Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Rev. lab. clín ; 6(3): 132-134, jul.-sept. 2013.
Article in Spanish | IBECS | ID: ibc-115470

ABSTRACT

El hipotiroidismo materno clínico y el subclínico tienen consecuencias graves tanto para la madre como para el feto. Debido a la compleja fisiología de la función tiroidea durante el embarazo, debería realizarse la evaluación hormonal según valores de referencia para cada trimestre de gestación y para cada zona y con las técnicas propias de cada laboratorio. Si no se dispone en el laboratorio de estos valores de referencia trimestrales propios, se recomiendan los siguientes valores de referencia de tirotropina: primer trimestre < 2,5 mUI/L; segundo y tercer trimestres < 3,0 mUI/L(AU)


Overt and subclinical maternal hypothyroidism is known to have serious adverse effects for both mother and fetus. Given the complex physiology of thyroid function during pregnancy, hormone assessment should be performed according to reference values for each gestational trimester and generated locally in each reference laboratory. If trimester-specific references intervals are not available in the laboratory, the following reference ranges of tirotropine are recommended: first trimester < 2,5 mUI/L; second and third trimesters < 3,0 mUI/L(AU)


Subject(s)
Humans , Female , Receptors, Thyrotropin/isolation & purification , Thyrotropin , Thyrotropin-Releasing Hormone , Receptors, Thyrotropin-Releasing Hormone/analysis , Pregnancy/blood , Reference Values , Congenital Hypothyroidism/blood , Hypothyroidism/blood , Congenital Hypothyroidism/complications , Congenital Hypothyroidism/diagnosis , Hypothyroidism/complications , Hypothyroidism/diagnosis
2.
Menopause ; 19(11): 1253-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22713859

ABSTRACT

OBJECTIVE: Thyroid gland dysfunction is associated with menstrual cycle disturbances, infertility, and increased risk of miscarriage, but the mechanisms are poorly understood. However, little is known about the regulation of these receptors in the uterus. The aim of this study was to determine the effects of long-term treatment with steroid hormones on the expression, distribution, and regulation of the receptors for thyrotropin-releasing hormone (TRHR) and thyroid-stimulating hormone (TSHR), thyroid hormone receptor α1/α2 (THRα1/α2), and THRß1 in the uterus of surgically menopausal monkeys. METHODS: Eighty-eight cynomolgus macaques were ovariectomized and treated orally with conjugated equine estrogens (CEE; n = 20), a combination of CEE and medroxyprogesterone acetate (MPA; n = 20), or tibolone (n = 28) for 2 years. The control group (OvxC; n = 20) received no treatment. Immunohistochemistry was used to evaluate the protein expression and distribution of the receptors in luminal epithelium, glands, stroma, and myometrium of the uterus. RESULTS: Immunostaining of TRHR, TSHR, and THRs was detected in all uterine compartments. Epithelial immunostaining of TRHR was down-regulated in the CEE + MPA group, whereas in stroma, both TRHR and TSHR were increased by CEE + MPA treatment as compared with OvxC. TRHR immunoreactivity was up-regulated, but THRα and THRß were down-regulated, in the myometrium of the CEE and CEE + MPA groups. The thyroid-stimulating hormone level was higher in the CEE and tibolone groups as compared with OvxC, but the level of free thyroxin did not differ between groups. CONCLUSIONS: All receptors involved in thyroid hormone function are expressed in monkey uterus, and they are all regulated by long-term steroid hormone treatment. These findings suggest that there is a possibility of direct actions of thyroid hormones, thyroid-stimulating hormone and thyrotropin-releasing hormone on uterine function.


Subject(s)
Gonadal Steroid Hormones/pharmacology , Macaca fascicularis , Receptors, Thyroid Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin/drug effects , Uterus/chemistry , Animals , Estrogens, Conjugated (USP)/administration & dosage , Female , Immunohistochemistry/veterinary , Medroxyprogesterone Acetate/administration & dosage , Norpregnenes/administration & dosage , Receptors, Thyroid Hormone/analysis , Receptors, Thyrotropin/analysis , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyroid Hormone Receptors alpha/analysis , Thyroid Hormone Receptors alpha/drug effects , Thyroid Hormone Receptors beta/analysis , Thyroid Hormone Receptors beta/drug effects , Uterus/physiology
4.
Hypertension ; 57(1): 103-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21135357

ABSTRACT

Local thyrotropin-releasing hormone (TRH) may be involved in cardiac pathophysiology, but its role in left ventricular hypertrophy (LVH) is still unknown. We studied whether local TRH is involved in LVH of spontaneously hypertensive rats (SHR) by investigating TRH expression and its long-term inhibition by interference RNA (TRH-iRNA) during LVH development at 2 stages (prehypertrophy and hypertrophy). SHR and their control rats (WKY) were compared. Cardiac hypertrophy was expressed as heart/total body weight (HW/BW) ratio. TRH content (radioimmuno assay), preproTRH, TRH receptor type I, brain natriuretic peptide (BNP), and collagen mRNA expressions (real-time polymerase chain reaction) were measured. For long-term inhibition of TRH, TRH-iRNA was injected into the left ventricle (LV) wall for 8 weeks. Hearts were processed for morphometric studies and immunohistochemical analysis using antibodies against α-smooth muscle actin and collagen type III. LV preproTRH-mRNA abundance was similar in both strains at 7 weeks of age. At the hypertrophic stage (18 weeks old), however, there was a 15-fold increase in SHR versus WKY, consistent with a significant increase in tripeptide levels and the expression of its receptor. Specific LV-TRH inhibition at the prehypertensive stage with TRH-iRNA, which decreased >50% preproTRH expression and tripeptide levels, prevented LVH development as shown by the normal HW/BW ratio observed in TRH-iRNA-treated SHR. In addition, TRH-iRNA impeded the increase in BNP and type III collagen expressions and prevented the increase in cardiomyocyte diameter evident in mismatch iRNA-treated adult SHR. These results show for the first time that the cardiac TRH system is involved in the development of LVH in SHR.


Subject(s)
Hypertrophy, Left Ventricular/metabolism , Thyrotropin-Releasing Hormone/metabolism , Actins/analysis , Animals , Collagen Type III/analysis , Hypertrophy, Left Ventricular/pathology , Male , Natriuretic Peptide, Brain/analysis , RNA Interference , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Thyrotropin-Releasing Hormone/analysis
5.
Pancreas ; 38(2): 197-202, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18948837

ABSTRACT

OBJECTIVES: Thyrotropin-releasing hormone (TRH) is expressed in rodent and human adult pancreata and in mouse pancreas during embryonic development. However, expression of TRH receptors (TRHRs) in the pancreas is controversial. We sought to provide evidence that the TRH/TRHR system might play a role in fetal development. METHODS: We used quantitative reverse transcription-polymerase chain reaction to measure TRH and TRHR messenger RNA (mRNA). To study the effects of TRHR expression in a pancreatic progenitor population, we expressed TRHRs in human islet-derived precursor cells (hIPCs) by infection with adenoviral vector AdCMVmTRHR. Thyrotropin-releasing hormone receptor signaling was measured as inositol phosphate production and intracellular calcium transients. Thyrotropin-releasing hormone receptor expression was measured by [H]methyl-TRH binding. Apoptosis was monitored by release of cytochrome c from mitochondria. RESULTS: We show that TRH mRNA is expressed in human fetal and adult pancreata, and that TRHR mRNA is expressed in fetal human pancreas but not in adult human pancreas. Thyrotropin-releasing hormone receptors expressed in hIPCs were shown to signal normally. Most importantly, TRH treatment for several days stimulated apoptosis in hIPCs expressing approximately 400,000 TRHRs per cell. CONCLUSIONS: These findings suggest a possible role for TRH/TRHR signaling in pancreatic precursors to promote programmed cell death, a normal constituent of morphogenesis during embryonic development in humans.


Subject(s)
Apoptosis , Pancreas/cytology , Pancreas/embryology , Receptors, Thyrotropin-Releasing Hormone/physiology , Humans , RNA, Messenger/analysis , Receptors, Thyrotropin-Releasing Hormone/analysis , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/genetics
6.
J Endocrinol ; 189(2): 271-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16648294

ABSTRACT

The hatching process in the chicken is accompanied by dramatic changes in plasma thyroid hormones. The cause of these changes, though crucial for hatching and the onset of endothermy, is not known. One hypothesis is that the pituitary gland becomes more sensitive to hypothalamic stimulation during this period. We have tested whether the responsiveness of the thyrotropes to hypothalamic stimuli changes throughout the last week of embryonic development and hatching by studying the mRNA expression of receptors involved in the control of the secretory activity of this cell type. We used a real-time PCR set-up to quantify whole pituitary mRNA expression of the beta subunit of thyrotrophin (TSH-beta), type 1 thyrotrophin-releasing hormone receptor (TRH-R1), corticotrophin-releasing hormone receptors (CRH-R1 and CRH-R2) and somatostatin subtype receptor 2 (SSTR2) on every day of the last week of embryonic development, including the day of hatch and the first day of posthatch life. The thyrotrope-specific expression was investigated by a combination of in situ hybridization with immunohistochemistry at selected ages. Although TSH-beta mRNA levels increased towards day 19 of incubation (E19), the expression of CRH-R2 and TRH-R1 mRNA by the thyrotropes tended to decrease during this period, suggesting a lower sensitivity of the thyrotropes to the stimulatory factors CRH and TRH. CRH-R1, which is not involved in the control of TSH secretion, increased steadily throughout the period tested. The expression of SSTR2 mRNA by the thyrotropes was low during embryonic development and increased just before hatching. We have concluded that the sensitivity of the pituitary thyrotropes to hypothalamic stimulation decreases throughout the last week of embryonic development, so that the higher expression of TSH-beta mRNA around E16-E19, and hence the increasing plasma thyroxine level, is unlikely to be the result of an increased stimulation by either TRH or CRH.


Subject(s)
Embryonic Development/physiology , Hypothalamus/embryology , Pituitary Gland, Anterior/embryology , Thyroxine/blood , Animals , Chick Embryo , Corticosterone/blood , Immunohistochemistry/methods , In Situ Hybridization/methods , Pituitary Gland, Anterior/cytology , RNA, Messenger/analysis , Receptors, Corticotropin-Releasing Hormone/analysis , Receptors, Somatostatin/analysis , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyrotropin, beta Subunit/analysis , Triiodothyronine/blood
7.
J Physiol ; 571(Pt 1): 27-42, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16339175

ABSTRACT

The erg1a (HERG) K+ channel subunit and its N-terminal splice variant erg1b are coexpressed in several tissues and both isoforms have been shown to form heteromultimeric erg channels in heart and brain. The reduction of erg1a current by thyrotropin-releasing hormone (TRH) is well studied, but no comparable data exist for erg1b. Since TRH and TRH receptors are widely expressed in the brain, we have now studied the different TRH effects on the biophysical properties of homomeric rat erg1b as well as heteromeric rat erg1a/1b channels. The erg channels were overexpressed in the clonal somatomammotroph pituitary cell line GH3/B6, which contains TRH receptors and endogenous erg channels. Compared to rerg1a, homomeric rerg1b channels exhibited not only faster deactivation kinetics, but also considerably less steady-state inactivation, and half-maximal activation occurred at about 10 mV more positive potentials. Coexpression of both isoforms resulted in erg currents with intermediate properties concerning the deactivation kinetics, whereas rerg1a dominated the voltage dependence of activation and rerg1b strongly influenced steady-state inactivation. Application of TRH induced a reduction of maximal erg conductance for all tested erg1 currents without effects on the voltage dependence of steady-state inactivation. Nevertheless, homomeric rerg1b channels significantly differed in their response to TRH from rerg1a channels. The TRH-induced shift in the activation curve to more positive potentials, the dramatic slowing of activation and the acceleration of deactivation typical for rerg1a modulation were absent in rerg1b channels. Surprisingly, most effects of TRH on heteromeric rerg1 channels were dominated by the rerg1b subunit.


Subject(s)
Ether-A-Go-Go Potassium Channels/physiology , Protein Subunits/physiology , Thyrotropin-Releasing Hormone/pharmacology , Action Potentials , Animals , Cell Line , ERG1 Potassium Channel , Electrophysiology , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation/drug effects , Membrane Potentials/drug effects , Pituitary Gland/chemistry , Pituitary Gland/cytology , Protein Isoforms , Protein Subunits/analysis , Rats , Receptors, Thyrotropin-Releasing Hormone/analysis , Receptors, Thyrotropin-Releasing Hormone/physiology , Reverse Transcriptase Polymerase Chain Reaction
8.
J Biochem ; 138(2): 111-25, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16091585

ABSTRACT

Some G protein-coupled receptors might be spacially targetted to discrete domains within the plasma membrane. Here we assessed the localization in membrane domains of the epitope-tagged, fluorescent version of thyrotropin-releasing hormone receptor (VSV-TRH-R-GFP) expressed in HEK293 cells. Our comparison of three different methods of cell fractionation (detergent extraction, alkaline treatment/sonication and mechanical homogenization) indicated that the dominant portion of plasma membrane pool of the receptor was totally solubilized by Triton X-100 and its distribution was similar to that of transmembrane plasma membrane proteins (glycosylated and non-glycosylated forms of CD147, MHCI, CD29, CD44, transmembrane form of CD58, Tapa1 and Na,K-ATPase). As expected, caveolin and GPI-bound proteins CD55, CD59 and GPI-bound form of CD58 were preferentially localized in detergent-resistant membrane domains (DRMs). Trimeric G proteins G(q)alpha/G(11)alpha, G(i)alpha1/G(i)alpha2, G(s)alphaL/G(s)alphaS and Gbeta were distributed almost equally between detergent-resistant and detergent-solubilized pools. In contrast, VSV-TRH-R-GFP, Galpha, Gbeta and caveolin were localized massively only in low-density membrane fragments of plasma membranes, which were generated by alkaline treatment/sonication or by mechanical homogenization of cells. These data indicate that VSV-TRH-R-GFP as well as other transmembrane markers of plasma membranes are excluded from TX-100-resistant, caveolin-enriched membrane domains. Trimeric G protein G(q)alpha/G(11)alpha occurs in both DRMs and in the bulk of plasma membranes, which is totally solubilized by TX-100.


Subject(s)
Cell Membrane/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Membrane Microdomains/chemistry , Receptors, Thyrotropin-Releasing Hormone/chemistry , Caveolin 1 , Caveolins/chemistry , Cell Culture Techniques , Centrifugation, Density Gradient , Detergents , GTP-Binding Protein alpha Subunits, Gq-G11/analysis , Humans , Immunoblotting , Octoxynol , Radioligand Assay , Receptors, Thyrotropin-Releasing Hormone/analysis , Solubility , Spectrometry, Fluorescence
9.
Mol Endocrinol ; 19(11): 2859-70, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16020481

ABSTRACT

To investigate the function of dimerization of the TRH receptor, a controlled dimerization system was developed. A variant FK506 binding protein (FKBP) domain was fused to the receptor C terminus and dimerization induced by incubating cells with dimeric FKBP ligand, AP20187. The TRH receptor-fusion bound hormone and signaled normally. Addition of dimerizer to cells expressing the receptor-FKBP fusion dramatically increased the fraction of receptor running as dimer on SDS-PAGE. AP20187 caused dimerization in a time- and concentration-dependent manner, acting within 1 min. Dimerizer had no effect on TRH receptors lacking the FKBP domain, and its effects were blocked by excess monomeric FKBP ligand. AP20187-induced dimerization did not cause receptor phosphorylation, inositol phosphate production, or ERK1/2 activation, and dimerizer did not alter signaling by TRH. Induced dimerization did, however, alter TRH receptor trafficking. TRH promoted greater receptor internalization in cells treated with AP20187 but not monomeric ligand, based on loss of surface binding sites and immunostaining. Dimerization increased the rate of internalization of TRH receptors and decreased the apparent rate of receptor recycling. AP20187 enhanced the small amount of TRH-induced receptor internalization when the receptor-FKBP fusion protein was expressed in cells lacking beta-arrestins. The results show that controlled dimerization of the TRH receptor potentiates hormone-induced receptor trafficking.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Arrestins/metabolism , CHO Cells , Calcium/metabolism , Cell Membrane/chemistry , Cricetinae , Dimerization , Enzyme Activation/drug effects , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Transport/drug effects , Receptors, Thyrotropin-Releasing Hormone/analysis , Receptors, Thyrotropin-Releasing Hormone/drug effects , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/analysis , Tacrolimus Binding Proteins/drug effects , Tacrolimus Binding Proteins/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Type C Phospholipases/metabolism , beta-Arrestins
10.
Histol Histopathol ; 20(3): 713-8, 2005 07.
Article in English | MEDLINE | ID: mdl-15944919

ABSTRACT

Thyrotropin-releasing hormone (TRH) synthesized in the hypothalamus has the capability of inducing the release of thyroid-stimulating hormone (TSH) from the anterior pituitary, which in turn stimulates the production of thyroid hormones in the thyroid gland. Immunoreactivity for TRH and TRH-like peptides has been found in some tissues outside the nervous system, including thyroid. It has been demonstrated that thyroid C-cells express authentic TRH, affecting thyroid hormone secretion by follicular cells. Therefore, C-cells could have a paracrine role in thyroid homeostasis. If this hypothesis is true, follicular cells should express TRH receptors (TRH-Rs) for the paracrine modulation carried out by C-cells. In order to elucidate whether or not C-cell TRH production could act over follicular cells modulating thyroid function, we studied TRH-Rs expression in PC C13 follicular cells from rat thyroid, by means of immunofluorescence technique and RT-PCR analysis. We also investigated the possibility that C-cells present TRH-Rs for the autocrine control of its own TRH production. Our results showed consistent expression for both receptors, TRH-R1 and TRH-R2, in 6-23 C-cells, and only for TRH-R2 in PC C13 follicular cells. Our data provide new evidence for a novel intrathyroidal regulatory pathway of thyroid hormone secretion via paracrine/autocrine TRH signaling.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/genetics , Thyroid Gland/metabolism , Animals , Cell Line , Fluorescent Antibody Technique/methods , Gene Expression , Paracrine Communication/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Thyrotropin-Releasing Hormone/analysis , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/cytology
11.
J Endocrinol ; 181(3): 401-12, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15171688

ABSTRACT

Thyrotropin-releasing hormone (TRH), a hypothalamic tripeptide, is expressed in pancreatic islets at peak levels during the late gestation and early neonate period. TRH increases insulin production in cultured beta-cells, suggesting that it might play a role in regulating pancreatic beta-cell function. However, there is limited information on TRH receptor expression in the pancreas. The aim of the present study was to explore the distribution of the TRH receptor in the pancreas and its function in pancreatic beta-cells. TRH receptor type 1 (TRHR1) gene expression was detected by RT-PCR and verified by Northern blotting and immunoblotting in the beta-cell lines, INS-1 and betaTC-6, and the rat pancreatic organ. The absence of TRH receptor type 2 expression in the tissue and cells indicated the tissue specificity of TRH receptor expression in the pancreas. The TRHR1 signals (detected by in situ hybridization) were distributed not only in islets but also in the surrounding areas of the pancreatic ductal and vasal epithelia. The apparent dissociation constant value for the affinity of [(3)H]3-methyl-histidine TRH (MeTRH) is 4.19 in INS-1 and 3.09 nM in betaTC-6. In addition, TRH induced epidermal growth factor (EGF) receptor phosphorylation with a half-maximum concentration of approximately 50 nM, whereas the high affinity analogue of TRH, MeTRH, was 1 nM. This suggested that the affinity of TRH ligands for the TRH receptor influences the activation of EGF receptor phosphorylation in betaTC-6 cells. Our observations suggested that the biological role of TRH in pancreatic beta-cells is via the activation of TRHR1. Further research is required to identify the role of TRHR1 in the pancreas aside from the islets.


Subject(s)
Islets of Langerhans/chemistry , RNA, Messenger/analysis , Receptors, Thyrotropin-Releasing Hormone/genetics , Animals , Base Sequence , Blotting, Western/methods , Cell Line , Insulin/analysis , Molecular Sequence Data , Pancreas/chemistry , Precipitin Tests , Rats , Receptors, Thyrotropin-Releasing Hormone/analysis , Reverse Transcriptase Polymerase Chain Reaction
12.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 15(10): 622-4, 2003 Oct.
Article in Chinese | MEDLINE | ID: mdl-14552689

ABSTRACT

OBJECTIVE: To study the dose and location of thyrotropin releasing hormone (TRH) and its receptor in Wistar rat's brain when early endotoxemia happened. METHODS: Twenty-two Wistar rats were selected. In 13 mouse the model was established with endotoxemia and hemorrhagic shock. The TRH and its receptor were observed with radioimmunoassay (RIA), and were compared to the 9 control rats. RESULTS: The dose of TRH decreased significantly in the early stage of endotoxemia. The maxium dose (Bmax) of TRH increased while Kd (affinity) decreased, so the effective TRH receptor had no significantly change. CONCLUSION: The dose of TRH is different in brain and serum. The decrease of TRH and the steady of active TRH receptor may protect the brain tissue and repress multiple organ dysfunction syndrome (MODS) in the early stage of endotoxemia.


Subject(s)
Brain Chemistry , Endotoxemia/metabolism , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyrotropin-Releasing Hormone/analysis , Animals , Male , Radioimmunoassay , Rats , Rats, Wistar
13.
Mol Cell Endocrinol ; 203(1-2): 33-9, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12782401

ABSTRACT

Thyrotropin or thyroid-stimulating hormone (TSH) secretion in the chicken is controlled by several hypothalamic hormones. It is stimulated by thyrotropin-releasing hormone (TRH) and corticotropin-releasing hormone (CRH), whereas somatostatin (SRIH) exerts an inhibitory effect. In order to determine the mechanism by which these hypothalamic hormones modulate chicken TSH release, we examined the cellular localization of TRH receptors (TRH-R), CRH receptors type 1 (CRH-R1) and somatostatin subtype 2 receptors (SSTR2) in the chicken pars distalis by in situ hybridization (ISH), combined with immunological staining of thyrotropes. We show that thyrotropes express TRH-Rs and SSTR2s, allowing a direct action of TRH and SRIH at the level of the thyrotropes. CRH-R1 expression is virtually confined to corticotropes, suggesting that CRH-induced adrenocorticotropin release is the result of a direct stimulation of corticotropes, whereas CRH-stimulated TSH release is not directly mediated by the known chicken CRH-R1. Possibly CRH-induced TSH secretion is mediated by a yet unknown type of CRH-R in the chicken. Alternatively, a pro-opiomelanocortin (POMC)-derived peptide, secreted by the corticotropes following CRH stimulation, could act as an activator of TSH secretion in a paracrine way.


Subject(s)
Pituitary Gland, Anterior/cytology , Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, Somatostatin/physiology , Receptors, Thyrotropin-Releasing Hormone/physiology , Thyrotropin/metabolism , Animals , Chickens , Female , Immunohistochemistry , In Situ Hybridization , Male , Pituitary Gland, Anterior/chemistry , RNA, Messenger/analysis , Receptors, Corticotropin-Releasing Hormone/analysis , Receptors, Somatostatin/analysis , Receptors, Thyrotropin-Releasing Hormone/analysis , Tissue Distribution
14.
Chin J Physiol ; 45(1): 19-24, 2002 Mar 31.
Article in English | MEDLINE | ID: mdl-12005348

ABSTRACT

The present study was to investigate the localization of preganglionic parasympathetic neurons of gallbladder in brain stem by anatomical and functional approaches. Male or female rabbits (n = 11) were anesthetized with sodium pentobarbital (30 mg/kg, i.v.). Cholera toxin B conjugated to horseradish peroxidase (CB-HRP) was injected into the gallbladder wall. Four days later, animals were re-anesthetized and perfused transcardially with paraformaldehyde solution in a 0.1 M phosphate buffer. The rabbit brain was then frozenly sectioned. The sections were processed for HRP label and stained with neutral red. Another group of rabbits (n = 54) were anesthetized by urethane (1 g/kg) after fasting for 18-24 hours, Gallbladder pressure (GP) was measured by inserting a frog bladder filled with normal saline into the gallbladder. Myoelectrical activity of the sphincter of Oddi (SO) was induced by a pair of copper electrodes. A glass tube (30 microm tip diameter) connected with a microsyringe was directed to the dorsal vagal complex (DVC) for microinjection. Majority of retrogradely labeled cells was found bilaterally in dorsal motor nucleus of the vagus nerve (DMV) throughout the length, except the rostral and caudal part. These cells were distributed in subnuclei parvicellularis or mediocellularis of DMV. Some labeled perikarya located in the medial subnucleus of the solitary tract (mNTS). Thyrotropin-releasing hormone (TRH, 1.3 mmol/L, 0.2 microl) microinjected into the rostral portion of the DVC (including DMV and NTS) enhanced the motility of gallbladder and SO. Microinjection of TRH at the middle part of DVC seldom induces excitatory effects on the gallbladder or SO. TRH microinjected into the caudal portion of the DVC elicited weaker response of gallbladder and SO than rostral portion. Our results indicated that DMV is one of the most important original nuclei of gallbladder's vagus nerves and mNTS may be also involved in the control of gallbladder's parasympathetic activity. Neurons that innervate the gallbladder distribute at most part of DVC, and are relatively dense at rostral and caudal position of DMV.


Subject(s)
Brain Stem/cytology , Gallbladder/innervation , Parasympathetic Nervous System/cytology , Animals , Brain Stem/physiology , Horseradish Peroxidase , Parasympathetic Nervous System/physiology , Rabbits , Receptors, Thyrotropin-Releasing Hormone/analysis , Receptors, Thyrotropin-Releasing Hormone/physiology
15.
Glia ; 33(4): 267-76, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11246225

ABSTRACT

The presence of thyrotropin-releasing hormone (Thyroliberin, TRH) and its receptor (TRH-R) in frozen coronal sections of the adult rat spinal cord and neonatal rat astroglial cultures was investigated by means of immunocytochemistry and Western blot using polyclonal antibodies generated against the hormone and monoclonal antibodies originated against discrete sequences of the type 1 rat TRH receptor (TRH-R1). TRH-R1 and TRH are present both in astroglial cells from adult rats and in cultured cells from newborn animals. The localization of TRH and TRH-R1 in nonneuronal cells in the central nervous system may reflect that some of the neurotrophic actions of TRH upon the central nervous system are mediated by glial cells.


Subject(s)
Astrocytes/chemistry , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyrotropin-Releasing Hormone/analysis , Animals , Antibodies, Monoclonal , Astrocytes/cytology , Blotting, Western , Cells, Cultured , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Immunohistochemistry , Male , Rats , Rats, Wistar , Receptors, Thyrotropin-Releasing Hormone/immunology , Spinal Cord/chemistry , Spinal Cord/cytology , Thyrotropin-Releasing Hormone/immunology
16.
J Clin Endocrinol Metab ; 85(8): 2709-13, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10946869

ABSTRACT

In many acromegalic patients the paradoxical release of GH in response to TRH has been well documented, but the mechanisms underlying this phenomenon are not understood. It has been suggested that aberrant GH secretion may result from TRH endogenously synthesized by the adenoma. In 32 adenomas from acromegalic patients, TRH-like immunoreactivity (TRH-LI) was measured using 2 well characterized antisera. TRH-LI was not detectable in 10 samples, and in 19 samples, TRH-LI was measured only by the less specific antibody. With the TRH-specific antibody, TRH-LI was identified only in 3 samples, 2 of which contained exceedingly high concentrations (40 and 96 pg/mg tissue). In the latter 2 samples, prepro-TRH messenger ribonucleic acid was identified by Northern blot analysis, but not in the control tissue sample of a patient without pituitary disease and also not in the other adenomas analyzed by this technique. Transcripts of the TRH receptor were almost undetectable in all adenomas analyzed. For the TRH-degrading ectoenzyme, a potential regulator of TRH signals at adenohypophyseal target sites, transcripts were significantly expressed only in the TRH-producing adenomas. We conclude that the TRH-signaling elements examined are, in general, not directly involved in the mechanisms causing paradoxical GH secretion in acromegalic patients.


Subject(s)
Acromegaly/physiopathology , Adenoma/chemistry , Pituitary Neoplasms/chemistry , Protein Precursors/genetics , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/physiology , Acromegaly/etiology , Adenoma/genetics , Adenoma/pathology , Adenoma/surgery , Adult , Aged , Aminopeptidases/analysis , Female , Humans , Male , Middle Aged , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery , Pyrrolidonecarboxylic Acid/analogs & derivatives , Receptors, Thyrotropin-Releasing Hormone/analysis , Signal Transduction , Thyrotropin-Releasing Hormone/analysis
17.
Neurol Res ; 22(4): 401-3, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10874690

ABSTRACT

To study the pathophysiology of olivopontocerebellar atrophy (OPCA), we destroyed inferior olive nuclei of male Wistar rats using 3-acetyl pyridine (3-AP) + harmaline + niacinamide. These rats showed a sluggish and ataxic gait. To elucidate the relationship between thyrotropin releasing hormone (TRH) in the Purkinje cell of cerebellum and the inferior olive nucleus, we investigated the concentrations of TRH in the cerebellar cortex, nuclei, and medulla oblongata including the inferior olive nuclei using radioimmunoassay method as well as TRH receptor in the Purkinje cells of cerebellum using immunohistochemical method. All statistical comparisons were done using non-parametric tests (Mann-Whitney U-test). We found that two weeks after the treatment, TRH concentrations in the cerebellar cortex as well as nuclei were significantly lower than in the controls but no significant difference in the medulla oblongata was observed between 3-AP treated rats and controls. Moreover, four weeks after the treatment, TRH-receptor positive Purkinje cell counts were significantly fewer than in the controls. These results suggest that TRH in the Purkinje cell of cerebellum may play a role in the ataxic gait observed in the rats whose inferior olive were destroyed.


Subject(s)
Olivary Nucleus/pathology , Purkinje Cells/chemistry , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyrotropin-Releasing Hormone/analysis , Animals , Ataxia/chemically induced , Ataxia/pathology , Cell Count , Denervation , Harmaline , Immunohistochemistry , Male , Neurotoxins , Niacinamide , Purkinje Cells/cytology , Pyridines , Radioimmunoassay , Rats , Rats, Wistar
18.
Endocr Regul ; 33(3): 135-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10571965

ABSTRACT

OBJECTIVE: To investigate the organ distribution of thyrotropin releasing hormone receptor (TRHR) type 2 in rats by immunohistochemical method. METHODS: TRHR type 2 was identified immunohistochemically in the rat tissues using specific anti-TRHR antiserum raised in New Zealand white rabbits immunized with a conjugate of synthetic TRHR type 2 (5-23) with bovine serum albumin. Immunohistochemical analysis was performed by avidin-biotin complex method. RESULTS: TRHR type 2 immunoreactivity was visualized in the central nervous system, anterior pituitary, gastric mucosa, Auerbach's and Meissner's nervous branch of the stomach, small intestine and colon, retina amd testis. Significant stain was detected in neural perikarya, axons and dendrites. When using antiserum preincubated with synthetic TRHR type 2(5-23) or anterior pituitary homogenates, no significant stain of anterior pituitary was detected. CONCLUSIONS: These findings suggest that TRHR type 2 is widely distributed and that the method used is valuable in studying the distribution of TRHR type 2 in rats.


Subject(s)
Immunohistochemistry , Receptors, Thyrotropin-Releasing Hormone/analysis , Animals , Avidin , Axons/chemistry , Biotin , Brain Chemistry , Colon/innervation , Dendrites/chemistry , Gastric Mucosa/chemistry , Intestine, Small/innervation , Male , Organ Specificity , Pituitary Gland, Anterior/chemistry , Rats , Rats, Wistar , Retina/chemistry , Spinal Cord/chemistry , Stomach/innervation , Testis/innervation
19.
Thyroid ; 8(10): 887-94, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9827655

ABSTRACT

Primary cultures of rat pituitary cells were stained with an antibody to the native thyrotropin-releasing hormone (TRH) receptor and with a bioactive, fluorescent analogue of TRH, Rhod-TRH. Rhod-TRH specifically stained 86% of lactotropes and 21% of nonlactotropes from primary pituitary cell cultures. Lactotropes and thyrotropes accounted for 90% of cells that stained with Rhod-TRH, but there were occasional lactotropes and thyrotropes that did not show detectable staining with antireceptor antibodies or with Rhod-TRH. The intensity of staining was generally higher in the GH3 line of tumor cells than in normal pituicytes, and 100% of the tumor cells stained with Rhod-TRH. To determine whether the TRH receptor undergoes ligand-directed endocytosis in normal cells, TRH receptor immunocytochemistry was performed before and after TRH binding. TRH receptors were localized on the surface of cells prior to TRH exposure, and Rhod-TRH fluorescence was confined to the plasma membrane when TRH binding was performed at 0 degrees C, where endocytosis is blocked. When cells were incubated with TRH at 37 degrees C, receptors were found in intracellular vesicles in both lactotropes and thyrotropes, and Rhod-TRH was rapidly internalized into endosomes at elevated temperatures. Internalization of Rhod-TRH was inhibited by hypertonic sucrose, indicating that it occurs through clathrin-coated pits. These findings show that some of the heterogeneity in the secretory and calcium responses of pituicytes to TRH occurs at the level of the TRH receptor.


Subject(s)
Pituitary Gland, Anterior/chemistry , Pituitary Gland, Anterior/metabolism , Prolactin/metabolism , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyrotropin/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Female , Fluorescent Antibody Technique , Fluorescent Dyes , Male , Molecular Sequence Data , Pituitary Neoplasms/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin-Releasing Hormone/metabolism , Rhodamines , Thyrotropin-Releasing Hormone/pharmacology
20.
Biol Pharm Bull ; 20(1): 86-7, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9013814

ABSTRACT

Thyrotropin-releasing hormone (TRH) and its analog, TA-0910, ameliorate the ataxia of the mutant mouse, rolling mouse Nagoya, by metabolic normalization in the ventral tegmental field (VTF). We here investigated the distribution of cerebral TRH receptors in the rolling mouse to clarify the sites of action of these drugs. TRH receptors were widely distributed in multiple brain areas, including in the VTF and the cuneiform nucleus (CnF) which terminates in the VTF. These results suggest that TRH and TA-0910 directly activate the VTF by acting on TRH receptors in the VTF and indirectly activate it through the receptors in the CnF.


Subject(s)
Ataxia/metabolism , Brain Chemistry , Receptors, Thyrotropin-Releasing Hormone/analysis , Animals , Male , Mice , Mice, Mutant Strains , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...