Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
1.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36499341

RESUMEN

Glucocorticoids are steroids involved in key physiological processes such as development, metabolism, inflammatory and stress responses and are mostly used exogenously as medications to treat various inflammation-based conditions. They act via the glucocorticoid receptor (GR) expressed in most cells. Exogenous glucocorticoids can negatively impact the function of the Leydig cells in the testis, leading to decreased androgen production. However, endogenous glucocorticoids are produced by the adrenal and within the testis, but whether their action on GR in Leydig cells regulates steroidogenesis is unknown. This study aimed to define the role of endogenous GR signalling in adult Leydig cells. We developed and compared two models; an inducible Cre transgene driven by expression of the Cyp17a1 steroidogenic gene (Cyp17-iCre) that depletes GR during development and a viral vector-driven Cre (AAV9-Cre) to deplete GR in adulthood. The delivery of AAV9-Cre ablated GR in adult mouse Leydig cells depleted Leydig cell GR more efficiently than the Cyp17-iCre model. Importantly, adult depletion of GR in Leydig cells caused reduced expression of luteinising hormone receptor (Lhcgr) and of steroidogenic enzymes required for normal androgen production. These findings reveal that Leydig cell GR signalling plays a physiological role in the testis and highlight that a normal balance of glucocorticoid activity in the testis is important for steroidogenesis.


Asunto(s)
Células Intersticiales del Testículo , Receptores de Glucocorticoides , Ratones , Masculino , Animales , Células Intersticiales del Testículo/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Glucocorticoides/genética , Glucocorticoides/metabolismo , Andrógenos/metabolismo , Ratones Noqueados , Testículo/metabolismo , Expresión Génica
2.
Nucleic Acids Res ; 50(20): 11635-11653, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36399508

RESUMEN

Understanding the function of non-coding genomic sequence variants represents a challenge for biomedicine. Many diseases are products of gene-by-environment interactions with complex mechanisms. This study addresses these themes by mechanistic characterization of non-coding variants that influence gene expression only after drug or hormone exposure. Using glucocorticoid signaling as a model system, we integrated genomic, transcriptomic, and epigenomic approaches to unravel mechanisms by which variant function could be revealed by hormones or drugs. Specifically, we identified cis-regulatory elements and 3D interactions underlying ligand-dependent associations between variants and gene expression. One-quarter of the glucocorticoid-modulated variants that we identified had already been associated with clinical phenotypes. However, their affected genes were 'unmasked' only after glucocorticoid exposure and often with function relevant to the disease phenotypes. These diseases involved glucocorticoids as risk factors or therapeutic agents and included autoimmunity, metabolic and mood disorders, osteoporosis and cancer. For example, we identified a novel breast cancer risk gene, MAST4, with expression that was repressed by glucocorticoids in cells carrying the risk genotype, repression that correlated with MAST4 expression in breast cancer and treatment outcomes. These observations provide a mechanistic framework for understanding non-coding genetic variant-chemical environment interactions and their role in disease risk and drug response.


Asunto(s)
Glucocorticoides , Secuencias Reguladoras de Ácidos Nucleicos , Glucocorticoides/genética , Glucocorticoides/metabolismo , Factores de Riesgo , Humanos , Farmacogenética , Sitios de Carácter Cuantitativo
3.
Mol Cell Endocrinol ; 550: 111652, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35461977

RESUMEN

Glucocorticoids are released in response to acute aerobic exercise. The objective was to define changes in the expression of glucocorticoid target genes in skeletal muscle in response to acute aerobic exercise at different times of day. We identified glucocorticoid target genes altered in skeletal muscle by acute exercise by comparing data sets from rodents subjected to acute aerobic exercise in the light or dark cycles to data sets from C2C12 myotubes treated with glucocorticoids. The role of glucocorticoid receptor signaling and REDD1 protein in mediating gene expression was assessed in exercised mice. Changes to expression of glucocorticoid genes were greater when exercise occurred in the dark cycle. REDD1 was required for the induction of genes induced at both times of day. In all, the time of day at which aerobic exercise is conducted dictates changes to the expression of glucocorticoid target genes in skeletal muscle with REDD1 contributing to those changes.


Asunto(s)
Glucocorticoides , Músculo Esquelético , Condicionamiento Físico Animal , Animales , Ritmo Circadiano , Glucocorticoides/genética , Glucocorticoides/metabolismo , Ratones , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
J Clin Invest ; 132(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34905510

RESUMEN

Through their ability to regulate gene expression in most organs, glucocorticoid (GC) hormones influence numerous physiological processes and are therefore key regulators of organismal homeostasis. In bone, GC hormones inhibit expression of the hormone Osteocalcin for poorly understood reasons. Here, we show that in a classical endocrine feedback loop, osteocalcin in return enhanced the biosynthesis of GC as well as mineralocorticoid hormones (adrenal steroidogenesis) in rodents and primates. Conversely, inactivation of osteocalcin signaling in adrenal glands significantly impaired adrenal growth and steroidogenesis in mice. Embryo-made osteocalcin was necessary for normal Sf1 expression in fetal adrenal cells and adrenal cell steroidogenic differentiation and therefore determined the number of steroidogenic cells present in the adrenal glands of adult animals. Embryonic, not postnatal, osteocalcin also governed adrenal growth, adrenal steroidogenesis, blood pressure, electrolyte equilibrium, and the rise in circulating corticosterone levels during the acute stress response in adult offspring. This osteocalcin-dependent regulation of adrenal development and steroidogenesis occurred even in the absence of a functional hypothalamus/pituitary/adrenal axis and explains why osteocalcin administration during pregnancy promoted adrenal growth and steroidogenesis and improved the survival of adrenocorticotropic hormone signaling-deficient animals. This study reveals that a bone-derived embryonic hormone influences lifelong adrenal functions and organismal homeostasis in the mouse.


Asunto(s)
Glándulas Suprarrenales/embriología , Homeostasis , Sistema Hipotálamo-Hipofisario/embriología , Osteocalcina/metabolismo , Sistema Hipófiso-Suprarrenal/embriología , Transducción de Señal , Animales , Femenino , Glucocorticoides/genética , Glucocorticoides/metabolismo , Macaca mulatta , Ratones , Ratones Noqueados , Osteocalcina/genética
5.
Eur J Endocrinol ; 186(2): 297-308, 2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-34914631

RESUMEN

OBJECTIVE: Cushing's syndrome represents a state of excessive glucocorticoids related to glucocorticoid treatments or to endogenous hypercortisolism. Cushing's syndrome is associated with high morbidity, with significant inter-individual variability. Likewise, adrenal insufficiency is a life-threatening condition of cortisol deprivation. Currently, hormone assays contribute to identify Cushing's syndrome or adrenal insufficiency. However, no biomarker directly quantifies the biological glucocorticoid action. The aim of this study was to identify such markers. DESIGN: We evaluated whole blood DNA methylome in 94 samples obtained from patients with different glucocorticoid states (Cushing's syndrome, eucortisolism, adrenal insufficiency). We used an independent cohort of 91 samples for validation. METHODS: Leukocyte DNA was obtained from whole blood samples. Methylome was determined using the Illumina methylation chip array (~850 000 CpG sites). Both unsupervised (principal component analysis) and supervised (Limma) methods were used to explore methylome profiles. A Lasso-penalized regression was used to select optimal discriminating features. RESULTS: Whole blood methylation profile was able to discriminate samples by their glucocorticoid status: glucocorticoid excess was associated with DNA hypomethylation, recovering within months after Cushing's syndrome correction. In Cushing's syndrome, an enrichment in hypomethylated CpG sites was observed in the region of FKBP5 gene locus. A methylation predictor of glucocorticoid excess was built on a training cohort and validated on two independent cohorts. Potential CpG sites associated with the risk for specific complications, such as glucocorticoid-related hypertension or osteoporosis, were identified, needing now to be confirmed on independent cohorts. CONCLUSIONS: Whole blood DNA methylome is dynamically impacted by glucocorticoids. This biomarker could contribute to better assessment of glucocorticoid action beyond hormone assays.


Asunto(s)
Síndrome de Cushing/genética , Metilación de ADN/genética , ADN/sangre , Epigenoma/genética , Glucocorticoides/sangre , Glucocorticoides/genética , Adolescente , Insuficiencia Suprarrenal/sangre , Insuficiencia Suprarrenal/genética , Adulto , Anciano , Biomarcadores/sangre , Islas de CpG/genética , Síndrome de Cushing/sangre , Femenino , Humanos , Hidrocortisona/análisis , Hidrocortisona/sangre , Hidrocortisona/orina , Leucocitos/química , Masculino , Persona de Mediana Edad , Saliva/química , Proteínas de Unión a Tacrolimus/genética
6.
FASEB J ; 36(1): e22130, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34959259

RESUMEN

This study aimed to investigate the causal relationship between chronic ingestion of a high-fat diet (HFD)-induced secretion of glucocorticoids (GCs) and the development of non-alcoholic fatty liver disease (NAFLD). We have produced a strain of transgenic mice (termed L/L mice) that have normal levels of circulating corticosterone (CORT), the major type of GCs in rodents, but unlike wild-type (WT) mice, their circulating CORT was not affected by HFD. Compared to WT mice, 12-week HFD-induced fatty liver was less pronounced with higher plasma levels of triglycerides in L/L mice. These changes were reversed by CORT supplement to L/L mice. By analyzing a sort of lipid metabolism-related proteins, we found that expressions of the hepatic cluster of differentiation 36 (CD36) were upregulated by HFD-induced CORT and involved in CORT-mediated fatty liver. Dexamethasone, an agonist of the glucocorticoid receptor (GR), upregulated expressions of CD36 in HepG2 hepatocytes and facilitated lipid accumulation in the cells. In conclusion, the fat ingestion-induced release of CORT contributes to NAFLD. This study highlights the pathogenic role of CORT-mediated upregulation of hepatic CD 36 in diet-induced NAFLD.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Glucocorticoides/sangre , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Triglicéridos/sangre , Animales , Glucocorticoides/genética , Células Hep G2 , Humanos , Ratones , Ratones Mutantes , Enfermedad del Hígado Graso no Alcohólico/genética , Triglicéridos/genética
7.
Cells ; 10(12)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34943949

RESUMEN

Glucocorticoids (GCs) represent a well-known class of lipophilic steroid hormones biosynthesised, with a circadian rhythm, by the adrenal glands in humans and by the inter-renal tissue in teleost fish (e.g., zebrafish). GCs play a key role in the regulation of numerous physiological processes, including inflammation, glucose, lipid, protein metabolism and stress response. This is achieved through binding to their cognate receptor, GR, which functions as a ligand-activated transcription factor. Due to their potent anti-inflammatory and immune-suppressive action, synthetic GCs are broadly used for treating pathological disorders that are very often linked to hypoxia (e.g., rheumatoid arthritis, inflammatory, allergic, infectious, and autoimmune diseases, among others) as well as to prevent graft rejections and against immune system malignancies. However, due to the presence of adverse effects and GC resistance their therapeutic benefits are limited in patients chronically treated with steroids. For this reason, understanding how to fine-tune GR activity is crucial in the search for novel therapeutic strategies aimed at reducing GC-related side effects and effectively restoring homeostasis. Recent research has uncovered novel mechanisms that inhibit GR function, thereby causing glucocorticoid resistance, and has produced some surprising new findings. In this review we analyse these mechanisms and focus on the crosstalk between GR and HIF signalling. Indeed, its comprehension may provide new routes to develop novel therapeutic targets for effectively treating immune and inflammatory response and to simultaneously facilitate the development of innovative GCs with a better benefits-risk ratio.


Asunto(s)
Glucocorticoides/genética , Inflamación/genética , Errores Innatos del Metabolismo/genética , Receptores de Glucocorticoides/deficiencia , Receptores de Glucocorticoides/genética , Animales , Regulación de la Expresión Génica/genética , Glucocorticoides/metabolismo , Homeostasis/genética , Humanos , Inflamación/metabolismo , Inflamación/patología , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Unión Proteica/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/genética , Pez Cebra/genética
8.
Mol Neurobiol ; 58(11): 5837-5856, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34409559

RESUMEN

Prenatal stress (PS) is a major risk factor for the development of emotional disorders in adulthood that may be mediated by an altered hypothalamic-pituitary-adrenal axis response to stress. Although the early onset of stress-related disorders is recognized as a major public health problem, to date, there are relatively few studies that have examined the incidence of early-life stressors in younger individuals. In this study, we assessed PS impact on the stress-coping response of juvenile offspring in behavioral tests and in the induced molecular changes in the hippocampus. Furthermore, we assessed if pregnancy stress could be driving changes in patterns of maternal behavior during early lactation. We found that PS modified stress-coping abilities of both sex offspring. In the hippocampus, PS increased the expression of bdnf-IV and crfr1 and induced sex difference changes on glucocorticoids and BDNF mRNA receptor levels. PS changed the hippocampal epigenetic landscape mainly in male offspring. Stress during pregnancy enhanced pup-directed behavior of stressed dams. Our study indicates that exposure to PS, in addition to enhanced maternal behavior, induces dynamic neurobehavioral variations at juvenile ages of the offspring that should be considered adaptive or maladaptive, depending on the characteristics of the confronting environment. Our present results highlight the importance to further explore risk factors that appear early in life that will be important to allow timely prevention strategies to later vulnerability to stress-related disorders.


Asunto(s)
Adaptación Psicológica , Complicaciones del Embarazo , Efectos Tardíos de la Exposición Prenatal , Restricción Física , Estrés Fisiológico , Estrés Psicológico , Animales , Femenino , Masculino , Embarazo , Ratas , Ansiedad/etiología , Ansiedad/genética , Ansiedad/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Corticosterona/sangre , Hormona Liberadora de Corticotropina/biosíntesis , Hormona Liberadora de Corticotropina/genética , Prueba de Laberinto Elevado , Regulación de la Expresión Génica , Glucocorticoides/biosíntesis , Glucocorticoides/genética , Hipocampo/embriología , Hipocampo/fisiología , Sistema Hipotálamo-Hipofisario/embriología , Sistema Hipotálamo-Hipofisario/fisiopatología , Lactancia/fisiología , Lactancia/psicología , Conducta Materna , Sistema Hipófiso-Suprarrenal/embriología , Sistema Hipófiso-Suprarrenal/fisiopatología , Complicaciones del Embarazo/fisiopatología , Complicaciones del Embarazo/psicología , Ratas Wistar , Receptor trkB/biosíntesis , Receptor trkB/genética , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Glucocorticoides/biosíntesis , Receptores de Glucocorticoides/genética , Restricción Física/efectos adversos , Caracteres Sexuales , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología , Natación
9.
Int J Mol Sci ; 22(16)2021 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-34445209

RESUMEN

Glucocorticoids (GCs) are hormones that aid the body under stress by regulating glucose and free fatty acids. GCs maintain energy homeostasis in multiple tissues, including those in the liver and skeletal muscle, white adipose tissue (WAT), and brown adipose tissue (BAT). WAT stores energy as triglycerides, while BAT uses fatty acids for heat generation. The multiple genomic and non-genomic pathways in GC signaling vary with exposure duration, location (adipose tissue depot), and species. Genomic effects occur directly through the cytosolic GC receptor (GR), regulating the expression of proteins related to lipid metabolism, such as ATGL and HSL. Non-genomic effects act through mechanisms often independent of the cytosolic GR and happen shortly after GC exposure. Studying the effects of GCs on adipose tissue breakdown and generation (lipolysis and adipogenesis) leads to insights for treatment of adipose-related diseases, such as obesity, coronary disease, and cancer, but has led to controversy among researchers, largely due to the complexity of the process. This paper reviews the recent literature on the genomic and non-genomic effects of GCs on WAT and BAT lipolysis and proposes research to address the many gaps in knowledge related to GC activity and its effects on disease.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Genómica , Glucocorticoides , Lipogénesis , Lipólisis , Animales , Glucocorticoides/genética , Glucocorticoides/metabolismo , Humanos
10.
Sci Rep ; 11(1): 12515, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131228

RESUMEN

The glucocorticoid receptor is a key regulator of essential physiological processes, which under the control of the Hsp90 chaperone machinery, binds to steroid hormones and steroid-like molecules and in a rather complicated and elusive response, regulates a set of glucocorticoid responsive genes. We here examine a human glucocorticoid receptor variant, harboring a point mutation in the last C-terminal residues, L773P, that was associated to Primary Generalized Glucocorticoid Resistance, a condition originating from decreased affinity to hormone, impairing one or multiple aspects of GR action. Using in vitro and in silico methods, we assign the conformational consequences of this mutation to particular GR elements and report on the altered receptor properties regarding its binding to dexamethasone, a NCOA-2 coactivator-derived peptide, DNA, and importantly, its interaction with the chaperone machinery of Hsp90.


Asunto(s)
Glucocorticoides/genética , Proteínas HSP90 de Choque Térmico/genética , Conformación Molecular/efectos de los fármacos , Receptores de Glucocorticoides/genética , Animales , ADN/genética , Dexametasona/farmacología , Glucocorticoides/química , Proteínas HSP90 de Choque Térmico/ultraestructura , Humanos , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Chaperonas Moleculares/genética , Chaperonas Moleculares/ultraestructura , Coactivador 2 del Receptor Nuclear/química , Coactivador 2 del Receptor Nuclear/genética , Péptidos/genética , Mutación Puntual/genética , Unión Proteica/genética , Receptores de Glucocorticoides/deficiencia , Receptores de Glucocorticoides/ultraestructura
11.
J Steroid Biochem Mol Biol ; 206: 105788, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33227378

RESUMEN

Congenital lipoid adrenal hyperplasia (LCAH), as the most severe form of congenital adrenal hyperplasia (CAH), is caused by mutations in the steroidogenic acute regulatory protein (STAR). Affected patients were typically characterized by adrenal insufficiency in the first year of life and present with female external genitalia regardless of karyotype. Non-classic LCAH patients usually present from 2 to 4 years old with glucocorticoid deficiency and mild mineralocorticoid deficiency, even develop naturally masculinized external genitalia at birth when they have 46,XY karyotype. We described thirty patients from unrelated Chinese families, including three non-classic LCAH ones. Four novel mutations were reported, including c.556A > G, c.179-15G > T, c.695delG and c.306 + 3_c.306 + 6delAAGT. The c.772C > T is the most common STAR mutation in Chinese population, suggesting a possibility of founder effect. Enzymatic activity assay combined with clinical characteristics showed a good genotype-phenotype correlation in this study. Residual STAR activity more than 20 % may be correlated with non-classic LCAH phenotype. We support the perspective that onset age may be affected by multiple factors and masculinization should be the main weighting factor for diagnosis of non-classic LCAH. Compared with 46,XX LCAH patients, less 46,XY ones were found in our report. A less comprehensive inspection and an easy diagnosis due to classical phenotype both would reduce the possibility of 46,XY LCAH patients to be referred to specialists or geneticists.


Asunto(s)
Hiperplasia Suprarrenal Congénita/genética , Insuficiencia Suprarrenal/genética , Trastorno del Desarrollo Sexual 46,XY/genética , Glucocorticoides/genética , Fosfoproteínas/genética , Hiperplasia Suprarrenal Congénita/epidemiología , Hiperplasia Suprarrenal Congénita/patología , Insuficiencia Suprarrenal/patología , Preescolar , China/epidemiología , Trastorno del Desarrollo Sexual 46,XY/epidemiología , Trastorno del Desarrollo Sexual 46,XY/patología , Femenino , Glucocorticoides/deficiencia , Humanos , Cariotipo , Masculino , Mutación/genética , Fenotipo
12.
Arch Gynecol Obstet ; 303(2): 427-434, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32886236

RESUMEN

PURPOSE: The aim of this study was to examine associations between single nucleotide polymorphisms (SNPs) that tag genetic variation in the glucocorticoid pathways (particularly in maternal genes FKBP5, NR3C1, and CRHR1) and birth weight. METHODS: The Franconian Maternal Health Evaluation Study (FRAMES) recruited healthy pregnant women prospectively for the assessment of maternal and fetal health. Germline DNA was collected from 375 pregnant women. Nine SNPs in the above-mentioned genes were genotyped. After reconstruction of haplotypes for each gene, a linear regression model was applied to the data to describe the association between haplotypes and birth weight. RESULTS: Female sex in the newborn (compared to male) was associated with lower birth weight, whereas a later week of gestation, higher body mass index pre-pregnancy, and higher parity were associated with higher birth weight. No association with birthweight was shown for the haplotypes of the selected SNPs. CONCLUSIONS: In this cohort of healthy unselected pregnant women, the analyzed candidate haplotypes in FKBP5, NR3C1, and CRHR1 did not show any association with birth weight. This might be in line with several other studies that have found no influence of fetal polymorphisms in the glucocorticoid receptor gene or triggers of the maternal HPA axis such as stress and psychosocial problems on birth weight. However, the small sample size in this study and the lack of consideration of individual risk factors and levels of stress in this cohort needs to be taken into account when interpreting the results.


Asunto(s)
Peso al Nacer , Glucocorticoides/genética , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Glucocorticoides/genética , Proteínas de Unión a Tacrolimus/genética , Adulto , Estudios de Cohortes , Femenino , Genotipo , Haplotipos , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Recién Nacido , Masculino , Sistema Hipófiso-Suprarrenal/metabolismo , Polimorfismo de Nucleótido Simple , Embarazo , Receptores de Glucocorticoides/metabolismo
13.
Leukemia ; 35(7): 1964-1975, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33168949

RESUMEN

The oncogenic protein Bcr-Abl has two major isoforms, p190Bcr-Abl and p210Bcr-Abl. While p210Bcr-Abl is the hallmark of chronic myeloid leukemia (CML), p190Bcr-Abl occurs in the majority of Philadelphia-positive acute lymphoblastic leukemia (Ph + ALL) patients. In CML, p190Bcr-Abl occurs in a minority of patients associating with distinct hematological features and inferior outcomes, yet the pathogenic role of p190Bcr-Abl and potential targeting therapies are largely uncharacterized. We employed next generation sequencing, phospho-proteomic profiling, and drug sensitivity testing to characterize p190Bcr-Abl in CML and hematopoietic progenitor cell line models (Ba/f3 and HPC-LSK). p190Bcr-Abl CML patients demonstrated poor response to imatinib and frequent mutations in epigenetic modifiers genes. In contrast with p210Bcr-Abl, p190Bcr-Abl exhibited specific transcriptional upregulation of interferon, interleukin-1 receptor, and P53 signaling pathways, associated with hyperphosphorylation of relevant signaling molecules including JAK1/STAT1 and PAK1 in addition to Src hyperphosphorylation. Comparable to p190Bcr-Abl CML patients, p190Bcr-Abl cell lines demonstrated similar transcriptional and phospho-signaling signatures. With the drug sensitivity screening we identified targeted drugs with specific activity in p190Bcr-Abl cell lines including IAP-, PAK1-, and Src inhibitors and glucocorticoids. Our results provide novel insights into the mechanisms underlying the distinct features of p190Bcr-Abl CML and promising therapeutic targets for this high-risk patient group.


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Transducción de Señal/genética , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica/métodos , Glucocorticoides/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Oncogenes/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteómica/métodos , Transcripción Genética/genética , Regulación hacia Arriba/genética
14.
Addict Biol ; 26(3): e12978, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33142367

RESUMEN

Genetically-selected Marchigian Sardinian alcohol-preferring (msP) rats display comorbid symptoms of increased alcohol preference and elevated anxiety-like behavior. Heightened stress sensitivity in msPs is influenced by genetic polymorphisms of the corticotropin-releasing factor receptor in the central nucleus of the amygdala (CeA), as well as reduced influence of anti-stress mechanisms that normally constrain the stress response. Given this propensity for stress dysregulation, in this study, we expand on the possibility that msPs may display differences in neuroendocrine processes that normally terminate the stress response. We utilized behavioral, biochemical, and molecular assays to compare basal and restraint stress-induced changes in the hypothalamic-pituitary-adrenal (HPA) axis of male and female msPs relative to their nonselected Wistar counterparts. The results showed that msPs display deficits in marble-burying behavior influenced by environmental factors and procedures that modulate arousal states in a sex-dependent manner. Whereas male msPs display evidence of dysregulated neuroendocrine function (higher adrenocorticotropic hormone levels and subthreshold reductions in corticosterone), females display restraint-induced elevations in corticosterone levels that were persistently higher in msPs. A dexamethasone challenge reduced the circulation of these stress hormones, although the reduction in corticosterone was generally attenuated in msP versus Wistar rats. Finally, we found evidence of diminished stress-induced glucocorticoid receptor (GR) phosphorylation in the hypothalamic paraventricular nucleus of msPs, as well as innate increases in phosphorylated GR levels in the CeA of male msPs. Collectively, these findings suggest that negative feedback processes regulating HPA responsiveness are diminished in msP rats, possibly underlying differences in the expression of anxiety-like behaviors.


Asunto(s)
Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Restricción Física , Consumo de Bebidas Alcohólicas/genética , Animales , Ansiedad/genética , Corticosterona/sangre , Retroalimentación Fisiológica , Femenino , Glucocorticoides/genética , Hipotálamo/metabolismo , Masculino , Ratas , Ratas Wistar , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
15.
Adv Neonatal Care ; 20(5): 392-400, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32868589

RESUMEN

BACKGROUND: Preterm infants experience a multitude of prenatal and postnatal stressors, resulting in cumulative stress exposure, which may jeopardize the timely attainment of developmental milestones, such as achieving oral feeding. Up to 70% of preterm infants admitted to the neonatal intensive care unit experience challenges while initiating oral feeding. Oral feeding skills require intact neurobehavioral development. Evolving evidence demonstrates that cumulative stress exposure results in epigenetic modification of glucocorticoid-related genes. Epigenetics is a field of study that focuses on phenotypic changes that do not involve alterations in the DNA sequence. Epigenetic modification of glucocorticoid-related genes alters cortisol reactivity to environmental stimuli, which may influence neurobehavioral development, and is the essence of the evolving field of Preterm Behavioral Epigenetics. It is plausible that early-life cumulative stress exposure and the ensuing epigenetic modification of glucocorticoid-related genes impair neurobehavioral development required for achievement of oral feeding skills in preterm infants. PURPOSE: The purpose of this article is to build upon the evolving science of Preterm Behavioral Epigenetics and present a conceptual model that explicates how cumulative stress exposure affects neurobehavioral development and achievement of oral feeding skills through epigenetic modification of glucocorticoid-related genes. METHODS/RESULTS: Using the Preterm Behavioral Epigenetics framework and supporting literature, we present a conceptual model in which early-life cumulative stress exposure, reflected by DNA methylation of glucocorticoid-related genes and altered cortisol reactivity, disrupts neurobehavioral development critical for achievement of oral feeding skills. IMPLICATIONS FOR PRACTICE AND RESEARCH: Future investigations guided by the proposed conceptual model will benefit preterm infant outcomes by introducing epigenetic-based approaches to assess and monitor preterm infant oral feeding skills. Furthermore, the proposed model can guide future investigations that develop and test epigenetic protective interventions to improve clinical outcomes, representing an innovation in neonatal care.


Asunto(s)
Conducta Alimentaria/fisiología , Glucocorticoides , Estrés Fisiológico , Estrés Psicológico/fisiopatología , Epigenómica , Femenino , Glucocorticoides/genética , Humanos , Hidrocortisona/genética , Recién Nacido , Recien Nacido Prematuro , Masculino , Modelos Teóricos , Embarazo
16.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629782

RESUMEN

Breast cancer (BC) patients experience increased stress with elevated cortisol levels, increasing risk of cancer recurrence. Cortisol binds to a cytoplasmic receptor, glucocorticoid receptor (GR) encoded by GR gene (NR3C1). We hypothesized that not only cancer cells, but even immune cells in the tumor microenvironment (TME) may contribute to GR expression in bulk tumor and influence prognosis. To test this, mRNA expression data was accessed from METABRIC and TCGA. "High" and "low" expression was based on highest and lowest quartiles of NR3C1 gene expression, respectively. Single-cell sequencing data were obtained from GSE75688 and GSE114725 cohorts. Computer algorithms CIBERSORT, Gene Set Enrichment Analysis and TIMER were used. GR-high BC has better median disease-free and disease-specific survival. Single cell sequencing data showed higher GR expression on immune cells compared to cancer and stromal cells. Positive correlation between GR-high BC and CD8+ T-cells was noted. In GR-high tumors, higher cytolytic activity (CYT) with decreased T-regulatory and T-follicular helper cells was observed. High GR expression was associated with lower proliferation index Ki67, enriched in IL-2_STAT5, apoptosis, KRAS, TGF-ß signaling, and epithelial-to-mesenchymal transition. Immune cells significantly contribute to GR expression of bulk BC. GR-high BC has a favorable TME with higher CYT with favorable outcomes.


Asunto(s)
Neoplasias de la Mama/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Receptores de Glucocorticoides/genética , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/fisiología , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Glucocorticoides/genética , Glucocorticoides/metabolismo , Humanos , Recurrencia Local de Neoplasia/genética , Pronóstico , Receptores de Estrógenos/metabolismo , Receptores de Glucocorticoides/inmunología , Receptores de Glucocorticoides/metabolismo , Microambiente Tumoral/inmunología
17.
Nucleic Acids Res ; 48(15): 8393-8407, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32619221

RESUMEN

The glucocorticoid receptor is an important immunosuppressive drug target and metabolic regulator that acts as a ligand-gated transcription factor. Generally, GR's anti-inflammatory effects are attributed to the silencing of inflammatory genes, while its adverse effects are ascribed to the upregulation of metabolic targets. GR binding directly to DNA is proposed to activate, whereas GR tethering to pro-inflammatory transcription factors is thought to repress transcription. Using mice with a point mutation in GR's zinc finger, that still tether via protein-protein interactions while being unable to recognize DNA, we demonstrate that DNA binding is essential for both transcriptional activation and repression. Performing ChIP-Seq, RNA-Seq and proteomics under inflammatory conditions, we show that DNA recognition is required for the assembly of a functional co-regulator complex to mediate glucocorticoid responses. Our findings may contribute to the development of safer immunomodulators with fewer side effects.


Asunto(s)
Proteínas de Unión al ADN/genética , ADN/genética , Inflamación/genética , Receptores de Glucocorticoides/genética , Animales , ADN/metabolismo , Regulación de la Expresión Génica/genética , Glucocorticoides/genética , Glucocorticoides/metabolismo , Humanos , Inflamación/patología , Ratones , Dominios y Motivos de Interacción de Proteínas/genética , RNA-Seq , Activación Transcripcional/genética
18.
Mol Diagn Ther ; 24(4): 473-485, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32607951

RESUMEN

BACKGROUND: Generalized glucocorticoid resistance is characterized by impaired cortisol signaling, resulting from mutations in the NR3C1 gene coding the human glucocorticoid receptor (hGR). Most of the pathogenic hGR variants are identified in the ligand-binding domain (LBD). However, we report a new case associated with a novel NR3C1 pathogenic variant in the N-terminal domain (NTD) of the hGR. METHODS: The index case was an Italian 31-year-old woman with a history of chronic fatigue, anxiety, hirsutism, irregular menstrual cycles, and infertility, but no clinical manifestations suggestive of Cushing's syndrome. Standard clinical methods were used to assess the patient's hypothalamic-pituitary-adrenal axis. Molecular analysis of the NR3C1 gene was performed by Sanger sequencing. In addition, we perform an extensive survey of all clinical pathogenic variants modifying the whole sequence of the NR3C1 gene. RESULTS: Endocrinologic evaluation revealed elevated serum cortisol, plasma adrenocorticotropic hormone, and androstenedione concentration and increased urinary free cortisol excretion. Morning serum cortisol levels remained elevated and were not suppressed during a 2-day, 2-mg dexamethasone suppression test. The identification of the novel p.(Glu123Ter) NR3C1 variant confirmed the diagnosis of glucocorticoid resistance. CONCLUSION: Our findings improve the understanding of NR3C1 mutational spectrum in patients affected by glucocorticoid resistance syndrome and contribute to precise diagnosis and genetic counseling.


Asunto(s)
Variación Genética/genética , Glucocorticoides/genética , Receptores de Glucocorticoides/genética , Adulto , Síndrome de Cushing/genética , Femenino , Humanos
19.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326396

RESUMEN

Gonadotropin-releasing hormone (GnRH) is essential for the initiation and maintenance of reproductive functions in vertebrates. To date, three distinct paralogue lineages, GnRH1, GnRH2, and GnRH3, have been identified with different functions and regulatory mechanisms. Among them, hypothalamic GnRH1 neurons are classically known as the hypophysiotropic form that is regulated by estrogen feedback. However, the mechanism of action underlying the estrogen-dependent regulation of GnRH1 has been debated, mainly due to the coexpression of low levels of estrogen receptor (ER) genes. In addition, the role of sex steroids in the modulation of GnRH2 and GnRH3 neurons has not been fully elucidated. Using single-cell real-time PCR, we revealed the expression of genes for estrogen, androgen, glucocorticoid, thyroid, and xenobiotic receptors in GnRH1, GnRH2, and GnRH3 neurons in the male Nile tilapia Oreochromis niloticus. We further quantified expression levels of estrogen receptor genes (ERα, ERß, and ERγ) in three GnRH neuron types in male tilapia of two different social statuses (dominant and subordinate) at the single cell level. In dominant males, GnRH1 mRNA levels were positively proportional to ERγ mRNA levels, while in subordinate males, GnRH2 mRNA levels were positively proportional to ERß mRNA levels. These results indicate that variations in the expression of nuclear receptors (and possibly steroid sensitivities) among individual GnRH cells may facilitate different physiological processes, such as the promotion of reproductive activities through GnRH1 neurons, and the inhibition of feeding and sexual behaviors through GnRH2 neurons.


Asunto(s)
Agresión/fisiología , Cíclidos/metabolismo , Hormona Liberadora de Gonadotropina/sangre , Neuronas/metabolismo , Distancia Psicológica , Receptores Citoplasmáticos y Nucleares/metabolismo , Esteroides/metabolismo , Andrógenos/sangre , Andrógenos/genética , Andrógenos/metabolismo , Animales , Cíclidos/genética , Estrógenos/sangre , Estrógenos/genética , Estrógenos/metabolismo , Glucocorticoides/sangre , Glucocorticoides/genética , Glucocorticoides/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Masculino , Precursores de Proteínas/sangre , Precursores de Proteínas/metabolismo , Ácido Pirrolidona Carboxílico/análogos & derivados , Ácido Pirrolidona Carboxílico/sangre , Ácido Pirrolidona Carboxílico/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Análisis de la Célula Individual , Esteroides/sangre , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Hormonas Tiroideas/sangre , Hormonas Tiroideas/genética , Hormonas Tiroideas/metabolismo , Xenobióticos/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-31962186

RESUMEN

Stress reactivity and glucocorticoid signaling alterations are reported in mouse models of autism spectrum disorder (ASD). BALB/c mice display decreased locomotor activity in the presence of stimulus mice and spend less time exploring enclosed stimulus mice; this mouse strain has been validated as an ASD model. VU0410120, a glycine type 1 transporter (GlyT1) inhibitor, improved sociability in BALB/c mice, consistent with data that NMDA Receptor (NMDAR) activation regulates sociability, and the endogenous tone of NMDAR-mediated neurotransmission is altered in this strain. Effects of a prosocial dose of VU0410120 on conspecific-provoked immobility, and relationships between conspecific-provoked immobility and corticosterone response were explored. VU0410120-treated BALB/c mice showed reduced immobility in the presence of conspecifics and increased the conspecific-provoked corticosterone response. However, the intensity of conspecific-provoked immobility in VU0410120-treated BALB/c mice did not differ as a function of corticosterone response. Expression profiles of 88 glucocorticoid signaling associated genes within frontal cortex and hippocampus were examined. BALB/c mice resistant to prosocial effects of VU0410120 had increased mRNA expression of Ddit4, a negative regulator of mTOR signaling. Dysregulated mTOR signaling activity is a convergent finding in several monogenic syndromic forms of ASD. Prosocial effects of VU0410120 in the BALB/c strain may be related to regulatory influences of NMDAR-activation on mTOR signaling activity. Because corticosterone response is a marker of social stress, the current data suggest that the stressfulness of a social encounter alone may not be the sole determinant of increased immobility in BALB/c mice; this strain may also display an element of social disinterest.


Asunto(s)
Corteza Cerebral/metabolismo , Corticosterona/sangre , Glucocorticoides/biosíntesis , Proteínas de Transporte de Glicina en la Membrana Plasmática/antagonistas & inhibidores , Hipocampo/metabolismo , Inmovilización/fisiología , Animales , Benzamidas/farmacología , Corteza Cerebral/efectos de los fármacos , Expresión Génica , Glucocorticoides/genética , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA