Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
Clin Chim Acta ; 553: 117749, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38169194

ABSTRACT

The measurement of steroid hormones in blood and urine, which reflects steroid biosynthesis and metabolism, has been recognized as a valuable tool for identifying and distinguishing steroidogenic disorders. The application of mass spectrometry enables the reliable and simultaneous analysis of large panels of steroids, ushering in a new era for diagnosing adrenal diseases. However, the interpretation of complex hormone results necessitates the expertise and experience of skilled clinicians. In this scenario, machine learning techniques are gaining worldwide attention within healthcare fields. The clinical values of combining mass spectrometry-based steroid profiles analysis with machine learning models, also known as steroid metabolomics, have been investigated for identifying and discriminating adrenal disorders such as adrenocortical carcinomas, adrenocortical adenomas, and congenital adrenal hyperplasia. This promising approach is expected to lead to enhanced clinical decision-making in the field of adrenal diseases. This review will focus on the clinical performances of steroid profiling, which is measured using mass spectrometry and analyzed by machine learning techniques, in the realm of decision-making for adrenal diseases.


Subject(s)
Adrenal Cortex Neoplasms , Adrenal Gland Diseases , Adrenocortical Adenoma , Adrenocortical Carcinoma , Humans , Adrenal Gland Diseases/diagnosis , Adrenal Gland Diseases/metabolism , Adrenocortical Adenoma/diagnosis , Adrenocortical Adenoma/pathology , Adrenocortical Carcinoma/diagnosis , Steroids/metabolism , Adrenal Cortex Neoplasms/diagnosis
3.
Mol Cell Endocrinol ; 522: 111120, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33338548

ABSTRACT

The Wnt signaling pathway is a critical mediator of the development and maintenance of several tissues. The adrenal cortex is highly dependent upon Wnt/ß-catenin signaling for proper zonation and endocrine function. Adrenocortical cells emerge in the peripheral capsule and subcapsular cortex of the gland as progenitor cells that centripetally differentiate into steroid hormone-producing cells of three functionally distinct concentric zones that respond robustly to various endocrine stimuli. Wnt/ß-catenin signaling mediates adrenocortical progenitor cell fate and tissue renewal to maintain the gland throughout life. Aberrant Wnt/ß-catenin signaling contributes to various adrenal disorders of steroid production and growth that range from hypofunction and hypoplasia to hyperfunction, hyperplasia, benign adrenocortical adenomas, and malignant adrenocortical carcinomas. Great strides have been made in defining the molecular underpinnings of adrenocortical homeostasis and disease, including the interplay between the capsule and cortex, critical components involved in maintaining the adrenocortical Wnt/ß-catenin signaling gradient, and new targets in adrenal cancer. This review seeks to examine these and other recent advancements in understanding adrenocortical Wnt/ß-catenin signaling and how this knowledge can inform therapeutic options for adrenal disease.


Subject(s)
Adrenal Cortex/metabolism , Adrenal Cortex/pathology , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/pathology , beta Catenin/metabolism , Animals , Humans , Ligands , Regeneration , Wnt Signaling Pathway
4.
Int J Mol Sci ; 21(14)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32709015

ABSTRACT

Cardiomyopathies are myocardial disorders in which heart muscle is structurally and/or functionally abnormal. Previously, structural cardiomyocyte disorders due to adrenal diseases, such as hyperaldosteronism, hypercortisolism, and hypercatecholaminism, were misunderstood, and endomyocardial biopsy (EMB) was not performed because was considered dangerous and too invasive. Recent data confirm that, if performed in experienced centers, EMB is a safe technique and gives precious information about physiopathological processes implied in clinical abnormalities in patients with different systemic disturbances. In this review, we illustrate the most important features in patients affected by primary aldosteronism (PA), Cushing's syndrome (CS), and pheochromocytoma (PHEO). Then, we critically describe microscopic and ultrastructural aspects that have emerged from the newest EMB studies. In PA, the autonomous hypersecretion of aldosterone induces the alteration of ion and water homeostasis, intracellular vacuolization, and swelling; interstitial oedema could be a peculiar feature of myocardial toxicity. In CS, cardiomyocyte hypertrophy and myofibrillolysis could be related to higher expression of atrogin-1. Finally, in PHEO, the hypercontraction of myofilaments with the formation of contraction bands and occasional cellular necrosis has been observed. We expect to clear the role of EMB in patients with cardiomyopathies and adrenal disease, and we believe EMB is a valid tool to implement new management and therapies.


Subject(s)
Adrenal Gland Diseases/complications , Adrenal Gland Diseases/pathology , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Adrenal Gland Diseases/diagnosis , Adrenal Gland Diseases/metabolism , Aldosterone/metabolism , Animals , Biopsy , Cardiomyopathies/diagnosis , Cardiomyopathies/metabolism , Catecholamines/metabolism , Endocardium/metabolism , Endocardium/pathology , Humans , Hydrocortisone/metabolism , Myocardium/metabolism , Myocardium/pathology
5.
Postgrad Med J ; 96(1137): 412-416, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32527756

ABSTRACT

Coronavirus infection outbreaks have occurred frequently in the last two decades and have led to significant mortality. Despite the focus on reducing mortality by preventing the spread of the virus, patients have died due to several other complications of the illness. The understanding of pathological mechanisms and their implications is continuously evolving. A number of symptoms occur in these patients due to the involvement of various endocrine glands. These clinical presentations went largely unnoticed during the first outbreak of severe acute respiratory syndrome (SARS) in 2002-2003. A few of these derangements continued during the convalescence phase and sometimes occurred after recovery. Similar pathological and biochemical changes are being reported with the novel coronavirus disease outbreak in 2020. In this review, we focus on these endocrine changes that have been reported in both SARS coronavirus and SARS coronavirus-2. As we battle the pandemic, it becomes imperative to address these underlying endocrine disturbances that are contributing towards or predicting mortality of these patients.


Subject(s)
Adrenal Gland Diseases/physiopathology , Betacoronavirus/physiology , Coronavirus Infections/physiopathology , Diabetes Mellitus/physiopathology , Pandemics , Pneumonia, Viral/physiopathology , Severe Acute Respiratory Syndrome/physiopathology , Severe acute respiratory syndrome-related coronavirus/physiology , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/virology , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Diabetes Mellitus/virology , Humans , Hyperglycemia , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Practice Guidelines as Topic , SARS-CoV-2 , Severe Acute Respiratory Syndrome/metabolism , Severe Acute Respiratory Syndrome/virology
6.
J Endocrinol ; 246(2): R13-R31, 2020 08.
Article in English | MEDLINE | ID: mdl-32380472

ABSTRACT

The circadian rhythm derives from the integration of many signals that shape the expression of clock-related genes in a 24-h cycle. Biological tasks, including cell proliferation, differentiation, energy storage, and immune regulation, are preferentially confined to specific periods. A gating system, supervised by the central and peripheral clocks, coordinates the endogenous and exogenous signals and prepares for transition to activities confined to periods of light or darkness. The fluctuations of cortisol and its receptor are crucial in modulating these signals. Glucocorticoids and the autonomous nervous system act as a bridge between the suprachiasmatic master clock and almost all peripheral clocks. Additional peripheral synchronizing mechanisms including metabolic fluxes and cytokines stabilize the network. The pacemaker is amplified by peaks and troughs in cortisol and their response to food, activity, and inflammation. However, when the glucocorticoid exposure pattern becomes chronically flattened at high- (as in Cushing's syndrome) or low (as in adrenal insufficiency) levels, the system fails. While endocrinologists are well aware of cortisol rhythm, too little attention has been given to interventions aimed at restoring physiological cortisol fluctuations in adrenal disorders. However, acting on glucocorticoid levels may not be the only way to restore clock-related activities. First, a counterregulatory mechanism on the glucocorticoid receptor itself controls signal transduction, and second, melatonin and/or metabolically active drugs and nutrients could also be used to modulate the clock. All these aspects are described herein, providing some insights into the emerging role of chronopharmacology, focusing on glucocorticoid excess and deficiency disorders.


Subject(s)
Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/pathology , Adrenal Gland Diseases/therapy , Adrenal Glands/metabolism , Adrenal Glands/physiology , Chronotherapy/methods , Glucocorticoids/metabolism , Animals , Circadian Rhythm/physiology , Humans , Hypothalamo-Hypophyseal System/metabolism
7.
Compr Physiol ; 9(4): 1443-1502, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31688964

ABSTRACT

Chromaffin cells (CCs) of the adrenal gland and the sympathetic nervous system produce the catecholamines (epinephrine and norepinephrine; EPI and NE) needed to coordinate the bodily "fight-or-flight" response to fear, stress, exercise, or conflict. EPI and NE release from CCs is regulated both neurogenically by splanchnic nerve fibers and nonneurogenically by hormones (histamine, corticosteroids, angiotensin, and others) and paracrine messengers [EPI, NE, adenosine triphosphate, opioids, γ-aminobutyric acid (GABA), etc.]. The "stimulus-secretion" coupling of CCs is a Ca2+ -dependent process regulated by Ca2+ entry through voltage-gated Ca2+ channels, Ca2+ pumps, and exchangers and intracellular organelles (RE and mitochondria) and diffusible buffers that provide both Ca2+ -homeostasis and Ca2+ -signaling that ultimately trigger exocytosis. CCs also express Na+ and K+ channels and ionotropic (nAChR and GABAA ) and metabotropic receptors (mACh, PACAP, ß-AR, 5-HT, histamine, angiotensin, and others) that make CCs excitable and responsive to autocrine and paracrine stimuli. To maintain high rates of E/NE secretion during stressful conditions, CCs possess a large number of secretory chromaffin granules (CGs) and members of the soluble NSF-attachment receptor complex protein family that allow docking, fusion, and exocytosis of CGs at the cell membrane, and their recycling. This article attempts to provide an updated account of well-established features of the molecular processes regulating CC function, and a survey of the as-yet-unsolved but important questions relating to CC function and dysfunction that have been the subject of intense research over the past 15 years. Examples of CCs as a model system to understand the molecular mechanisms associated with neurodegenerative diseases are also provided. Published 2019. Compr Physiol 9:1443-1502, 2019.


Subject(s)
Adrenal Gland Diseases/pathology , Adrenal Glands/cytology , Adrenal Glands/physiology , Chromaffin Cells/physiology , Adrenal Gland Diseases/metabolism , Gene Expression Regulation , Humans , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology
8.
Endocr Rev ; 40(6): 1605-1625, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31294783

ABSTRACT

Steroid biosynthesis and metabolism are reflected by the serum steroid metabolome and, in even more detail, by the 24-hour urine steroid metabolome, which can provide unique insights into alterations of steroid flow and output indicative of underlying conditions. Mass spectrometry-based steroid metabolome profiling has allowed for the identification of unique multisteroid signatures associated with disorders of steroid biosynthesis and metabolism that can be used for personalized approaches to diagnosis, differential diagnosis, and prognostic prediction. Additionally, steroid metabolome analysis has been used successfully as a discovery tool, for the identification of novel steroidogenic disorders and pathways as well as revealing insights into the pathophysiology of adrenal disease. Increased availability and technological advances in mass spectrometry-based methodologies have refocused attention on steroid metabolome profiling and facilitated the development of high-throughput steroid profiling methods soon to reach clinical practice. Furthermore, steroid metabolomics, the combination of mass spectrometry-based steroid analysis with machine learning-based approaches, has facilitated the development of powerful customized diagnostic approaches. In this review, we provide a comprehensive up-to-date overview of the utility of steroid metabolome analysis for the diagnosis and management of inborn disorders of steroidogenesis and autonomous adrenal steroid excess in the context of adrenal tumors.


Subject(s)
Adrenal Gland Diseases/metabolism , Metabolome , Steroid Metabolism, Inborn Errors/metabolism , Adrenal Gland Diseases/diagnosis , Diagnosis, Differential , Humans , Steroid Metabolism, Inborn Errors/diagnosis
9.
Cell Stem Cell ; 25(2): 290-296.e2, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31104943

ABSTRACT

Evolution has resulted in profound differences between males and females that extend to non-reproductive organs and are reflected in the susceptibility and progression of diseases. However, the cellular and molecular basis for these differences remains largely unknown. Here we report that adrenal gland tissue renewal is highly active and sexually dimorphic, with female mice showing a 3-fold higher turnover than males. Moreover, in males, homeostasis relies on proliferation of cells within the steroidogenic zone, but females employ an additional stem and/or progenitor compartment situated in the adrenal capsule. Using lineage tracing, sex reversal models, gonadectomy, and dihydrotestosterone treatments, we further show that sex-specific stem cell activity is driven by male hormones that repress recruitment of Gli1+ stem cells from the capsule and cell proliferation. Taken together, our findings provide a molecular and cellular basis for adrenal sex dimorphism that may contribute to the increased incidence of adrenal diseases in females.


Subject(s)
Adrenal Gland Diseases/metabolism , Sex Factors , Stem Cells/physiology , Adrenal Cortex/physiology , Adrenal Gland Diseases/epidemiology , Adrenal Gland Diseases/pathology , Biological Evolution , Castration , Cell Proliferation , Cell Self Renewal , Cells, Cultured , Disease Susceptibility , Female , Homeostasis , Humans , Incidence , Male , Regeneration , Sex Characteristics
10.
J Steroid Biochem Mol Biol ; 190: 273-280, 2019 06.
Article in English | MEDLINE | ID: mdl-30707926

ABSTRACT

Adrenal steroidogenesis is a robust process, involving a series of enzymatic reactions that facilitate conversion of cholesterol into biologically active steroid hormones under the stimulation of angiotensin II, adrenocorticotropic hormone and other regulators. The biosynthesis of mineralocorticoids, glucocorticoids, and adrenal-derived androgens occur in separate adrenocortical zones as a result of the segregated expression of steroidogenic enzymes and cofactors. This mini review provides the principles of adrenal steroidogenesis, including the classic and under-appreciated 11-oxygenated androgen pathways. Several adrenal diseases result from dysregulated adrenal steroid synthesis. Herein, we review growing evidence that adrenal diseases exhibit characteristic modifications from normal adrenal steroid pathways that provide opportunities for the discovery of biomarker steroids that would improve diagnosis and monitoring of adrenal disorders.


Subject(s)
Adrenal Gland Diseases/metabolism , Steroids/metabolism , Adrenal Gland Diseases/blood , Adrenal Gland Diseases/diagnosis , Adrenal Gland Diseases/urine , Adrenal Glands/metabolism , Biomarkers/blood , Biomarkers/metabolism , Biomarkers/urine , Biosynthetic Pathways , Humans , Steroids/blood , Steroids/urine
11.
Toxicol Lett ; 302: 7-17, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30528684

ABSTRACT

Glucocorticoids play a major factor in fetal maturation and fate decision after birth. We have previously demonstrated that prenatal caffeine exposure (PCE) resulted in adrenal dysplasia. However, its molecular mechanism has not been clarified. In the present study, a rat model of intrauterine growth retardation (IUGR) was established by PCE, and offspring were sacrificed. Moreover, NCI-H295 A cells were used to confirm glucocorticoid-related molecular mechanism. Results showed that PCE fetal weight decreased, and the IUGR rate increased, while serum corticosterone levels increased but insulin-like growth factor 1 (IGF1) levels decreased. Fetal adrenals exhibited an activated glucocorticoid-activation system, and the downregulated expression of IGF1 signal pathway and steroidal synthetases. For adult rats, there was no significant change in the glucocorticoid-activation system in the PCE group, the IGF1 signal pathway showed increased trend, and the expression levels of adrenal steroidal synthetases were close to normal. The data in vitro showed that the cortisol of 1200 nM can inhibit the expression of adrenocortical cell steroidal synthetases and IGF1 signal pathway when compared with the control. Meanwhile, the glucocorticoid-activation system was activated while GR inhibitor mifepristone can reverse the effect of cortisol. Furthermore, cortisol can also promote GR into the nucleus after its activation. Based on these findings, we speculated that high concentrations of glucocorticoid in utero led to GR in the nucleus through its activation and then inhibited the IGF1 signaling pathway by activating the glucocorticoid-activation system, which could further downregulate steroid synthesis.


Subject(s)
Adrenal Gland Diseases/chemically induced , Adrenal Glands/drug effects , Caffeine/toxicity , Central Nervous System Stimulants/toxicity , Corticosterone/metabolism , Fetal Growth Retardation/chemically induced , Hydrocortisone/metabolism , Insulin-Like Growth Factor I/metabolism , Prenatal Exposure Delayed Effects , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/pathology , Adrenal Glands/metabolism , Adrenal Glands/pathology , Age Factors , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Corticosterone/blood , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Male , Maternal Exposure , Phosphoproteins/metabolism , Pregnancy , Rats, Wistar
12.
Neuroendocrinology ; 107(4): 340-354, 2018.
Article in English | MEDLINE | ID: mdl-30208370

ABSTRACT

BACKGROUND/AIMS: Abuse of toluene products (e.g., glue-sniffing) primarily occurs during adolescence and has been associated with appetite suppression and weight impairments. However, the metabolic phenotype arising from adolescent inhalant abuse has never been fully characterised, and its persistence during abstinence and underlying mechanisms remain unknown. METHODS: Adolescent male Wistar rats (post-natal day 27) were exposed to inhaled toluene (10,000 ppm) (n = 32) or air (n = 48) for 1 h/day, 3 days/week for 4 weeks, followed by 4 weeks of abstinence. Twenty air rats were pair-fed to the toluene group, to differentiate the direct effects of toluene from under-nutrition. Food intake, weight, and growth were monitored. Metabolic hormones were measured after exposure and abstinence periods. Energy expenditure was measured using indirect calorimetry. Adrenal function was assessed using adrenal histology and hormone testing. RESULTS: Inhalant abuse suppressed appetite and increased energy expenditure. Reduced weight gain and growth were observed in both the toluene and pair-fed groups. Compared to the pair-fed group, and despite normalisation of food intake, the suppression of weight and growth for toluene-exposed rats persisted during abstinence. After exposure, toluene-exposed rats had low fasting blood glucose and insulin compared to the air and pair-fed groups. Consistent with adrenal insufficiency, adrenal hypertrophy and increased basal adrenocorticotropic hormone were observed in the toluene-exposed rats, despite normal basal corticosterone levels. CONCLUSIONS: Inhalant abuse results in negative energy balance, persistent growth impairment, and endocrine changes suggestive of adrenal insufficiency. We conclude that adrenal insufficiency contributes to the negative energy balance phenotype, potentially presenting a significant additional health risk for inhalant users.


Subject(s)
Adrenal Gland Diseases/chemically induced , Growth Disorders/chemically induced , Inhalant Abuse/complications , Metabolic Diseases/chemically induced , Sexual Maturation , Adolescent , Adolescent Behavior/drug effects , Adolescent Behavior/physiology , Adolescent Development/drug effects , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/physiopathology , Adrenal Glands/physiopathology , Animals , Appetite/drug effects , Body Weight/drug effects , Disease Models, Animal , Eating/drug effects , Growth Disorders/metabolism , Growth Disorders/physiopathology , Humans , Inhalant Abuse/metabolism , Inhalant Abuse/pathology , Inhalant Abuse/physiopathology , Male , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Motor Activity/drug effects , Phenotype , Rats , Rats, Wistar , Sexual Maturation/drug effects , Sexual Maturation/physiology , Toluene/toxicity
13.
Cell Mol Life Sci ; 75(6): 1057-1070, 2018 03.
Article in English | MEDLINE | ID: mdl-29058015

