Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 113(8): 2146-51, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858405

RESUMEN

Epigenetic modifications play significant roles in adaptive evolution. The tumor suppressor p53, well known for controlling cell fate and maintaining genomic stability, is much less known as a master gene in environmental adaptation involving methylation modifications. The blind subterranean mole rat Spalax eherenbergi superspecies in Israel consists of four species that speciated peripatrically. Remarkably, the northern Galilee species Spalax galili (2n = 52) underwent adaptive ecological sympatric speciation, caused by the sharply divergent chalk and basalt ecologies. This was demonstrated by mitochondrial and nuclear genomic evidence. Here we show that the expression patterns of the p53 regulatory pathway diversified between the abutting sympatric populations of S. galili in sharply divergent chalk-basalt ecologies. We identified higher methylation on several sites of the p53 promoter in the population living in chalk soil (chalk population). Site mutagenesis showed that methylation on these sites linked to the transcriptional repression of p53 involving Cut-Like Homeobox 1 (Cux1), paired box 4 (Pax 4), Pax 6, and activator protein 1 (AP-1). Diverse expression levels of p53 between the incipiently sympatrically speciating chalk-basalt abutting populations of S. galili selectively affected cell-cycle arrest but not apoptosis. We hypothesize that methylation modification of p53 has adaptively shifted in supervising its target genes during sympatric speciation of S. galili to cope with the contrasting environmental stresses of the abutting divergent chalk-basalt ecologies.


Asunto(s)
Metilación de ADN , Genes p53 , Spalax/genética , Spalax/metabolismo , Adaptación Biológica , Animales , Carbonato de Calcio , Puntos de Control del Ciclo Celular/genética , Ecosistema , Evolución Molecular , Expresión Génica , Especiación Genética , Genética de Población , Pulmón/metabolismo , Regiones Promotoras Genéticas , Silicatos , Suelo , Spalax/clasificación , Simpatría
2.
Proc Natl Acad Sci U S A ; 111(36): 13199-204, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25146699

RESUMEN

Cerebral edema is a potentially life-threatening illness, but knowledge of its underlying mechanisms is limited. Here we report that hypobaric hypoxia induces rat cerebral edema and neuronal apoptosis and increases the expression of corticotrophin releasing factor (CRF), CRF receptor type 1 (CRFR1), aquaporin-4 (AQP4), and endothelin-1 (ET-1) in the cortex. These effects, except for the increased expression of CRF itself, could all be blocked by pretreatment with an antagonist of the CRF receptor CRFR1. We also show that, in cultured primary astrocytes: (i) both CRFR1 and AQP4 are expressed; (ii) exogenous CRF, acting through CRFR1, triggers signaling of cAMP/PKA, intracellular Ca(2+), and PKCε; and (iii) the up-regulated cAMP/PKA signaling contributes to the phosphorylation and expression of AQP4 to enhance water influx into astrocytes and produces an up-regulation of ET-1 expression. Finally, using CHO cells transfected with CRFR1(+) and AQP4(+), we show that transfected CRFR1(+) contributes to edema via transfected AQP4(+). In conclusion, hypoxia triggers cortical release of CRF, which acts on CRFR1 to trigger signaling of cAMP/PKA in cortical astrocytes, leading to activation of AQP4 and cerebral edema.


Asunto(s)
Acuaporina 4/metabolismo , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Hipoxia/complicaciones , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Animales , Apoptosis/genética , Acuaporina 4/genética , Astrocitos/metabolismo , Edema Encefálico/patología , Células CHO , Hormona Liberadora de Corticotropina/metabolismo , Cricetinae , Cricetulus , Endotelina-1/metabolismo , Hipoxia/metabolismo , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal , Transfección , Regulación hacia Arriba/genética
3.
J Neuroinflammation ; 13(1): 63, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26968975

RESUMEN

BACKGROUND: High-altitude cerebral edema (HACE) is the severe type of acute mountain sickness (AMS) and life threatening. A subclinical inflammation has been speculated, but the exact mechanisms underlying the HACE are not fully understood. METHODS: Human volunteers ascended to high altitude (3860 m, 2 days), and rats were exposed to hypoxia in a hypobaric chamber (5000 m, 2 days). Human acute mountain sickness was evaluated by the Lake Louise Score (LLS), and plasma corticotrophin-releasing hormone (CRH) and cytokines TNF-α, IL-1ß, and IL-6 were measured in rats and humans. Subsequently, rats were pre-treated with lipopolysaccharide (LPS, intraperitoneal (ip) 4 mg/kg, 11 h) to induce inflammation prior to 1 h hypoxia (7000 m elevation). TNF-α, IL-1ß, IL-6, nitric oxide (NO), CRH, and aquaporin-4 (AQP4) and their gene expression, Evans blue, Na(+)-K(+)-ATPase activity, p65 translocation, and cell swelling were measured in brain by ELISA, Western blotting, Q-PCR, RT-PCR, immunohistochemistry, and transmission electron micrography. MAPKs, NF-κB pathway, and water permeability of primary astrocytes were demonstrated. All measurements were performed with or without LPS challenge. The release of NO, TNF-α, and IL-6 in cultured primary microglia by CRH stimulation with or without PDTC (NF-κB inhibitor) or CP154,526 (CRHR1 antagonist) were measured. RESULTS: Hypobaric hypoxia enhanced plasma TNF-α, IL-1ß, and IL-6 and CRH levels in human and rats, which positively correlated with AMS. A single LPS injection (ip, 4 mg/kg, 12 h) into rats increased TNF-α and IL-1ß levels in the serum and cortex, and AQP4 and AQP4 mRNA expression in cortex and astrocytes, and astrocyte water permeability but did not cause brain edema. However, LPS treatment 11 h prior to 1 h hypoxia (elevation, 7000 m) challenge caused cerebral edema, which was associated with activation of NF-κB and MAPKs, hypoxia-reduced Na(+)-K(+)-ATPase activity and blood-brain barrier (BBB) disruption. Both LPS and CRH stimulated TNF-α, IL-6, and NO release in cultured rat microglia via NF-κB and cAMP/PKA. CONCLUSIONS: Preexisting systemic inflammation plus a short severe hypoxia elicits cerebral edema through upregulated AQP4 and water permeability by TLR4 and CRH/CRHR1 signaling. This study revealed that both infection and hypoxia can cause inflammatory response in the brain. Systemic inflammation can facilitate onset of hypoxic cerebral edema through interaction of astrocyte and microglia by activation of TLR4 and CRH/CRHR1 signaling. Anti-inflammatory agents and CRHR1 antagonist may be useful for prevention and treatment of AMS and HACE.


Asunto(s)
Mal de Altura/fisiopatología , Edema Encefálico/etiología , Edema Encefálico/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Inflamación/fisiopatología , Adolescente , Animales , Barrera Hematoencefálica/efectos de los fármacos , Agua Corporal/metabolismo , Permeabilidad de la Membrana Celular , Hormona Liberadora de Corticotropina/sangre , Citocinas/sangre , Voluntarios Sanos , Humanos , Lipopolisacáridos , Masculino , Ratas , Ratas Sprague-Dawley , Adulto Joven
4.
Proc Natl Acad Sci U S A ; 110(51): 20639-44, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297887

RESUMEN

Mutational changes in p53 correlate well with tumorigenesis. Remarkably, however, relatively little is known about the role that p53 variations may play in environmental adaptation. Here we report that codon asparagine-104 (104N) and glutamic acid-104 (104E), respectively, of the p53 gene in the wild zokor (Myospalax baileyi) and root vole (Microtus oeconomus) are adaptively variable, meeting the environmental stresses of the Tibetan plateau. They differ from serine-104 (104S) seen in other rodents, including the lowland subterranean zokor Myospalax cansus, and from serine 106 (106S) in humans. Based on site-directed mutational analysis in human cell lines, the codon 104N variation in M. baileyi is responsible for the adaptive balance of the transactivation of apoptotic genes under hypoxia, cold, and acidic stresses. The 104E p53 variant in Microtus oeconomus suppresses apoptotic gene transactivation and cell apoptosis. Neither 104N nor 104E affects the cell-cycle genes. We propose that these variations in p53 codon 104 are an outcome of environmental adaptation and evolutionary selection that enhance cellular strategies for surviving the environmental stresses of hypoxia and cold (in M. baileyi and M. oeconomus) and hypercapnia (in M. baileyi) in the stressful environments of the Qinghai-Tibet plateau.


Asunto(s)
Adaptación Fisiológica/genética , Apoptosis/genética , Arvicolinae/genética , Frío , Evolución Molecular , Hipoxia/genética , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/genética , Animales , Arvicolinae/metabolismo , Humanos , Hipoxia/metabolismo , Tibet , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Neuro Endocrinol Lett ; 35(6): 429-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25433848

RESUMEN

Prenatal stress (PNS) is associated with increased biological risk for mental disorders such as anxiety and depression later in life, and stress appear to be additive to the PNS influences. Among the most widely cited and accepted alternative hypotheses of anxiety and depression is dysfunction of the HPA axis, a system that is central in orchestrating the stress response. Therefore, understanding how PNS exerts profound effects on the HPA axis and stress-sensitive brain functions including anxiety and depression has significant clinical importance. In this mini-review, we will focus on novel and evolving concepts regarding the potential mechanisms underlying the short and long-term effects of PNS involving CRH peptide family. We present evidence demonstrating prenatal hypoxia exposure induced anxiety-like behavior in adult male rat offspring and CRHR1 in PVN of the hypothalamus is involved.


Asunto(s)
Trastornos de Ansiedad/fisiopatología , Hormona Liberadora de Corticotropina/fisiología , Trastorno Depresivo/fisiopatología , Hipoxia/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Animales , Femenino , Masculino , Embarazo , Ratas , Estrés Fisiológico/fisiología
6.
Neuro Endocrinol Lett ; 35(5): 417-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25275258

RESUMEN

OBJECTIVES: To investigate whether CRHR1 and CRHR2 are colocalized in CRH-specific neurons in rat brain. METHODS: Double/triple immunofluorescence, and combined in situ hybridization were performed in the PVN, amygdala and hippocampus, and triple immunofluorescence was applied to the median eminence (ME), dorsal raphe (DR) and locus coeruleus (LC). RESULTS: Both CRHR1 and CRHR2 immunoreactivity were highly coexpressed in the PVN, central nucleus of the amygdala (CeA) and hippocampus. Triple immunofluorescence under confocal microscopy confirmed that CRHR1 and CRHR2 are coexpressed in CRH-producing neurons in these regions. The results of in situ hybridization combined with double immunofluorescence further strengthened the finding that CRHR1 and CRHR2 were coexpressed in CRH-specific neurons in the PVN, CeA and hippocampus. In addition, CRH immunoreactivity signals were evidently distributed in the ME, DR and LC, and were coexpressed with both receptors. CONCLUSION: CRH receptors colocalize in CRH-containing neurons in the PVN, CeA and hippocampus, and CRH, CRHR1, and CRHR2 coexist in the DR and LC. Our results implicate CRHR1 and CRHR2 in coordinating the regulation of CRH neuronal activity in stress and behavioral responses.


Asunto(s)
Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Núcleo Dorsal del Rafe/metabolismo , Técnica del Anticuerpo Fluorescente , Hipocampo/metabolismo , Hibridación in Situ , Locus Coeruleus/metabolismo , Masculino , Eminencia Media/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/genética
7.
Nat Commun ; 15(1): 3976, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729948

RESUMEN

Bleeding and thrombosis are known as common complications of polycythemia for a long time. However, the role of coagulation system in erythropoiesis is unclear. Here, we discover that an anticoagulant protein tissue factor pathway inhibitor (TFPI) plays an essential role in erythropoiesis via the control of heme biosynthesis in central macrophages. TFPI levels are elevated in erythroblasts of human erythroblastic islands with JAK2V617F mutation and hypoxia condition. Erythroid lineage-specific knockout TFPI results in impaired erythropoiesis through decreasing ferrochelatase expression and heme biosynthesis in central macrophages. Mechanistically, the TFPI interacts with thrombomodulin to promote the downstream ERK1/2-GATA1 signaling pathway to induce heme biosynthesis in central macrophages. Furthermore, TFPI blockade impairs human erythropoiesis in vitro, and normalizes the erythroid compartment in mice with polycythemia. These results show that erythroblast-derived TFPI plays an important role in the regulation of erythropoiesis and reveal an interplay between erythroblasts and central macrophages.


Asunto(s)
Eritroblastos , Eritropoyesis , Factor de Transcripción GATA1 , Hemo , Lipoproteínas , Macrófagos , Policitemia , Policitemia/metabolismo , Policitemia/genética , Policitemia/patología , Eritroblastos/metabolismo , Hemo/metabolismo , Humanos , Animales , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Ratones , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA1/genética , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Trombomodulina/metabolismo , Trombomodulina/genética , Ratones Noqueados , Ferroquelatasa/metabolismo , Ferroquelatasa/genética , Masculino , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Femenino
8.
Horm Behav ; 63(1): 25-31, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23164543

RESUMEN

We previously reported that gestational intermittent hypoxia (GIH) causes anxiety-like behavior in neonatal rats. Here, we showed that the anxiogenic effect was correlated with upregulation of corticotropin-releasing hormone receptor 1 (CRHR1) in the hypothalamic paraventricular nuclei (PVN) by GIH, and was selective to male offspring. The anxiety-like behavior was assessed by both the open field (OF) and elevated plus maze (EPM) tests. We demonstrated that GIH triggered anxiety-like behavior in male offspring, but not in female offspring or in the postpartum dams. Microinjection of antalarmin, a CRHR1-selective antagonist, into the PVN of the male offspring significantly increased the distance traveled and time spent in the central portion of the OF, and the time spent in the open arms in the EPM compared with controls. However, microinjection of the CRHR2 agonist, urocortin III, into the PVN did not affect anxiogenic behavior in the male offspring. These findings clearly demonstrate a gender-selective effect of GIH to increase anxiety-like behavior and this anxiogenic effect might be linked to embryogenically-driven upregulation of PVN CRHR1.


Asunto(s)
Ansiedad/genética , Hipoxia/genética , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Animales , Ansiedad/etiología , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Femenino , Hipoxia/complicaciones , Hipoxia/metabolismo , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Pirimidinas/farmacología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Factores Sexuales
9.
Front Endocrinol (Lausanne) ; 14: 1061235, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817607

RESUMEN

The production and secretion of saliva is an essential function of the salivary glands. Saliva is a complicated liquid with different functions, including moistening, digestion, mineralization, lubrication, and mucosal protection. This review focuses on the mechanism and neural regulation of salivary secretion, and saliva is secreted in response to various stimuli, including odor, taste, vision, and mastication. The chemical and physical properties of saliva change dynamically during physiological and pathophysiological processes. Moreover, the central nervous system modulates salivary secretion and function via various neurotransmitters and neuroreceptors. Smell, vision, and taste have been investigated for the connection between salivation and brain function. The immune and endocrine functions of the salivary glands have been explored recently. Salivary glands play an essential role in innate and adaptive immunity and protection. Various immune cells such as B cells, T cells, macrophages, and dendritic cells, as well as immunoglobins like IgA and IgG have been found in salivary glands. Evidence supports the synthesis of corticosterone, testosterone, and melatonin in salivary glands. Saliva contains many potential biomarkers derived from epithelial cells, gingival crevicular fluid, and serum. High level of matrix metalloproteinases and cytokines are potential markers for oral carcinoma, infectious disease in the oral cavity, and systemic disease. Further research is required to monitor and predict potential salivary biomarkers for health and disease in clinical practice and precision medicine.


Asunto(s)
Glándulas Endocrinas , Glándulas Salivales , Glándulas Salivales/fisiología , Saliva/química , Inmunidad
11.
Front Endocrinol (Lausanne) ; 13: 893238, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36147561

