Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.534
Filtrer
1.
Sci Rep ; 14(1): 13209, 2024 06 08.
Article de Anglais | MEDLINE | ID: mdl-38851835

RÉSUMÉ

Hypertension remains a leading cause of cardiovascular and kidney diseases. Failure to control blood pressure with ≥ 3 medications or control requiring ≥ 4 medications is classified as resistant hypertension (rHTN) and new therapies are needed to reduce the resulting increased risk of morbidity and mortality. Here, we report genetic evidence that relaxin family peptide receptor 2 (RXFP2) is associated with rHTN in men, but not in women. This study shows that adrenal gland gene expression of RXFP2 is increased in men with hypertension and the RXFP2 natural ligand, INSL3, increases adrenal steroidogenesis and corticosteroid secretion in human adrenal cells. To address the hypothesis that RXFP2 activation is an important mechanism in rHTN, we discovered and characterized small molecule and monoclonal antibody (mAb) blockers of RXFP2. The novel chemical entities and mAbs show potent, selective inhibition of RXFP2 and reduce aldosterone and cortisol synthesis and release. The RXFP2 mAbs have suitable rat pharmacokinetic profiles to evaluate the role of RXFP2 in the development and maintenance of rHTN. Overall, we identified RXFP2 activity as a potential new mechanism in rHTN and discovered RXFP2 antagonists for the future interrogation of RXFP2 in cardiovascular and renal diseases.


Sujet(s)
Hypertension artérielle , Récepteurs couplés aux protéines G , Récepteurs peptidiques , Humains , Mâle , Hypertension artérielle/traitement médicamenteux , Hypertension artérielle/génétique , Récepteurs couplés aux protéines G/antagonistes et inhibiteurs , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Animaux , Récepteurs peptidiques/génétique , Récepteurs peptidiques/métabolisme , Récepteurs peptidiques/antagonistes et inhibiteurs , Rats , Femelle , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Glandes surrénales/métabolisme , Glandes surrénales/effets des médicaments et des substances chimiques , Résistance aux substances/génétique , Antihypertenseurs/pharmacologie , Aldostérone/métabolisme
2.
Int J Mol Sci ; 25(10)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38791102

RÉSUMÉ

Congenital Adrenal Hyperplasia (CAH) is an autosomal recessive disorder impairing cortisol synthesis due to reduced enzymatic activity. This leads to persistent adrenocortical overstimulation and the accumulation of precursors before the blocked enzymatic step. The predominant form of CAH arises from mutations in CYP21A2, causing 21-hydroxylase deficiency (21-OHD). Despite emerging treatment options for CAH, it is not always possible to physiologically replace cortisol levels and counteract hyperandrogenism. Moreover, there is a notable absence of an effective in vivo model for pre-clinical testing. In this work, we developed an animal model for CAH with the clinically relevant point mutation p.R484Q in the previously humanized CYP21A2 mouse strain. Mutant mice showed hyperplastic adrenals and exhibited reduced levels of corticosterone and 11-deoxycorticosterone and an increase in progesterone. Female mutants presented with higher aldosterone concentrations, but blood pressure remained similar between wildtype and mutant mice in both sexes. Male mutant mice have normal fertility with a typical testicular appearance, whereas female mutants are infertile, exhibit an abnormal ovarian structure, and remain in a consistent diestrus phase. Conclusively, we show that the animal model has the potential to contribute to testing new treatment options and to prevent comorbidities that result from hormone-related derangements and treatment-related side effects in CAH patients.


Sujet(s)
Hyperplasie congénitale des surrénales , Modèles animaux de maladie humaine , Steroid 21-hydroxylase , Animaux , Hyperplasie congénitale des surrénales/génétique , Hyperplasie congénitale des surrénales/anatomopathologie , Hyperplasie congénitale des surrénales/métabolisme , Steroid 21-hydroxylase/génétique , Steroid 21-hydroxylase/métabolisme , Souris , Femelle , Mâle , Humains , Corticostérone/métabolisme , Corticostérone/sang , Aldostérone/métabolisme , Glandes surrénales/métabolisme , Glandes surrénales/anatomopathologie , Mutation , Progestérone/métabolisme
3.
J Neuroimmunol ; 391: 578366, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38733741

RÉSUMÉ

Disturbance in neuroendocrine signaling has been consistently documented in multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS) representing the main cause of non-traumatic brain injury among young adults. In fact, MS patients display altered hormonal levels and psychiatric symptoms along with the pathologic hallmarks of the disease, which include demyelination, neuroinflammation and axonal injury. In addition, we have recently shown that extensive transcriptional changes take place in the hypothalamus of mice upon the MS model experimental autoimmune encephalomyelitis (EAE). We also detected structural and functional aberrancies in endocrine glands of EAE animals. Specifically, we described the hyperplasia of adrenal glands and the atrophy of ovaries at disease peak. To further expand the characterization of these phenotypes, here we profiled the transcriptomes of both glands by means of RNA-seq technology. Notably, we identified fatty acid and cholesterol biosynthetic pathways as the most dysregulated molecular processes in adrenals and ovaries, respectively. Furthermore, we demonstrated that key genes encoding neuropeptides and hormone receptors undergo distinct expression dynamics in the hypothalamus along disease progression. Altogether, our results corroborate the dysfunction of the neuroendocrine system as a major pathological event of autoimmune demyelination and highlight the crosstalk between the CNS and the periphery in mediating such disease phenotypes.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Métabolisme lipidique , Souris de lignée C57BL , Animaux , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Femelle , Souris , Métabolisme lipidique/physiologie , Ovaire/métabolisme , Ovaire/anatomopathologie , Glandes surrénales/métabolisme , Glandes surrénales/anatomopathologie , Transcriptome
4.
Cell Metab ; 36(6): 1411-1429.e10, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38701776

RÉSUMÉ

Mitochondria have diverse functions critical to whole-body metabolic homeostasis. Endurance training alters mitochondrial activity, but systematic characterization of these adaptations is lacking. Here, the Molecular Transducers of Physical Activity Consortium mapped the temporal, multi-omic changes in mitochondrial analytes across 19 tissues in male and female rats trained for 1, 2, 4, or 8 weeks. Training elicited substantial changes in the adrenal gland, brown adipose, colon, heart, and skeletal muscle. The colon showed non-linear response dynamics, whereas mitochondrial pathways were downregulated in brown adipose and adrenal tissues. Protein acetylation increased in the liver, with a shift in lipid metabolism, whereas oxidative proteins increased in striated muscles. Exercise-upregulated networks were downregulated in human diabetes and cirrhosis. Knockdown of the central network protein 17-beta-hydroxysteroid dehydrogenase 10 (HSD17B10) elevated oxygen consumption, indicative of metabolic stress. We provide a multi-omic, multi-tissue, temporal atlas of the mitochondrial response to exercise training and identify candidates linked to mitochondrial dysfunction.


Sujet(s)
Mitochondries , Conditionnement physique d'animal , Animaux , Mâle , Femelle , Mitochondries/métabolisme , Rats , Muscles squelettiques/métabolisme , Humains , Rat Sprague-Dawley , Tissu adipeux brun/métabolisme , Glandes surrénales/métabolisme , Multi-omique
5.
Zool Res ; 45(3): 617-632, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38766745

RÉSUMÉ

The Chinese tree shrew ( Tupaia belangeri chinensis) has emerged as a promising model for investigating adrenal steroid synthesis, but it is unclear whether the same cells produce steroid hormones and whether their production is regulated in the same way as in humans. Here, we comprehensively mapped the cell types and pathways of steroid metabolism in the adrenal gland of Chinese tree shrews using single-cell RNA sequencing, spatial transcriptome analysis, mass spectrometry, and immunohistochemistry. We compared the transcriptomes of various adrenal cell types across tree shrews, humans, macaques, and mice. Results showed that tree shrew adrenal glands expressed many of the same key enzymes for steroid synthesis as humans, including CYP11B2, CYP11B1, CYB5A, and CHGA. Biochemical analysis confirmed the production of aldosterone, cortisol, and dehydroepiandrosterone but not dehydroepiandrosterone sulfate in the tree shrew adrenal glands. Furthermore, genes in adrenal cell types in tree shrews were correlated with genetic risk factors for polycystic ovary syndrome, primary aldosteronism, hypertension, and related disorders in humans based on genome-wide association studies. Overall, this study suggests that the adrenal glands of Chinese tree shrews may consist of closely related cell populations with functional similarity to those of the human adrenal gland. Our comprehensive results (publicly available at http://gxmujyzmolab.cn:16245/scAGMap/) should facilitate the advancement of this animal model for the investigation of adrenal gland disorders.


Sujet(s)
Glandes surrénales , Stéroïdes , Animaux , Glandes surrénales/métabolisme , Humains , Stéroïdes/biosynthèse , Stéroïdes/métabolisme , Transcriptome , Souris , Tupaiidae , Femelle , Multi-omique
6.
Hypertension ; 81(7): 1500-1510, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38747164

RÉSUMÉ

BACKGROUND: Inter-individual variation in blood pressure (BP) arises in part from sequence variants within enhancers modulating the expression of causal genes. We propose that these genes, active in tissues relevant to BP physiology, can be identified from tissue-level epigenomic data and genotypes of BP-phenotyped individuals. METHODS: We used chromatin accessibility data from the heart, adrenal, kidney, and artery to identify cis-regulatory elements (CREs) in these tissues and estimate the impact of common human single-nucleotide variants within these CREs on gene expression, using machine learning methods. To identify causal genes, we performed a gene-wise association test. We conducted analyses in 2 separate large-scale cohorts: 77 822 individuals from the Genetic Epidemiology Research on Adult Health and Aging and 315 270 individuals from the UK Biobank. RESULTS: We identified 309, 259, 331, and 367 genes (false discovery rate <0.05) for diastolic BP and 191, 184, 204, and 204 genes for systolic BP in the artery, kidney, heart, and adrenal, respectively, in Genetic Epidemiology Research on Adult Health and Aging; 50% to 70% of these genes were replicated in the UK Biobank, significantly higher than the 12% to 15% expected by chance (P<0.0001). These results enabled tissue expression prediction of these 988 to 2875 putative BP genes in individuals of both cohorts to construct an expression polygenic score. This score explained ≈27% of the reported single-nucleotide variant heritability, substantially higher than expected from prior studies. CONCLUSIONS: Our work demonstrates the power of tissue-restricted comprehensive CRE analysis, followed by CRE-based expression prediction, for understanding BP regulation in relevant tissues and provides dual-modality supporting evidence, CRE and expression, for the causality genes.


Sujet(s)
Pression sanguine , Polymorphisme de nucléotide simple , Humains , Pression sanguine/génétique , Pression sanguine/physiologie , Mâle , Femelle , Éléments activateurs (génétique)/génétique , Adulte d'âge moyen , Régulation de l'expression des gènes , Hypertension artérielle/génétique , Hypertension artérielle/physiopathologie , Sujet âgé , Rein/métabolisme , Adulte , Glandes surrénales/métabolisme , Étude d'association pangénomique/méthodes
7.
Sci Rep ; 14(1): 12297, 2024 05 29.
Article de Anglais | MEDLINE | ID: mdl-38811798

RÉSUMÉ

The current study aimed to investigate the effect of Sox9-Cre-directed Nr5a1-conditional knockout (Sox9-Cre;Nr5a1flox/flox) on adrenal development. We showed that SOX9 is expressed by adrenocortical cells at E10.5-E11.5 but is extinguished no later than E12.5. The number of adrenocortical cells significantly reduced in Sox9-Cre;Nr5a1flox/flox mice while the number of cleaved caspase 3-positive cells increased compared to that in the controls at E11.5-E12.5, when the adrenal primordium (AP) is about to expand. This indicated that fetal adrenocortical cells are lost via apoptosis due to Nr5a1 ablation by E12.5. Both medulla formation and encapsulation were perturbed, accompanied by a smaller AP size, in Sox9-Cre;Nr5a1flox/flox mice during embryonic development. Adult Sox9-Cre;Nr5a1flox/flox adrenals were hypoplastic and exhibited irregular organization of the medulla with aberrant sex differentiation in the X zone. Additionally, there were histologically eosin-negative vacuolated cells, which were negative for both the X-zone marker 20αHSD and the steroidogenesis marker 3ßHSD at the innermost cortex of Sox9-Cre;Nr5a1flox/flox adrenals. Although Nr5a1+/- adrenals were hypoplastic, a small number of chromaffin cells were properly located in the center, having normal sex differences in the X-zone. The results collectively provided in-vivo evidence that Nr5a1 plays a critical role in AP expansion and subsequent adrenal development.


Sujet(s)
Glandes surrénales , Facteur de transcription SOX-9 , Facteur stéroïdogène-1 , Animaux , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Souris , Facteur stéroïdogène-1/métabolisme , Facteur stéroïdogène-1/génétique , Glandes surrénales/métabolisme , Glandes surrénales/embryologie , Integrases/métabolisme , Integrases/génétique , Souris knockout , Femelle , Mâle
8.
Nature ; 629(8014): 1082-1090, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38750354

RÉSUMÉ

Cell types with specialized functions fundamentally regulate animal behaviour, and yet the genetic mechanisms that underlie the emergence of novel cell types and their consequences for behaviour are not well understood1. Here we show that the monogamous oldfield mouse (Peromyscus polionotus) has recently evolved a novel cell type in the adrenal gland that expresses the enzyme AKR1C18, which converts progesterone into 20α-hydroxyprogesterone. We then demonstrate that 20α-hydroxyprogesterone is more abundant in oldfield mice, where it induces monogamous-typical parental behaviours, than in the closely related promiscuous deer mice (Peromyscus maniculatus). Using quantitative trait locus mapping in a cross between these species, we ultimately find interspecific genetic variation that drives expression of the nuclear protein GADD45A and the glycoprotein tenascin N, which contribute to the emergence and function of this cell type in oldfield mice. Our results provide an example by which the recent evolution of a new cell type in a gland outside the brain contributes to the evolution of social behaviour.


Sujet(s)
Glandes surrénales , Évolution biologique , Comportement paternel , Peromyscus , Animaux , Femelle , Mâle , 20alpha-Dihydroprogestérone/métabolisme , Glandes surrénales/cytologie , Glandes surrénales/enzymologie , Glandes surrénales/métabolisme , Oestradiol dehydrogenases/génétique , Oestradiol dehydrogenases/métabolisme , /génétique , Variation génétique , Hybridation génétique , Peromyscus/classification , Peromyscus/génétique , Peromyscus/physiologie , Progestérone/métabolisme , Locus de caractère quantitatif , Comportement social , Ténascine/génétique
10.
Circ Res ; 134(11): 1566-1580, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38781299

RÉSUMÉ

This interdisciplinary review explores the intricate nexus between HIV infection, nutrition, adrenal gland function, and cardiovascular health, highlighting a critical aspect of HIV management often overlooked in current literature. With the advent of antiretroviral therapy, the life expectancy of people living with HIV has dramatically improved, transforming HIV into a manageable chronic condition. However, this success brings forth new challenges, notably an increased risk of cardiovascular diseases among people living with HIV. We examine the normal physiology of the adrenal gland, including its role in mineral metabolism, a crucial facet of nutrition. We discuss the evolution of knowledge tying adrenal pathology to cardiovascular disease. We explore the impact of HIV on adrenal gland findings from a gross pathology perspective, as well as the clinical impact of adrenal insufficiency in HIV. The review further elucidates the role of nutrition in this context, considering the double burden of undernutrition and obesity prevalent in regions heavily affected by HIV. By aggregating findings from longitudinal studies and recent clinical trials, the review presents compelling evidence of increased cardiovascular disease among people living with HIV compared with people without HIV. It highlights the critical role of the adrenal glands in regulating nutrient metabolism and its implications for cardiovascular health, drawing attention to the potential for dietary interventions and targeted therapies to mitigate these risks. This review urges a paradigm shift in the management of HIV, advocating for a holistic approach that incorporates nutritional assessment and interventions into routine HIV care to address the complex interplay between HIV, adrenal function, and cardiovascular health. Through this lens, we offer insights into novel therapeutic strategies aimed at reducing cardiovascular risk in people living with HIV, contributing to the ongoing efforts to enhance the quality of life and longevity in this population.


Sujet(s)
Glandes surrénales , Maladies cardiovasculaires , Infections à VIH , État nutritionnel , Humains , Infections à VIH/complications , Maladies cardiovasculaires/étiologie , Glandes surrénales/métabolisme , Glandes surrénales/physiopathologie , Insuffisance surrénale/physiopathologie , Système cardiovasculaire/physiopathologie , Système cardiovasculaire/métabolisme
11.
J Cell Mol Med ; 28(10): e18376, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38780511

RÉSUMÉ

Taking into account homeostatic disorders resulting from arterial hypertension and the key importance of CacyBP/SIP, ß-catenin and endocannabinoids in the functioning of many organs, it was decided to assess the presence and distribution of CacyBP/SIP, ß-catenin, CB1 and CB2 in the adrenal glands of hypertensive rats of various aetiology. The study was conducted on the adrenal glands of rats with spontaneous and renovascular hypertension. The expression of CacyBP/SIP, ß-catenin, CB1 and CB2 was detected by immunohistochemistry and real-time PCR method. The results of the present study revealed both lower gene expression and immunoreactivity of CacyBP/SIP in the adrenal glands of all hypertensive groups compared to the normotensive rats. This study demonstrated a reduction in the immunoreactivity and expression of the ß-catenin, CB1 and CB2 genes in the adrenals of 2K1C rats. While in SHR, the reaction showing ß-catenin and CB1 was very weak or negative, and the expression of CB2 in the adrenal glands of these rats increased. The results of this study show, for the first time, marked differences in the expression of CacyBP/SIP, ß-catenin and CB1 and CB2 cannabinoid receptors in the adrenal glands of rats with primary (SHR) and secondary hypertension (2K1C).


Sujet(s)
Glandes surrénales , Hypertension artérielle , Récepteur cannabinoïde de type CB1 , Récepteur cannabinoïde de type CB2 , bêta-Caténine , Animaux , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Mâle , Hypertension artérielle/métabolisme , Hypertension artérielle/génétique , Glandes surrénales/métabolisme , Glandes surrénales/anatomopathologie , Récepteur cannabinoïde de type CB1/métabolisme , Récepteur cannabinoïde de type CB1/génétique , Récepteur cannabinoïde de type CB2/métabolisme , Récepteur cannabinoïde de type CB2/génétique , Rats , Rats de lignée SHR , Rat Wistar , Immunohistochimie , Récepteurs de cannabinoïdes/métabolisme , Récepteurs de cannabinoïdes/génétique , Hypertension rénovasculaire/métabolisme , Hypertension rénovasculaire/génétique , Hypertension rénovasculaire/anatomopathologie
12.
Int J Mol Sci ; 25(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38791437

RÉSUMÉ

Gap junctions (GJs) are important in the regulation of cell growth, morphology, differentiation and migration. However, recently, more attention has been paid to their role in the pathogenesis of different diseases as well as tumorigenesis, invasion and metastases. The expression pattern and possible role of connexins (Cxs), as major GJ proteins, under both physiological and pathological conditions in the adrenal gland, were evaluated in this review. The databases Web of Science, PubMed and Scopus were searched. Studies were evaluated if they provided data regarding the connexin expression pattern in the adrenal gland, despite current knowledge of this topic not being widely investigated. Connexin expression in the adrenal gland differs according to different parts of the gland and depends on ACTH release. Cx43 is the most studied connexin expressed in the adrenal gland cortex. In addition, Cx26, Cx32 and Cx50 were also investigated in the human adrenal gland. Cx50 as the most widespread connexin, along with Cx26, Cx29, Cx32, Cx36 and Cx43, has been expressed in the adrenal medulla with distinct cellular distribution. Considerable effort has recently been directed toward connexins as therapeutically targeted molecules. At present, there exist several viable strategies in the development of potential connexin-based therapeutics. The differential and hormone-dependent distribution of gap junctions within adrenal glands, the relatively large gap junction within this gland and the increase in the gap junction size and number following hormonal treatment would indicate that gap junctions play a pivotal role in cell functioning in the adrenal gland.


Sujet(s)
Connexines , Jonctions communicantes , Humains , Connexines/métabolisme , Jonctions communicantes/métabolisme , Tumeurs de la surrénale/métabolisme , Tumeurs de la surrénale/anatomopathologie , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Glandes surrénales/métabolisme , Glandes surrénales/anatomopathologie , Animaux , Régulation de l'expression des gènes tumoraux
13.
PLoS One ; 19(4): e0287421, 2024.
Article de Anglais | MEDLINE | ID: mdl-38653001

RÉSUMÉ

This study examined the psychogenic stress (PS) effects on changes in oxidative stress and the antioxidant capacity of an organism at different growth stages. The experimental animals were male Wistar rats of five different ages from growth periods (GPs) to old age. The growth stages were randomly classified into control (C) and experimental (PS) groups. The PS was performed using restraint and water immersion once daily for 3 h for 4 weeks. Reactive oxygen metabolites (d-ROMs) and the biological antioxidant potential (BAP) were measured before and after the experiment. In addition, the liver and adrenal glands were removed, and the wet weight was measured. The d-ROM and BAP of all growth stages given PS increased significantly. The d-ROM in the C group without PS increased significantly in GPs while decreased significantly in old-aged rats. In addition, the BAP of the C group in GP and early adulthood were all significantly elevated. There were significant differences in organ weights between the C and PS groups at all growth stages. Oxidative stress and antioxidant capacity differed depending on the organism's developmental status and growth stage, and PS also showed different effects. In particular, the variability in oxidative stress was remarkable, suggesting that the effect of PS was more significant in the organism's immature organs.


Sujet(s)
Antioxydants , Stress oxydatif , Rat Wistar , Stress psychologique , Animaux , Antioxydants/métabolisme , Mâle , Rats , Stress psychologique/métabolisme , Espèces réactives de l'oxygène/métabolisme , Foie/métabolisme , Taille d'organe , Glandes surrénales/métabolisme , Glandes surrénales/croissance et développement
14.
Poult Sci ; 103(6): 103712, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38603935

RÉSUMÉ

The effects of the administration of the opioid agonist, morphine, on plasma and tissue concentrations of Met-enkephalin were determined in 14 wk old female chickens. In addition, effects of morphine on proenkephalin (PENK) expression were examined. Plasma concentrations of Met-enkephalin were reduced 10 minutes after morphine administration. Plasma concentrations of peptides that contain Met-enkephalin motifs were decreased 30 minutes after morphine administration. Tissue concentrations of Met-enkephalin tended to be depressed following morphine administration. Adrenal concentrations of PENK peptides containing Met-enkephalin motifs were decreased in chickens challenged with morphine. Expression of PENK in the anterior pituitary gland and adrenal glands were decreased in morphine treated compared to control pullets. In contrast, plasma concentrations of corticosterone were elevated 10 min after morphine treatment. Morphine also induced changes in mu (µ) opioid receptors and delta (δ) opioid receptors in both anterior pituitary tissue and adrenal tissues.


Sujet(s)
Poulets , Corticostérone , Méthionine-enképhaline , Enképhalines , Morphine , Précurseurs de protéines , Animaux , Morphine/administration et posologie , Morphine/pharmacologie , Poulets/métabolisme , Méthionine-enképhaline/métabolisme , Femelle , Corticostérone/sang , Précurseurs de protéines/métabolisme , Enképhalines/métabolisme , Analgésiques morphiniques/administration et posologie , Analgésiques morphiniques/pharmacologie , Glandes surrénales/effets des médicaments et des substances chimiques , Glandes surrénales/métabolisme , Protéines aviaires/métabolisme , Protéines aviaires/génétique
15.
Sci Rep ; 14(1): 8044, 2024 04 05.
Article de Anglais | MEDLINE | ID: mdl-38580769

RÉSUMÉ

The crosstalk between the chromaffin and adrenocortical cells is essential for the endocrine activity of the adrenal glands. This interaction is also likely important for tumorigenesis and progression of adrenocortical cancer and pheochromocytoma. We developed a unique in vitro 3D model of the whole adrenal gland called Adrenoid consisting in adrenocortical carcinoma H295R and pheochromocytoma MTT cell lines. Adrenoids showed a round compact morphology with a growth rate significantly higher compared to MTT-spheroids. Confocal analysis of differential fluorescence staining of H295R and MTT cells demonstrated that H295R organized into small clusters inside Adrenoids dispersed in a core of MTT cells. Transmission electron microscopy confirmed the strict cell-cell interaction occurring between H295R and MTT cells in Adrenoids, which displayed ultrastructural features of more functional cells compared to the single cell type monolayer cultures. Adrenoid maintenance of the dual endocrine activity was demonstrated by the expression not only of cortical and chromaffin markers (steroidogenic factor 1, and chromogranin) but also by protein detection of the main enzymes involved in steroidogenesis (steroidogenic acute regulatory protein, and CYP11B1) and in catecholamine production (tyrosine hydroxylase and phenylethanolamine N-methyltransferase). Mass spectrometry detection of steroid hormones and liquid chromatography measurement of catecholamines confirmed Adrenoid functional activity. In conclusion, Adrenoids represent an innovative in vitro 3D-model that mimics the spatial and functional complexity of the adrenal gland, thus being a useful tool to investigate the crosstalk between the two endocrine components in the pathophysiology of this endocrine organ.


Sujet(s)
Tumeurs de la surrénale , Phéochromocytome , Humains , Glandes surrénales/métabolisme , Catécholamines/métabolisme , Chromogranine/métabolisme
16.
Cryobiology ; 115: 104895, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38616031

RÉSUMÉ

The study is devoted to the effect of lowered resuscitation temperature (26 °C) on cryopreserved porcine adrenal glands functional activity in vitro and in vivo under xenotransplantation. The adrenals were collected from newborn pigs, cryopreserved with 5 % DMSO at a rate of 1 °C/min, resuscitated at 26 or 37 °C for 48 h (5 % CO2, DMEM), embedded into small intestinal submucosa, and transplanted to bilaterally adrenalectomized rats. It has been shown that the glands resuscitated at 26 °C have suppressed free-radical processes and can produce cortisol and aldosterone in vitro, and may lead to elevated blood levels of these hormones. Moreover, the adrenal grafts maintain blood glucose levels and promote the formation of glycogen stores. Thus, the resuscitation at 26 °C can improve the quality of grafts and favor the introduction and application of the cryopreserved organs and tissues for transplantation in clinical and experimental practice.


Sujet(s)
Glandes surrénales , Cryoconservation , Hydrocortisone , Transplantation hétérologue , Animaux , Glandes surrénales/métabolisme , Transplantation hétérologue/méthodes , Cryoconservation/méthodes , Suidae , Hydrocortisone/sang , Rats , Glycémie/métabolisme , Glycémie/analyse , Aldostérone/sang , Aldostérone/métabolisme , Mâle , Glycogène/métabolisme , Réanimation/méthodes , Conservation d'organe/méthodes , Cryoprotecteurs/pharmacologie , Diméthylsulfoxyde/pharmacologie
17.
Mol Pharm ; 21(5): 2435-2440, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38626389

RÉSUMÉ

Among clinically used radiopharmaceuticals, iodine-123 labeled metaiodobenzylguanidine ([123I]mIBG) serves for diagnosing neuroendocrine tumors and obtaining images of myocardial sympathetic innervation. mIBG, a structural analogue of norepinephrine (NE), a neurotransmitter acting in peripheral and central nerves, follows a pathway similar to NE, transmitting signals through the NE transporter (NET) located at synaptic terminals. It moves through the body without decomposing, enabling noninvasive image evaluation. In this study, we aimed to quantify [123I]mIBG uptake in the adrenal glands using small animal single-photon emission computed tomography/computed tomography (SPECT/CT) images post [123I]mIBG administration. We investigated the possibility of assessing the effectiveness of ß-adrenergic receptor blockers by quantifying SPECT/CT images and biodistribution results to determine the degree of [123I]mIBG uptake in the adrenal glands treated with labetalol, a known ß-adrenergic receptor blocker. Upon intravenous administration of [123I]mIBG to mice, SPECT/CT images were acquired over time to confirm the in vivo distribution pattern, revealing a clear uptake in the adrenal glands. Labetalol inhibited the uptake of [123I]mIBG in cell lines expressing NET. A decrease in [123I]mIBG uptake in the adrenal glands was observed in the labetalol-treated group compared with the normal group through SPECT/CT imaging and biodistribution studies. These results demonstrate that SPECT/CT imaging with [123I]mIBG could be applicable for evaluating the preclinical efficacy of new antihypertensive drug candidates such as labetalol, a ß-adrenergic receptor blocker.


Sujet(s)
3-Iodobenzyl-guanidine , Antagonistes bêta-adrénergiques , Radio-isotopes de l'iode , Labétalol , Animaux , Humains , Mâle , Souris , Glandes surrénales/imagerie diagnostique , Glandes surrénales/effets des médicaments et des substances chimiques , Glandes surrénales/métabolisme , Antagonistes bêta-adrénergiques/pharmacologie , Antagonistes bêta-adrénergiques/pharmacocinétique , Lignée cellulaire tumorale , Études de faisabilité , Transporteurs de la norépinéphrine/métabolisme , Transporteurs de la norépinéphrine/antagonistes et inhibiteurs , Radiopharmaceutiques/pharmacocinétique , Tomographie par émission monophotonique couplée à la tomodensitométrie , Distribution tissulaire
18.
Pflugers Arch ; 476(6): 911-922, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38538989

RÉSUMÉ

Aldosterone is a steroid hormone that is important for maintaining the volume and ionic composition of extracellular fluids and is produced in the zona glomerulosa of the adrenal cortex. The basic mechanisms controlling aldosterone secretion are known. However, more detailed studies on the regulation of aldosterone secretion often fail due to the lack of suitable models: although secretion can be studied in cultured adrenocortical cells under defined conditions, the differentiation status of the cells is difficult to control and the complex anatomy of the adrenal cortex is lost. In living animals, the physiological context is intact, but the influences are manifold and the examination conditions cannot be sufficiently controlled. One method that closes the gap between cell models and studies in living animals is the isolated perfused adrenal gland. In the past, this method has provided important data on the pathophysiology of adrenal glands from larger animals, but the technique was not used in mice. Here, we developed a method for isolation and perfusion of the mouse adrenal gland to study aldosterone secretion. This technique preserves the complex anatomical and functional context of the mouse adrenal cortex, to ensure defined experimental conditions and to minimize extra-adrenal influences. Initial series of experiments with the ex vivo perfused mouse adrenal gland show that this model offers the possibility for unique insights into pathophysiological regulatory principles and is suitable for the use of genetically modified mouse models.


Sujet(s)
Glandes surrénales , Aldostérone , Animaux , Aldostérone/métabolisme , Souris , Glandes surrénales/métabolisme , Souris de lignée C57BL , Mâle , Perfusion/méthodes , Zone glomérulée/métabolisme
19.
Nat Aging ; 4(3): 396-413, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38503993

RÉSUMÉ

Adrenal glands, vital for steroid secretion and the regulation of metabolism, stress responses and immune activation, experience age-related decline, impacting systemic health. However, the regulatory mechanisms underlying adrenal aging remain largely uninvestigated. Here we established a single-nucleus transcriptomic atlas of both young and aged primate suprarenal glands, identifying lipid metabolism and steroidogenic pathways as core processes impacted by aging. We found dysregulation in centripetal adrenocortical differentiation in aged adrenal tissues and cells in the zona reticularis region, responsible for producing dehydroepiandrosterone sulfate (DHEA-S), were highly susceptible to aging, reflected by senescence, exhaustion and disturbed hormone production. Remarkably, LDLR was downregulated in all cell types of the outer cortex, and its targeted inactivation in human adrenal cells compromised cholesterol uptake and secretion of dehydroepiandrosterone sulfate, as observed in aged primate adrenal glands. Our study provides crucial insights into endocrine physiology, holding therapeutic promise for addressing aging-related adrenal insufficiency and delaying systemic aging.


Sujet(s)
Glandes surrénales , Vieillissement , Animaux , Humains , Sujet âgé , Sulfate de déhydroépiandrostérone/métabolisme , Glandes surrénales/métabolisme , Vieillissement/génétique , Zone réticulée , Primates/métabolisme
20.
Front Endocrinol (Lausanne) ; 15: 1291775, 2024.
Article de Anglais | MEDLINE | ID: mdl-38419957

RÉSUMÉ

Objective: To investigate the diagnostic efficiency and prognostic value of 68Ga-Pentixafor PET/CT in comparison with adrenal vein sampling (AVS) for functional lateralization in primary aldosteronism (PA). Histology and long-term clinical follow-up normally serve as the gold standard for such diagnosis. Methods: We prospectively recruited 26 patients diagnosed with PA. All patients underwent 68Ga-Pentixafor PET/CT and AVS. Postsurgical biochemical and clinical outcomes of patients with unilateral primary aldosteronism (UPA), as diagnosed by PET/CT or AVS, were assessed by applying standardized Primary Aldosteronism Surgical Outcome (PASO) criteria. Immunohistochemistry (IHC) was performed to detect the expression of aldosterone synthase (CYP11B2) and CXCR4. Results: On total, 19 patients were diagnosed with UPA; of these, 13 patients were lateralized by both PET/CT and AVS, four patients were lateralized by PET-only, and two by AVS-only. Seven subjects with no lateralization on AVS and PET received medical therapy. All patients achieved complete biochemical success except one with nodular hyperplasia lateralized by AVS alone. The consistency between PET/CT and AVS outcomes was 77% (20/26). Moreover, CYP11B2-positive nodules were all CXCR4-positive and showed positive findings on PET. Patients who achieved complete biochemical and clinical success had a higher uptake on PET as well as stronger expression levels of CXCR4 and CYP11B2. Conclusion: Our analysis showed that 68Ga-Pentixafor PET/CT could enable non-invasive diagnosis in most patients with PA and identify additional cases of unilateral and surgically curable PA which could not be classified by AVS. 68Ga-Pentixafor PET/CT should be considered as a first-line test for the future classification of PA.


Sujet(s)
Complexes de coordination , Hyperaldostéronisme , Peptides cycliques , Tomographie par émission de positons couplée à la tomodensitométrie , Humains , Glandes surrénales/métabolisme , Radio-isotopes du gallium/métabolisme , Cytochrome P-450 CYP11B2/métabolisme , Hyperaldostéronisme/diagnostic , Hyperaldostéronisme/chirurgie , Hyperaldostéronisme/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...