ABSTRACT

Neuroblastoma (NB) is a tumor of the sympathoadrenal system arising in children under 15 years of age. In Germany, NB accounts for 7% of childhood cancer cases, but 11% of cancer deaths. It originates from highly migratory progenitor cells that leave the dorsal neural tube and contribute neurons and glial cells to sympathetic ganglia, and chromaffin and supportive cells to the adrenal medulla and paraganglia. Clinically, histologically and molecularly, NBs present as extremely heterogeneous, ranging from very good to very poor prognosis. The etiology of NB still remains unclear and needs to be elucidated, however, aberrant auto- and paracrine embryonic cell communications seem to be likely candidates to initiate or facilitate the emergence, progression and regression of NB. The wingless-type MMTV integration site (WNT) family of proteins represents an evolutionary highly conserved signaling system that orchestrates embryogenesis. At least 19 ligands in the human, numerous receptors and co-receptors are known, which control not only proliferation, but also cell polarity, migration and differentiation. Here we seek to interconnect aspects of WNT signaling with sympathoadrenal and paraganglionic development to define new WNT signaling cues in the etiology and progression of NB.


Subject(s)
Adrenal Gland Diseases/genetics , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Paraganglioma/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , Adolescent , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/mortality , Adrenal Gland Diseases/pathology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adrenal Glands/pathology , Child , Child, Preschool , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/growth & development , Neural Crest/metabolism , Neural Crest/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Paraganglioma/metabolism , Paraganglioma/mortality , Paraganglioma/pathology , Stem Cells/metabolism , Stem Cells/pathology , Survival Analysis , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Wnt Proteins/metabolism
14.
Arch Med Res ; 48(6): 488-497, 2017 08.
Article in English | MEDLINE | ID: mdl-29203053

ABSTRACT

BACKGROUND: Our previous study demonstrated that prenatal food restriction (PFR) could induce the dysfunction of the hypothalamic-pituitary-adrenal axis and glucocorticoid-related glucose and lipid metabolic alterations in adult offspring rats. AIM OF THE STUDY: To investigate the intrauterine programming mechanism of adrenal dysfunction in the PFR offspring rats. METHODS: From gestational days (GDs) 11-20, pregnant Wistar rats were fed a restricted diet (50% of the daily food intake of control rats, 60 g/kg·d). Some were executed at GD20, while the others survived to full-term delivery; all pups were fed a high-fat diet (HFD) after weaning. The serum corticosterone concentration, expression level of adrenal steroidal synthetase, and insulin-like growth factor 1 (IGF1) signaling pathway were tested. RESULTS: We confirmed that the fetal body weight of the PFR group was lower than that of the control group, and the mRNA expression of adrenal steroidogenic acute regulatory protein, cytochrome P450 cholesterol side chain cleavage, 3ß-hydroxysteroid dehydrogenase, and steroid 11ß-hydroxylase (P450c11) were decreased in the PFR fetal rats. The maternal and fetal serum corticosterone levels were significantly increased in the PFR groups. Furthermore, the expression of the adrenal IGF1 signaling pathway (including IGF1, IGF1R, and Akt1) was suppressed. However, after a post-weaning HFD, the body weight gain rates and serum corticosterone levels were elevated, and the expression of adrenal steroid 21-hydroxylase and P450c11, as well as the IGF1 signaling pathway, were significantly increased in the PFR group. CONCLUSIONS: These results showed that a higher level of circulation corticosterone by PFR in utero inhibited adrenal IGF1 signaling and steroidogenesis, whereas post-weaning HFD induced adrenal steroidogenesis by an enhanced IGF1 signaling.


Subject(s)
Diet, Reducing/adverse effects , Hypothalamo-Hypophyseal System/physiology , Insulin-Like Growth Factor I/physiology , Pituitary-Adrenal System/physiology , Prenatal Exposure Delayed Effects/metabolism , Adrenal Gland Diseases/metabolism , Animals , Body Weight/physiology , Cholesterol/metabolism , Corticosterone/blood , Disease Models, Animal , Female , Glucose/metabolism , Insulin-Like Growth Factor I/metabolism , Phosphoproteins/metabolism , Pregnancy , Rats , Rats, Wistar
15.
Hormones (Athens) ; 16(3): 251-265, 2017 Jul.
Article in English | MEDLINE | ID: mdl-29278511

ABSTRACT

The adrenal gland is a key component of the stress system in the human body. Multiple direct and paracrine interactions between different cell types and their progenitors take place within the adrenal gland microenvironment. These unique interactions are supported by high vascularization and the adrenal cortex extracellular matrix. Alterations in the adrenal gland microenvironment are known to influence the progression of several pathological conditions, such as obesity and sepsis, and to be influenced by these disorders. For example, it has been suggested that activation of immune-adrenal crosstalk during sepsis induces elevated adrenal glucocorticoid levels, whereas crosstalk between adrenocortical cells and sonic hedgehog responsive stem cells was found to contribute to the increased size of the adrenal cortex during obesity. By contrast to sepsis, where activation of adrenal glucocorticoid production has protective effects, chronic exposure to high levels of glucocorticoids induces adverse effects, typically manifested in patients with Cushing syndrome, such as increased body weight, dyslipidemia, glucose intolerance, and hypertension. Therefore, a better understanding of factors involved in the regulation of the adrenal gland microenvironment is crucial. This review highlights bidirectional interactions occurring between the adrenal gland microenvironment and systemic responses during obesity and sepsis. Furthermore, it presents and discusses recent advancements and challenges in attempts to restore or regenerate adrenal gland function, including the use of oxygenated immune-isolating devices.


Subject(s)
Adrenal Gland Diseases/metabolism , Adrenal Glands/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Adrenal Gland Diseases/physiopathology , Adrenal Glands/physiopathology , Humans , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Regeneration/physiology
16.
BMJ Case Rep ; 20172017 Jul 24.
Article in English | MEDLINE | ID: mdl-28739615

ABSTRACT

Primary adrenal disorders contribute 20%â€"30% of patients with endogenous Cushing's syndrome. Most of the primary adrenal diseases are unilateral and include adenoma and adrenocortical carcinoma, whereas bilateral adrenal lesions are uncommon and include primary pigmented nodular adrenocortical disease, primary bilateral macronodular adrenocortical hyperplasia, isolated micronodular adrenocortical disease, bilateral adenomas or carcinomas, and rarely pituitary adrenocorticotropic hormone-dependent adrenal nodular disease. Cyclic adenosine monophosphate-dependent protein kinase A signalling is the major activator of cortisol secretion in primary adrenal nodular disorders. We report two cases of bilateral adrenal nodular disease with endogenous Cushing's syndrome, including one each of primary pigmented nodular adrenocortical disease and primary bilateral macronodular adrenocortical hyperplasia.


Subject(s)
Adrenal Gland Diseases/complications , Adrenal Glands/pathology , Cushing Syndrome/etiology , Adrenal Cortex Diseases/complications , Adrenal Cortex Diseases/metabolism , Adrenal Cortex Diseases/pathology , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/pathology , Adrenal Glands/metabolism , Adult , Child, Preschool , Cushing Syndrome/metabolism , Cushing Syndrome/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Hydrocortisone/metabolism , Hyperplasia , Male
17.
Horm Metab Res ; 49(4): 269-275, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28103616

ABSTRACT

In the normal human adrenal gland, serotonin (5-HT) stimulates aldosterone secretion through the 5-HT4 receptor (5-HT4R). However, the physiological role of the serotonergic control of adrenocortical function is not known. In the present study, we have investigated the ability of l-Lysine, which has been shown to act as a 5-HT4 receptor antagonist, to counteract in vitro and in vivo the stimulatory effect of 5-HT4R agonists on aldosterone production. l-Lysine was found to inhibit aldosterone production induced by 5-HT and the 5-HT4R agonists BIMU8 from cultured human adrenocortical cells. The action of l-Lysine (4.95 g/day orally) on the adrenal cortex was also evaluated in 20 healthy volunteers in a double blind, cross-over, placebo controlled study. l-Lysine had no significant influence on basal plasma aldosterone levels and the aldosterone responses to upright posture, tetracosactide, and low sodium diet (10 mmol/day for 3 days). Conversely, l-Lysine significantly reduced the surge of plasma aldosterone induced by metoclopramide indicating that l-Lysine is able to efficiently antagonize the adrenal 5-HT4 receptors in vivo. These results suggest that l-Lysine supplementation may represent a new treatment of primary adrenal diseases in which corticosteroid hypersecretion is driven by overexpressed 5-HT4 receptors.


Subject(s)
Adrenal Gland Diseases/drug therapy , Adrenal Glands/metabolism , Aldosterone/metabolism , Lysine/administration & dosage , Receptors, Serotonin, 5-HT4/metabolism , Serotonin 5-HT4 Receptor Antagonists/administration & dosage , Serotonin Agents/administration & dosage , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/pathology , Adrenal Glands/pathology , Cells, Cultured , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Serotonin/metabolism
18.
Minerva Endocrinol ; 42(2): 151-163, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27973460

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the developed world and its pathogenesis is complex and multifactorial. It is considered the hepatic manifestation of the metabolic syndrome and is the leading cause of hepatic cirrhosis. This review aims to present current knowledge on the involvement of the adrenal glands in the development of NAFLD. Clinical and animal studies have shown that excess glucocorticoids (GC) have been implicated in the pathogenesis of NAFLD. Patients with NAFLD seem to have a subtle chronic activation of the hypothalamic pituitary adrenal axis leading to a state of subclinical hypercortisolism. Regulators of GC such as 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), an enzyme that regenerates cortisol from inactive cortisone, and 5α/5ß-reductases, enzymes that increase cortisol clearance, are implicated in the development of NAFLD by amplifying local GC action. Adrenal androgen (dehydroepiandrosterone) abnormalities and increased aldosterone levels may also have a role in the development of NAFLD whereas the contribution of adrenergic signaling in NAFLD pathogenesis remains unclear.


Subject(s)
Adrenal Cortex Hormones/metabolism , Adrenal Gland Diseases/complications , Non-alcoholic Fatty Liver Disease/complications , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/therapy , Animals , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy
19.
Curr Pharm Des ; 22(36): 5588-5602, 2016.
Article in English | MEDLINE | ID: mdl-27510480

ABSTRACT

BACKGROUND: The prevalence of adrenal hyperandrogenism (AH), as defined by increased circulating dehydroepiandrosterone-sulfate (DHEAS) levels, ranges from 15 to 45% in women with polycystic ovary syndrome (PCOS). METHODS: The aim of this review is to update the pathogenesis and consequences of AH in PCOS, from molecular genetics to the clinical setting. RESULTS: Mounting evidence derived from animal models suggests that genetically or enviromentally determined prenatal androgen excess, by influencing the hormonal and metabolic phenotype of susceptible female fetuses later in life, may be the capital event for the development of AH in PCOS. Because human placental aromatase activity is likely to prevent any deleterious effect of maternal hyperandrogenemia on the fetus, inheritance of the maternal steroidogenic defect is the more likely culprit, even though other factors such as changes in placental steroidogenesis itself or its nutritional efflux may also be involved in the building a deregulated enzymatic pathway from utero to adult life. Anyhow, the most important issue is whether or not AH influences the cardiometabolic risk of women with PCOS. On the one hand, AH has shown a controversial relationship with carbohydrate metabolism and adiposity, and is also associated with abnormalities in blood pressure regulation in these patients. On the other hand, DHEAS may exert a beneficial effect on the lipid profile of both lean and obese patients. Lastly, available studies in women with PCOS cast doubt upon a protective role of DHEAS levels on subclinical atherosclerosis, despite opposite data from the general population. CONCLUSION: AH is frequent in patients with PCOS yet unraveling its consequences for the management of this disorder requires future longitudinal studies.


Subject(s)
Adrenal Gland Diseases/metabolism , Hyperandrogenism/genetics , Polycystic Ovary Syndrome/genetics , Adrenal Gland Diseases/etiology , Animals , Female , Humans , Hyperandrogenism/ethnology , Hyperandrogenism/metabolism , Polycystic Ovary Syndrome/ethnology , Polycystic Ovary Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...