RESUMEN

Hypothalamus-pituitary-adrenal (HPA) axis plays critical roles in stress responses under challenging conditions such as hypoxia, via regulating gene expression and integrating activities of hypothalamus-pituitary-targets cells. However, the transcriptional regulatory mechanisms and signaling pathways of hypoxic stress in the pituitary remain to be defined. Here, we report that hypoxia induced dynamic changes in the transcription factors, hormones, and their receptors in the adult rat pituitary. Hypoxia-inducible factors (HIFs), oxidative phosphorylation, and cAMP signaling pathways were all differentially enriched in genes induced by hypoxic stress. In the pituitary gene network, hypoxia activated c-Fos and HIFs with specific pituitary transcription factors (Prop1), targeting the promoters of hormones and their receptors. HIF and its related signaling pathways can be a promising biomarker during acute or constant hypoxia. Hypoxia stimulated the transcription of marker genes for microglia, chemokines, and cytokine receptors of the inflammatory response. Corticotropin-releasing hormone receptor 1 (CRHR1) mediated the transcription of Pomc, Sstr2, and Hif2a, and regulated the function of HPA axis. Together with HIF, c-Fos initiated and modulated dynamic changes in the transcription of hormones and their receptors. The receptors were also implicated in the regulation of functions of target cells in the pituitary network under hypoxic stress. CRHR1 played an integrative role in the hypothalamus-pituitary-target axes. This study provides new evidence for CRHR1 involved changes of hormones, receptors, signaling molecules and pathways in the pituitary induced by hypoxia.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Receptores de Hormona Liberadora de Corticotropina , Animales , Hormonas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Proopiomelanocortina/genética , ARN Mensajero/genética , Ratas , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Citocinas/metabolismo , Factores de Transcripción/metabolismo
12.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 40(5): 559-66, 2011 09.
Artículo en Zh | MEDLINE | ID: mdl-21984162

RESUMEN

HIF-1 is composed of HIF-1α and HIF-1ß subunits. It promotes target genes transcription under hypoxia and plays essential roles in cell development, physiological adaptations, and pathological processes. In the past 10 years, the research on signaling pathways of HIF-1 in response to cell hypoxia stress, especially on HIF-1α-mediated gene transcription has made great progress.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Transducción de Señal , Animales , Hipoxia de la Célula/fisiología , Humanos
13.
Front Endocrinol (Lausanne) ; 12: 652363, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33796080

RESUMEN

Somatostatin (SST) and somatostatin receptors (SSTRs) play an important role in the brain and gastrointestinal (GI) system. SST is produced in various organs and cells, and the inhibitory function of somatostatin-containing cells is involved in a range of physiological functions and pathological modifications. The GI system is the largest endocrine organ for digestion and absorption, SST-endocrine cells and neurons in the GI system are a critical effecter to maintain homeostasis via SSTRs 1-5 and co-receptors, while SST-SSTRs are involved in chemo-sensory, mucus, and hormone secretion, motility, inflammation response, itch, and pain via the autocrine, paracrine, endocrine, and exoendocrine pathways. It is also a power inhibitor for tumor cell proliferation, severe inflammation, and post-operation complications, and is a first-line anti-cancer drug in clinical practice. This mini review focuses on the current function of producing SST endocrine cells and local neurons SST-SSTRs in the GI system, discusses new development prognostic markers, phosphate-specific antibodies, and molecular imaging emerging in diagnostics and therapy, and summarizes the mechanism of the SST family in basic research and clinical practice. Understanding of endocrines and neuroendocrines in SST-SSTRs in GI will provide an insight into advanced medicine in basic and clinical research.


Asunto(s)
Tracto Gastrointestinal/fisiología , Receptores de Somatostatina/fisiología , Somatostatina/fisiología , Animales , Antineoplásicos/farmacología , Comunicación Celular/efectos de los fármacos , Proliferación Celular , Modelos Animales de Enfermedad , Sistema Nervioso Entérico/fisiología , Homeostasis , Humanos , Inflamación , Ligandos , Neuronas/metabolismo , Sistema Nervioso Parasimpático/fisiología , Pronóstico , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Células Secretoras de Somatostatina/metabolismo , Sistema Nervioso Simpático/fisiología
14.
Gen Comp Endocrinol ; 161(3): 400-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19233187

RESUMEN

Here, we report the cloning and characterization of growth hormone (GH), insulin-like growth factor-I (IGF-I) and IGF-II from naked carp (Gymnocypris przewalskii), a native teleost fish of Lake Qinghai in the Qinghai-Tibet Plateau of China. The GH of naked carp encodes for a predicted amino acid sequence showing identities of 63%, 63%, 91% and 94% with cherry salmon, rainbow trout, zebrafish and grass carp, respectively. Compared to common carp and goldfish, evolutionary analysis showed that genome duplication has had less influence on the relaxation of purifying selection in the evolution of naked carp GH. Sequence analysis of naked carp IGF-I (ncIGF-I) and ncIGF-II showed a high degree of homology with known fish IGF-I and IGF-II. To investigate effects of salinity and ionic composition of the aquatic environment on the GH-IGF axis in naked carp, male fish held in river water were assigned randomly to 4 groups: RW (river-water), RW+Na (NaCl in RW), RW+Mg (MgCl(2) in RW) and LW (lake-water) groups. The concentrations of Na(+) in RW+Na and Mg(2+) in RW+Mg were equal to the concentrations of these ions in lake-water. After 2 days of exposure, the plasma IGF-I levels in the RW+Na and LW groups were significantly higher than the control group (RW), and the plasma GH levels of the LW group were also significantly higher than the RW group. The somatostatin (SS) levels in the hypothalamus significantly increased in the RW+Na group. After 5 days of exposure, these hormone levels did not differ significantly among groups. These results indicate that while the plasma GH and IGF-I levels are osmosensitive, the absence of a change in GH secretion in RW+Na might be partly due to a transiently increased release of hypothalamic SS induced by the stress of neutral-saline water. This is the first report of a salinity-induced increase of GH-IGF-I circulating levels in Cypriniformes.


Asunto(s)
Carpas/metabolismo , Regulación de la Expresión Génica , Hormona del Crecimiento/metabolismo , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , China , Evolución Molecular , Hormona del Crecimiento/sangre , Hormona del Crecimiento/química , Hormona del Crecimiento/clasificación , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Factor I del Crecimiento Similar a la Insulina/química , Factor I del Crecimiento Similar a la Insulina/clasificación , Factor II del Crecimiento Similar a la Insulina/química , Factor II del Crecimiento Similar a la Insulina/clasificación , Masculino , Salinidad , Sales (Química)/farmacología , Alineación de Secuencia , Somatostatina/metabolismo
15.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 38(1): 95-9, 2009 01.
Artículo en Zh | MEDLINE | ID: mdl-19253435

RESUMEN

OBJECTIVE: To evaluate the correlation of venous return disturbance (VRD), bone marrow edema (BME) and the amount of joint fluid with clinical symptoms in osteonecrosis of the femoral head (ONFH). METHODS: Twenty patients (25 hips) with ONFH proved by pathology were parallelly examined by 1.5 T MR and medullograph. RESULT: In total 25 hips VRD was observed in 16 hips with medullograph and 13 hips with the clinical symptoms (P <0.05). BME was found in 17 with MRI (P <0.01), and the was found in 15 hips with MRI (Spearman=0.949). VRD were found in 16 hydrarthrus. CONCLUSION: ONFH patients with clinical symptoms have VRD, BME and the hydrarthrus. The medullograph is of value in monitoring the development of ONFH.


Asunto(s)
Necrosis de la Cabeza Femoral/patología , Cabeza Femoral/irrigación sanguínea , Imagen por Resonancia Magnética , Flujo Sanguíneo Regional/fisiología , Adulto , Médula Ósea/patología , Médula Ósea/fisiopatología , Edema/patología , Femenino , Necrosis de la Cabeza Femoral/fisiopatología , Articulación de la Cadera/patología , Humanos , Masculino , Persona de Mediana Edad , Venas/fisiopatología
16.
Artículo en Inglés | MEDLINE | ID: mdl-30915036

RESUMEN

The common understanding of p53 function is a genome guardian, which is activated by diverse stresses stimuli and mediates DNA repair, apoptosis, and cell cycle arrest. Increasing evidence has demonstrated p53 new cellular functions involved in abundant endocrine and metabolic response for maintaining homeostasis. However, TP53 is frequently mutant in human cancers, and the mutant p53 (Mut-p53) turns to an "evil" cancer-assistant. Mut-p53-induced epithelial-mesenchymal transition (EMT) plays a crucial role in the invasion and metastasis of endocrine carcinomas, and Mut-p53 is involved in cancer immune evasion by upregulating PD-L1 expression. Therefore, Mut-p53 is a valuable treatment target for malignant tumors. Targeting Mut-p53 in correcting sequence and conformation are increasingly concerned. Interestingly, in wild animals, p53 variations contribute to cancer resistant and high longevity. This review has discussed the multiple functions of p53 in health, diseases, and nature evolution, summarized the frequently mutant sites of p53, and the mechanisms of Mut-p53-mediated metastasis and immune evasion in endocrine cancers. We have provided a new insight for multiple roles of p53 in human and wild animals.

17.
J Zhejiang Univ Sci B ; 9(9): 739-46, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18837481

RESUMEN

OBJECTIVE: To study the relationships among magnetic resonance imaging (MRI), histological findings, and insulin-like growth factor-I (IGF-I) in steroid-induced osteonecrosis of the femoral head in rabbits. METHODS: Thirty rabbits were randomly divided into experimental Group A (n=15) and control Group B (n=15). The 7.5 mg/kg (2 ml) of dexamethasone (DEX) and physiological saline (2 ml) were injected into the right gluteus medius muscle twice at one-week intervals in animals of Groups A and B, respectively. At 4, 8 and 16 weeks after obtaining an MRI, the rabbits were sacrificed and the femoral head from one side was removed for histological study of lacunae empty of osteocytes, subchondral vessels, and size of fat cells under microscopy, and the femoral head from the other side was removed for enzyme-linked immunoadsorbent assay (ELISA) for IGF-I. RESULTS: At 4, 8 and 16 weeks after treatment, no necrotic lesions were detected in Group B, while they were detected in Group A. Light microscopy revealed that the fat cells of the marrow cavity were enlarged, subchondral vessels were evidently decreased, and empty bone lacunae were clearly increased. The IGF-I levels in Group A were significantly higher than those in Group B. At 8 weeks after the DEX injection, the MRI of all 20 femora showed an inhomogeneous, low signal intensity area in the femoral head, and at 16 weeks, the findings of all 10 femora showed a specific "line-like sign". The MRI findings of all femora in Group B were normal. CONCLUSION: MRI is a highly sensitive means of diagnosing early experimental osteonecrosis of the femoral head. However, the abnormal marrow tissues appeared later than 4 weeks when the expression of IGF-I increased. This reparative factor has an early and important role in response to steroid-induced osteonecrosis of the femoral head, and provides a theoretical foundation for understanding the pathology and designing new therapies.


Asunto(s)
Dexametasona , Modelos Animales de Enfermedad , Necrosis de la Cabeza Femoral/metabolismo , Necrosis de la Cabeza Femoral/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Esteroides , Animales , Necrosis de la Cabeza Femoral/inducido químicamente , Imagen por Resonancia Magnética , Conejos
20.
Endocrinology ; 148(7): 3271-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17379644

RESUMEN

To determine the influence of continuous hypoxia on body weight, food intake, hepatic glycogen, circulatory glucose, insulin, glucagon, leptin, and corticosterone, and the involvement of the corticotropin-releasing factor receptor type 1 (CRFR1) in modulation of these hormones, rats were exposed to a simulated altitude of 5 km (approximately 10.8% O2) in a hypobaric chamber for 1, 2, 5, 10, and 15 d. Potential involvement of CRFR1 was assessed through five daily sc injections of a CRFR1 antagonist (CP-154,526) prior to hypoxia. Results showed that the levels of body weight, food intake, blood glucose, and plasma insulin were significantly reduced; the content of hepatic glycogen initially and transiently declined, whereas the early plasma glucagon and leptin remarkably increased; plasma corticosterone was markedly increased throughout the hypoxic exposure of 1-15 d. Compared with hypoxia alone, CRFR1 antagonist pretreatment in the hypoxic groups prevented the rise in corticosterone, whereas the levels of body weight and food intake were unchanged. At the same time, the reduction in blood glucose was greater and the pancreatic glucose was increased, plasma insulin reverted toward control, and plasma glucagon decreased. In summary, prolonged hypoxia reduced body weight, food intake, blood glucose, and plasma insulin but transiently enhanced plasma glucagon and leptin. In conclusion, CRFR1 is potentially involved in the plasma insulin reduction and transient glucagon increase in hypoxic rats.


Asunto(s)
Glucemia/metabolismo , Glucagón/sangre , Hipoxia/fisiopatología , Insulina/sangre , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Altitud , Animales , Peso Corporal/efectos de los fármacos , Corticosterona/sangre , Ingestión de Alimentos/efectos de los fármacos , Leptina/sangre , Glucógeno Hepático/metabolismo , Masculino , Pirimidinas/farmacología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA