring. Currently, many factors are known that affect the regulation and proper maintenance of pregnancy in humans, domestic animals, or rodents. Among the determinants of reproductive success should be distinguished: the maternal microenvironment, genes, and proteins as well as numerous pregnancy hormones that regulate the most important processes and ensure organism homeostasis. It is well known that white adipose tissue, as the largest endocrine gland in our body, participates in the synthesis and secretion of numerous hormones belonging to the adipokine family, which also may regulate the course of pregnancy. Unfortunately, overweight and obesity lead to the expansion of adipose tissue in the body, and its excess in both women and animals contributes to changes in the synthesis and release of adipokines, which in turn translates into dramatic changes during pregnancy, including those taking place in the organ that is crucial for the proper progress of pregnancy, i.e. the placenta. In this chapter, we are summarizing the current knowledge about levels of adipokines and their role in the placenta, taking into account the physiological and pathological conditions of pregnancy, e.g. gestational diabetes mellitus, preeclampsia, or intrauterine growth restriction in humans, domestic animals, and rodents.


Sujet(s)
Adipokines , Grossesse , Humains , Adipokines/métabolisme , Femelle , Animaux , Placenta/métabolisme , Diabète gestationnel/métabolisme
19.
Reprod Domest Anim ; 59(5): e14624, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38798196

RÉSUMÉ

The study aimed to assess the local gene expression of adipokine members, namely vaspin, adiponectin, visfatin, resistin and their associated receptors - heat shock 70 protein 5 (HSPA5), adiponectin receptor 1 (AdipoR1) and adiponectin receptor 2 (AdipoR2) - in bovine follicles during the preovulatory period and early corpus luteum development. Follicles were collected before gonadotropin-releasing hormone (GnRH) treatment (0 h) and at 4, 10, 20, 25 and 60 h after GnRH application through transvaginal ovariectomy (n = 5 samples/group). Relative mRNA expression levels were quantified using real-time reverse transcription polymerase chain reaction (RT-qPCR). Vaspin exhibited high mRNA levels immediately 4 h after GnRH application, followed by a significant decrease. Adiponectin mRNA levels were elevated at 25 h after GnRH treatment. AdipoR2 exhibited late-stage upregulation, displaying increased expression at 20, 25 and 60 h following GnRH application. Visfatin showed upregulation at 20 h post-GnRH application. In conclusion, the observed changes in adipokine family members within preovulatory follicles, following experimentally induced ovulation, may constitute crucial components of the local mechanisms regulating final follicle growth and development.


Sujet(s)
Adipokines , Corps jaune , Hormone de libération des gonadotrophines , Follicule ovarique , Ovulation , Animaux , Femelle , Bovins/physiologie , Corps jaune/métabolisme , Corps jaune/effets des médicaments et des substances chimiques , Follicule ovarique/effets des médicaments et des substances chimiques , Follicule ovarique/métabolisme , Ovulation/physiologie , Hormone de libération des gonadotrophines/pharmacologie , Hormone de libération des gonadotrophines/métabolisme , Adipokines/métabolisme , Adipokines/génétique , ARN messager/métabolisme , ARN messager/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Nicotinamide phosphoribosyltransferase/génétique , Nicotinamide phosphoribosyltransferase/métabolisme
20.
Reprod Biol Endocrinol ; 22(1): 38, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38575956

RÉSUMÉ

The present study aimed to examine the effects of progranulin and omentin on basic ovarian cell functions. For this purpose, we investigated the effects of the addition of progranulin and omentin (0, 0.1, 1, or 10 ng/ml) on the viability, proliferation, apoptosis and steroidogenesis of cultured rabbit ovarian granulosa cells. To determine the importance of the interrelationships between granulosa cells and theca cells, we compared the influence of progranulin and omentin on progesterone and estradiol release in cultured granulosa cells and ovarian fragments containing both granulosa cells and theca cells. Cell viability, proliferation, cytoplasmic apoptosis and release of progesterone and estradiol were measured by Cell Counting Kit-8 (CCK-8), BrdU incorporation, cell death detection, and ELISA. Both progranulin and omentin increased granulosa cell viability and proliferation and decreased apoptosis. Progranulin increased progesterone release by granulosa cells but reduced progesterone output by ovarian fragments. Progranulin decreased estradiol release by granulosa cells but increased it in ovarian fragments. Omentin reduced progesterone release in both models. Omentin reduced estradiol release by granulosa cells but promoted this release in ovarian fragments. The present observations are the first to demonstrate that progranulin and omentin can be direct regulators of basic ovarian cell functions. Furthermore, the differences in the effects of these adipokines on steroidogenesis via granulosa and ovarian fragments indicate that these peptides could target both granulosa and theca cells.


Sujet(s)
Adipokines , Progestérone , Femelle , Animaux , Lapins , Progestérone/métabolisme , Progranulines/métabolisme , Progranulines/pharmacologie , Adipokines/métabolisme , Adipokines/pharmacologie , Ovaire/métabolisme , Cellules de la granulosa/métabolisme , Oestradiol/métabolisme , Apoptose , Cellules cultivées , Prolifération cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE