1
|
Niego A, Benítez-Burraco A. Are feralization and domestication truly mirror processes? ETHOL ECOL EVOL 2021. [DOI: 10.1080/03949370.2021.1975314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Amy Niego
- PhD Program, Faculty of Philology, University of Seville, C/Palos de la Frontera s/n, 41004 Sevilla, Spain
| | - Antonio Benítez-Burraco
- Department of Spanish, Linguistics, and Theory of Literature (Linguistics), Faculty of Philology, University of Seville, C/Palos de la Frontera s/n, 41004 Sevilla, Spain (E-mail: )
| |
Collapse
|
2
|
Alvarez-Salas E, González A, Amaya MI, de Gortari P. Accumbal TRH is downstream of the effects of isolation stress on hedonic food intake in rats. Nutr Neurosci 2021; 24:554-563. [PMID: 31438781 DOI: 10.1080/1028415x.2019.1657658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Emotional stress, through elevating corticosterone (CORT) levels may reduce feeding in rodents however when offered palatable food, stressed animals ingest more food compared to non-stressed controls. Nucleus accumbens (NAc) is part of the mesocorticolimbic system and participates in processing rewarding characteristics of food modulating palatable food intake, mainly when glucocorticoids are elevated. A possible mediator of CORT effects is accumbal thyrotropin-releasing hormone (TRH), which reduces chow intake in rats when administered into the NAc. We aimed to study the TRH role in hedonic feeding in stressed rats. For 14 days, animals with ad libitum access to chow or chow plus chocolate milk were either group-housed or singly-housed to induce stress. Rats with access to chocolate milk showed hyperphagia and decreased accumbal TRH mRNA levels, which were potentiated by stress. Results suggest that TRH downregulation was permissive of the increased palatable food intake. TRH injections into NAc of singly-housed animals with palatable food access reduced their food intake and increased serum CORT levels. The accumbal injections of a glucocorticoid receptor antagonist (mifepristone) in stressed rats with palatable food access, reduced only palatable food intake and increased accumbal TRH expression and serum CORT levels. This modulation of TRH mRNA when CORT signaling is modified suggests that accumbal TRH is downstream of glucocorticoids activity, which specifically increase palatable food intake. Our results strengthen the TRH involvement in regulating emotional aspects of hedonic feeding in stressed animals. Finding new therapies directed towards increasing TRHergic activity in NAc may be protective against overeating.
Collapse
Affiliation(s)
- Elena Alvarez-Salas
- Molecular Neurophysiology Laboratory, Neurosciences Research Department, National Institute of Psychiatry Ramón de la Fuente, México City, México
| | - Aldo González
- Molecular Neurophysiology Laboratory, Neurosciences Research Department, National Institute of Psychiatry Ramón de la Fuente, México City, México
| | - Maria Isabel Amaya
- Molecular Neurophysiology Laboratory, Neurosciences Research Department, National Institute of Psychiatry Ramón de la Fuente, México City, México
| | - Patricia de Gortari
- Molecular Neurophysiology Laboratory, Neurosciences Research Department, National Institute of Psychiatry Ramón de la Fuente, México City, México
| |
Collapse
|
3
|
Cardiovascular and body weight regulation changes in transgenic mice overexpressing thyrotropin-releasing hormone (TRH). J Physiol Biochem 2020; 76:599-608. [PMID: 32914279 DOI: 10.1007/s13105-020-00765-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/02/2020] [Indexed: 11/27/2022]
Abstract
Thyrotropin-releasing hormone (TRH) plays several roles as a hormone/neuropeptide. Diencephalic TRH (dTRH) participates in the regulation of blood pressure in diverse animal models, independently of the thyroid status. The present study aimed to evaluate whether chronic overexpression of TRH in mice affects cardiovascular and metabolic variables. We developed a transgenic (TG) mouse model that overexpresses dTrh. Despite having higher food consumption and water intake, TG mice showed significantly lower body weight respect to controls. Also, TG mice presented higher blood pressure, heart rate, and locomotor activity independently of thyroid hormone levels. These results and the higher urine noradrenaline excretion observed in TG mice suggest a higher metabolic rate mediated by sympathetic overflow. Cardiovascular changes were impeded by siRNA inhibition of the diencephalic Trh overexpression. Also, the silencing of dTRH in the TG mice normalized urine noradrenaline excretion, supporting the view that the cardiovascular effects of TRH involve the sympathetic system. Overall, we show that congenital dTrh overexpression leads to an increase in blood pressure accompanied by changes in body weight and food consumption mediated by a higher sympathetic overflow. These results provide new evidence confirming the participation of TRH in cardiovascular and body weight regulation.
Collapse
|
4
|
Tang J, Shen X, Ouyang H, Luo W, Huang Y, Tian Y, Zhang X. Transcriptome analysis of pituitary gland revealed candidate genes and gene networks regulating the growth and development in goose. Anim Biotechnol 2020; 33:429-439. [PMID: 32779547 DOI: 10.1080/10495398.2020.1801457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Goose is important meat poultry and its growth and development has always been the focus of attention, but the regulation mechanisms of genes and gene network underlying growth and development of goose are still unclear. Three males of large-size Shitou goose and three males of small size Wuzong goose at 5 weeks of age were used for transcriptome analysis with deep sequencing. After slaughter, their pituitary gland was taken for RNA-seq. A total of 290 DEGs were identified by fold change ≥2 and false discovery rate (FDR) <0.05, where there were 148 upregulated genes and 142 downregulated genes in Shitou goose compared to Wuzong goose. Results also showed that the DEGs related to insulin signaling pathway could increase protein synthesis and fat production, and the interaction network of DEGs was mainly related to development, endocrine system, inflammatory diseases, tissue damage and abnormality. The DEGs involved in the growth and function of the pituitary organs may regulate the growth and development of the body by affecting the synthesis and secretion of pituitary hormones. The results of this study will help to understand the regulatory mechanism of goose growth and development.
Collapse
Affiliation(s)
- Jun Tang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, China
| | - Xu Shen
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, China
| | - Hongjia Ouyang
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, China
| | - Wen Luo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yunmao Huang
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, China
| | - Yunbo Tian
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Zhang N, Zhang HY, Bi SA, Moran TH, Bi S. Differential regulation of thyrotropin-releasing hormone mRNA expression in the paraventricular nucleus and dorsomedial hypothalamus in OLETF rats. Neurosci Lett 2019; 703:79-85. [PMID: 30902570 DOI: 10.1016/j.neulet.2019.03.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 02/05/2023]
Abstract
Thyrotropin-releasing hormone (TRH) plays an important role in the regulation of energy balance. While the regulation of TRH in the paraventricular nucleus (PVN) in response to changes of energy balance has been well studied, how TRH is regulated in the dorsomedial hypothalamus (DMH) in maintaining energy homeostasis remains unclear. Here, we assessed the effects of food restriction and exercise on hypothalamic Trh expression using Otsuka Long-Evens Tokushima Fatty (OLETF) rats. Sedentary ad lib fed OLETF rats (OLETF-SED) became hyperphagic and obese. These alterations were prevented in OLETF rats with running wheel access (OLETF-RW) or food restriction in which their food was pair-fed (OLETF-PF) to the intake of lean control rats (LETO-SED). Evaluation of hypothalamic gene expression revealed that Trh mRNA expression was increased in the PVN of OLETF-SED rats and normalized in OLETF-RW and OLETF-PF rats compared to LETO-SED rats. In contrast, the expression of Trh in the DMH was decreased in OLETF-SED rats relative to LETO-SED rats. This alteration was reversed in OLETF-RW rats as seen in LETO-SED rats, but food restriction resulted in a significant increase in DMH Trh expression in OLETF-PF rats compared to LETO-SED rats. Strikingly, while Trh mRNA expression was decreased in the PVN of intact rats in response to acute food deprivation, food deprivation resulted in increased expression of Trh in the DMH. Together, these results demonstrate the differential regulation of Trh expression in the PVN and DMH in OLETF rats and suggest that DMH TRH also contributes to hypothalamic regulation of energy balance.
Collapse
Affiliation(s)
- Ni Zhang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai-Ying Zhang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sophia A Bi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sheng Bi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
Watanave M, Matsuzaki Y, Nakajima Y, Ozawa A, Yamada M, Hirai H. Contribution of Thyrotropin-Releasing Hormone to Cerebellar Long-Term Depression and Motor Learning. Front Cell Neurosci 2018; 12:490. [PMID: 30618637 PMCID: PMC6299015 DOI: 10.3389/fncel.2018.00490] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/29/2018] [Indexed: 11/17/2022] Open
Abstract
Thyrotropin-releasing hormone (TRH) regulates various physiological activities through activation of receptors expressed in a broad range of cells in the central nervous system. The cerebellum expresses TRH receptors in granule cells and molecular layer interneurons. However, the function of TRH in the cerebellum remains to be clarified. Here, using TRH knockout (KO) mice we studied the role of TRH in the cerebellum. Immunohistochemistry showed no gross morphological differences between KO mice and wild-type (WT) littermates in the cerebellum. In the rotarod test, the initial performance of KO mice was comparable to that of WT littermates, but the learning speed of KO mice was significantly lower than that of WT littermates, suggesting impaired motor learning. The motor learning deficit in KO mice was rescued by intraperitoneal injection of TRH. Electrophysiology revealed absence of long-term depression (LTD) at parallel fiber-Purkinje cell synapses in KO mice, which was rescued by bath-application of TRH. TRH was shown to increase cyclic guanosine monophosphate (cGMP) content in the cerebellum. Since nitric oxide (NO) stimulates cGMP synthesis in the cerebellum, we examined whether NO-cGMP pathway was involved in TRH-mediated LTD rescue in KO mice. Pharmacological blockade of NO synthase and subsequent cGMP production prevented TRH-induced LTD expression in KO mice, whereas increase in cGMP signal in Purkinje cells by 8-bromoguanosine cyclic 3',5'-monophosphate, a membrane-permeable cGMP analog, restored LTD without TRH application. These results suggest that TRH is involved in cerebellar LTD presumably by upregulating the basal cGMP level in Purkinje cells, and, consequently, in motor learning.
Collapse
Affiliation(s)
- Masashi Watanave
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasunori Matsuzaki
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuyo Nakajima
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Atsushi Ozawa
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
- Research Program for Neural Signalling, Division of Endocrinology, Metabolism and Signal Research, Gunma University Initiative for Advanced Research, Maebashi, Japan
| |
Collapse
|
7
|
Zhang Z, Machado F, Zhao L, Heinen CA, Foppen E, Ackermans MT, Zhou J, Bisschop PH, Boelen A, Fliers E, Kalsbeek A. Administration of Thyrotropin-Releasing Hormone in the Hypothalamic Paraventricular Nucleus of Male Rats Mimics the Metabolic Cold Defense Response. Neuroendocrinology 2018; 107:267-279. [PMID: 30092582 PMCID: PMC6390456 DOI: 10.1159/000492785] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/08/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cold exposure increases thyrotropin-releasing hormone (TRH) expression primarily in the hypothalamic paraventricular nucleus (PVN). The PVN is a well-known hypothalamic hub in the control of energy metabolism. TRH terminals and receptors are found on PVN neurons. We hypothesized that TRH release in the PVN plays an important role in the control of thermogenesis and energy mobilization during cold exposure. METHODS Male Wistar rats were exposed to a cold environment (4°C) or TRH retrodialysis in the PVN for 2 h. We compared the effects of cold exposure and TRH administration in the PVN on plasma glucose, corticosterone, and thyroid hormone concentrations, body temperature, locomotor activity, as well as metabolic gene expression in the liver and brown adipose tissue. RESULTS Cold exposure increased body temperature, locomotor activity, and plasma corticosterone concentrations, but blood glucose concentrations were similar to that of room temperature control animals. TRH administration in the PVN also promptly increased body temperature, locomotor activity and plasma corticosterone concentrations. However, TRH administration in the PVN markedly increased blood glucose concentrations and endogenous glucose production (EGP) compared to saline controls. Selective hepatic sympathetic or parasympathetic denervation reduced the TRH-induced increase in glucose concentrations and EGP. Gene expression data indicated increased gluconeogenesis in liver and lipolysis in brown adipose tissue, both after cold exposure and TRH administration. CONCLUSIONS We conclude that TRH administration in the rat PVN largely mimics the metabolic and behavioral changes induced by cold exposure indicating a potential link between TRH release in the PVN and cold defense.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Frederico Machado
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Li Zhao
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Charlotte A Heinen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ewout Foppen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jiangning Zhou
- CAS Key Laboratory of Brain Function and Disease, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anita Boelen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the
| |
Collapse
|
8
|
Affiliation(s)
- I. W. Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, United Kingdom
| |
Collapse
|
9
|
Abstract
Thyroid hormones (TH) are endocrine messengers essential for normal development and function of virtually every vertebrate. The hypothalamic-pituitary-thyroid axis is exquisitely modulated to maintain nearly constant TH (T4 and T3) levels in circulation. However peripheral tissues and the CNS control the intracellular availability of TH, suggesting that circulating concentrations of TH are not fully representative of what each cell type sees. Indeed, recent work in the field has identified that TH transporters, deiodinases and thyroid hormone receptor coregulators can strongly control tissue-specific sensitivity to a set amount of TH. Furthermore, the mechanism by which the thyroid hormone receptors regulate target gene expression can vary by gene, tissue and cellular context. This review will highlight novel insights into the machinery that controls the cellular response to TH, which include unique signaling cascades. These findings shed new light into the pathophysiology of human diseases caused by abnormal TH signaling.
Collapse
Affiliation(s)
- Arturo Mendoza
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Kolaj M, Zhang L, Renaud LP. L-type calcium channels and MAP kinase contribute to thyrotropin-releasing hormone-induced depolarization in thalamic paraventricular nucleus neurons. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1120-7. [PMID: 27009047 PMCID: PMC4935505 DOI: 10.1152/ajpregu.00082.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 03/18/2016] [Indexed: 12/11/2022]
Abstract
In rat paraventricular thalamic nucleus (PVT) neurons, activation of thyrotropin-releasing hormone (TRH) receptors enhances neuronal excitability via concurrent decrease in a G protein-coupled inwardly rectifying K (GIRK)-like conductance and opening of a cannabinoid receptor-sensitive transient receptor potential canonical (TRPC)-like conductance. Here, we investigated the calcium (Ca(2+)) contribution to the components of this TRH-induced response. TRH-induced membrane depolarization was reduced in the presence of intracellular BAPTA, also in media containing nominally zero [Ca(2+)]o, suggesting a critical role for both intracellular Ca(2+) release and Ca(2+) influx. TRH-induced inward current was unchanged by T-type Ca(2+) channel blockade, but was decreased by blockade of high-voltage-activated Ca(2+) channels (HVACCs). Both the pharmacologically isolated GIRK-like and the TRPC-like components of the TRH-induced response were decreased by nifedipine and increased by BayK8644, implying Ca(2+) influx via L-type Ca(2+) channels. Only the TRPC-like conductance was reduced by either thapsigargin or dantrolene, suggesting a role for ryanodine receptors and Ca(2+)-induced Ca(2+) release in this component of the TRH-induced response. In pituitary and other cell lines, TRH stimulates MAPK. In PVT neurons, only the GIRK-like component of the TRH-induced current was selectively decreased in the presence of PD98059, a MAPK inhibitor. Collectively, the data imply that TRH-induced depolarization and inward current in PVT neurons involve both a dependency on extracellular Ca(2+) influx via opening of L-type Ca(2+) channels, a sensitivity of a TRPC-like component to intracellular Ca(2+) release via ryanodine channels, and a modulation by MAPK of a GIRK-like conductance component.
Collapse
Affiliation(s)
- Miloslav Kolaj
- Ottawa Hospital Research Institute, Neuroscience Program and University of Ottawa, Department of Medicine, Ottawa, Ontario, Canada
| | - Li Zhang
- Ottawa Hospital Research Institute, Neuroscience Program and University of Ottawa, Department of Medicine, Ottawa, Ontario, Canada
| | - Leo P Renaud
- Ottawa Hospital Research Institute, Neuroscience Program and University of Ottawa, Department of Medicine, Ottawa, Ontario, Canada
| |
Collapse
|
11
|
Shevchenko KV, Vyunova TV, Radilov AS, Andreeva LA, Nagaev IY, Shevchenko VP, Rembovsky VR, Myasoedov NF. Penetration of thyroliberin in the blood and brain regions at intranasal or intravenous administration. DOKL BIOCHEM BIOPHYS 2016; 465:436-9. [PMID: 26728743 DOI: 10.1134/s1607672915060228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Indexed: 11/22/2022]
Abstract
The maximum amounts of the thyroliberin in the blood and brain of rats at intranasal and intravenous administration were determined. It is found that rat hippocampal, cortical, and cerebellar membranes contain two types of specific binding sites (high- and low-affinity) for the labeled ligand. It was shown that, at intranasal and intravenous administration, maximum amounts of the thyroliberin were detected in the cerebellum and then in the cortex and hippocampus. The degradation of the thyroliberin in the rat brain and its regions at intranasal and intravenous administration was studied. It is shown that the degree of degradation and the formation of proteolytic products of the thyroliberin is different in different regions of the rat brain.
Collapse
Affiliation(s)
- K V Shevchenko
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| | - T V Vyunova
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| | - A S Radilov
- Research Institute of Hygiene, Occupational Pathology, and Human Ecology, Federal Medical-Biological Agency, Ministry of Public Health of the Russian Federation, p/o Kuz'molovskii, Leningrad oblast, 188663, Russia
| | - L A Andreeva
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| | - I Yu Nagaev
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| | - V P Shevchenko
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| | - V R Rembovsky
- Research Institute of Hygiene, Occupational Pathology, and Human Ecology, Federal Medical-Biological Agency, Ministry of Public Health of the Russian Federation, p/o Kuz'molovskii, Leningrad oblast, 188663, Russia.
| | - N F Myasoedov
- Institute of Molecular Genetics, Russian Academy of Sciences, pl. Akademika Kurchatova 46, Moscow, 123182, Russia
| |
Collapse
|
12
|
Meena CL, Ingole S, Rajpoot S, Thakur A, Nandeker PP, Sangamwar AT, Sharma SS, Jain R. Discovery of a low affinity thyrotropin-releasing hormone (TRH)-like peptide that exhibits potent inhibition of scopolamine-induced memory impairment in mice. RSC Adv 2015; 5:56872-56884. [PMID: 26191403 PMCID: PMC4501038 DOI: 10.1039/c5ra06935a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
TRH-like peptides were synthesized in which the critical N-terminus residue L-pGlu was replaced with various heteroaromatic rings, and the central residue histidine with 1-alkyl-L-histidines. All synthesized TRH-like peptides were evaluated in vitro as agonists in HEK mTRH-R1 and HEK mTRH-R2 cell lines, an expressing receptor binding assay (IC50), and cell signaling assay (EC50). The analeptic potential of the synthesized peptides was evaluated in vivo by using the antagonism of a pentobarbital-induced sleeping time. The peptides 6a, 6c and 6e were found to activate TRH-R2 with potencies (EC50) of 0.002 μM, 0.28 μM and 0.049 μM, respectively. In contrast, for signaling activation of TRH-R1, the same peptides required higher concentration of 0.414 μM, 50 μM and 19.1 μM, respectively in the FLIPR assay. The results showed that these peptides were 207, 178 and 389-fold selective towards TRH-R2 receptor subtype. In the antagonism of a pentobarbital-induced sleeping time assay, peptide 6c showed a 58.5% reduction in sleeping time. The peptide 6c exhibited high stability in rat blood plasma, a superior effect on the scopolamine-induced cognition impairment mice model, safe effects on the cardiovascular system, and general behavior using a functional observation battery (FOB).
Collapse
Affiliation(s)
- Chhuttan L. Meena
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Shubdha Ingole
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Satyendra Rajpoot
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Avinash Thakur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Prajwal P. Nandeker
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Abhay T. Sangamwar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Shyam S. Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| |
Collapse
|
13
|
Fekete C, Lechan RM. Central regulation of hypothalamic-pituitary-thyroid axis under physiological and pathophysiological conditions. Endocr Rev 2014; 35:159-94. [PMID: 24423980 PMCID: PMC3963261 DOI: 10.1210/er.2013-1087] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 11/05/2013] [Indexed: 12/18/2022]
Abstract
TRH is a tripeptide amide that functions as a neurotransmitter but also serves as a neurohormone that has a critical role in the central regulation of the hypothalamic-pituitary-thyroid axis. Hypophysiotropic TRH neurons involved in this neuroendocrine process are located in the hypothalamic paraventricular nucleus and secrete TRH into the pericapillary space of the external zone of the median eminence for conveyance to anterior pituitary thyrotrophs. Under basal conditions, the activity of hypophysiotropic TRH neurons is regulated by the negative feedback effects of thyroid hormone to ensure stable, circulating, thyroid hormone concentrations, a mechanism that involves complex interactions between hypophysiotropic TRH neurons and the vascular system, cerebrospinal fluid, and specialized glial cells called tanycytes. Hypophysiotropic TRH neurons also integrate other humoral and neuronal inputs that can alter the setpoint for negative feedback regulation by thyroid hormone. This mechanism facilitates adaptation of the organism to changing environmental conditions, including the shortage of food and a cold environment. The thyroid axis is also affected by other adverse conditions such as infection, but the central mechanisms mediating suppression of hypophysiotropic TRH may be pathophysiological. In this review, we discuss current knowledge about the mechanisms that contribute to the regulation of hypophysiotropic TRH neurons under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Csaba Fekete
- Department of Endocrine Neurobiology (C.F.), Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083 Budapest, Hungary; Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism (C.F., R.M.L.), Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Department of Neuroscience (R.M.L.), Tufts University School of Medicine, Boston, Massachusetts 02111
| | | |
Collapse
|
14
|
Henkin RI, Potolicchio SJ, Levy LM. Olfactory Hallucinations without Clinical Motor Activity: A Comparison of Unirhinal with Birhinal Phantosmia. Brain Sci 2013; 3:1483-553. [PMID: 24961619 PMCID: PMC4061890 DOI: 10.3390/brainsci3041483] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 01/14/2023] Open
Abstract
Olfactory hallucinations without subsequent myoclonic activity have not been well characterized or understood. Herein we describe, in a retrospective study, two major forms of olfactory hallucinations labeled phantosmias: one, unirhinal, the other, birhinal. To describe these disorders we performed several procedures to elucidate similarities and differences between these processes. From 1272, patients evaluated for taste and smell dysfunction at The Taste and Smell Clinic, Washington, DC with clinical history, neurological and otolaryngological examinations, evaluations of taste and smell function, EEG and neuroradiological studies 40 exhibited cyclic unirhinal phantosmia (CUP) usually without hyposmia whereas 88 exhibited non-cyclic birhinal phantosmia with associated symptomology (BPAS) with hyposmia. Patients with CUP developed phantosmia spontaneously or after laughing, coughing or shouting initially with spontaneous inhibition and subsequently with Valsalva maneuvers, sleep or nasal water inhalation; they had frequent EEG changes usually ipsilateral sharp waves. Patients with BPAS developed phantosmia secondary to several clinical events usually after hyposmia onset with few EEG changes; their phantosmia could not be initiated or inhibited by any physiological maneuver. CUP is uncommonly encountered and represents a newly defined clinical syndrome. BPAS is commonly encountered, has been observed previously but has not been clearly defined. Mechanisms responsible for phantosmia in each group were related to decreased gamma-aminobutyric acid (GABA) activity in specific brain regions. Treatment which activated brain GABA inhibited phantosmia in both groups.
Collapse
Affiliation(s)
- Robert I Henkin
- Center for Molecular Nutrition and Sensory Disorders, The Taste and Smell Clinic, 5125 MacArthur Blvd, NW, Suite 20, Washington, DC 20016, USA.
| | - Samuel J Potolicchio
- Department of Neurology, The George Washington University Medical Center, 2150 Pennsylvania Avenue, NW, 7th Floor, Washington, DC 20037, USA.
| | - Lucien M Levy
- Department of Radiology, The George Washington University Medical Center, 900 23rd Street, NW, Washington, DC 20037, USA.
| |
Collapse
|
15
|
Chrétien M. How the prohormone theory solved two important controversies in hormonal and neural Peptide biosynthesis. Front Endocrinol (Lausanne) 2013; 4:148. [PMID: 24167501 PMCID: PMC3805937 DOI: 10.3389/fendo.2013.00148] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 10/02/2013] [Indexed: 11/29/2022] Open
Abstract
This Prohormone Theory was simultaneously proposed in 1967 by two independent groups using two different approaches and two experimental models. Donald Steiner, in elegant pulse-chase experiments, proposed the existence of proinsulin when he observed that a human insulinoma was producing higher MW forms of immunoreactive insulin, subsequently transformed into insulin-like material (1). Simultaneously and independently, Michel Chrétien, based on amino acid sequence homologies between three pituitary peptides, β-lipotropic hormone (β-LPH), γ-LPH, and β-melanocyte-stimulating hormone (β-MSH), concluded that active peptide hormones are derived from endoproteolytic cleavages of inactive precursors, apparently at pairs of basic amino acids (2). One year later, Donald Chance confirmed that the cleavage sites in proinsulin were also made of paired basic amino acids (3). This novel paradigm solved two major controversies on the biosynthesis of both insulin and neuropeptides. This short review describes how.
Collapse
Affiliation(s)
- Michel Chrétien
- Functional Endoproteolysis Laboratory, Institut de Recherches Cliniques de Montréal, Ottawa Hospital Research Institute , Montreal, QC , Canada
| |
Collapse
|
16
|
Glutaminyl cyclase-mediated toxicity of pyroglutamate-beta amyloid induces striatal neurodegeneration. BMC Neurosci 2013; 14:108. [PMID: 24083638 PMCID: PMC3850634 DOI: 10.1186/1471-2202-14-108] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
Background Posttranslational modifications of beta amyloid (Aβ) have been shown to affect its biophysical and neurophysiological properties. One of these modifications is N-terminal pyroglutamate (pE) formation. Enzymatic glutaminyl cyclase (QC) activity catalyzes cyclization of truncated Aβ(3-x), generating pE3-Aβ. Compared to unmodified Aβ, pE3-Aβ is more hydrophobic and neurotoxic. In addition, it accelerates aggregation of other Aβ species. To directly investigate pE3-Aβ formation and toxicity in vivo, transgenic (tg) ETNA (E at the truncated N-terminus of Aβ) mice expressing truncated human Aβ(3–42) were generated and comprehensively characterized. To further investigate the role of QC in pE3-Aβ formation in vivo, ETNA mice were intercrossed with tg mice overexpressing human QC (hQC) to generate double tg ETNA-hQC mice. Results Expression of truncated Aβ(3–42) was detected mainly in the lateral striatum of ETNA mice, leading to progressive accumulation of pE3-Aβ. This ultimately resulted in astrocytosis, loss of DARPP-32 immunoreactivity, and neuronal loss at the sites of pE3-Aβ formation. Neuropathology in ETNA mice was associated with behavioral alterations. In particular, hyperactivity and impaired acoustic sensorimotor gating were detected. Double tg ETNA-hQC mice showed similar Aβ levels and expression sites, while pE3-Aβ were significantly increased, entailing increased astrocytosis and neuronal loss. Conclusions ETNA and ETNA-hQC mice represent novel mouse models for QC-mediated toxicity of truncated and pE-modified Aβ. Due to their significant striatal neurodegeneration these mice can also be used for analysis of striatal regulation of basal locomotor activity and sensorimotor gating, and possibly for DARPP-32-dependent neurophysiology and neuropathology. The spatio-temporal correlation of pE3-Aβ and neuropathology strongly argues for an important role of this Aβ species in neurodegenerative processes in these models.
Collapse
|
17
|
Quintanar JL, Guzmán-Soto I. Hypothalamic neurohormones and immune responses. Front Integr Neurosci 2013; 7:56. [PMID: 23964208 PMCID: PMC3741963 DOI: 10.3389/fnint.2013.00056] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/16/2013] [Indexed: 01/19/2023] Open
Abstract
The aim of this review is to provide a comprehensive examination of the current literature describing the neural-immune interactions, with emphasis on the most recent findings of the effects of neurohormones on immune system. Particularly, the role of hypothalamic hormones such as Thyrotropin-releasing hormone (TRH), Corticotropin-releasing hormone (CRH) and Gonadotropin-releasing hormone (GnRH). In the past few years, interest has been raised in extrapituitary actions of these neurohormones due to their receptors have been found in many non-pituitary tissues. Also, the receptors are present in immune cells, suggesting an autocrine or paracrine role within the immune system. In general, these neurohormones have been reported to exert immunomodulatory effects on cell proliferation, immune mediators release and cell function. The implications of these findings in understanding the network of hypothalamic neuropeptides and immune system are discussed.
Collapse
Affiliation(s)
- J Luis Quintanar
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes Aguascalientes, México
| | | |
Collapse
|
18
|
Development of the serotonergic cells in murine raphe nuclei and their relations with rhombomeric domains. Brain Struct Funct 2012; 218:1229-77. [PMID: 23052546 PMCID: PMC3748323 DOI: 10.1007/s00429-012-0456-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 09/08/2012] [Indexed: 12/18/2022]
Abstract
The raphe nuclei represent the origin of central serotonergic projections. The literature distinguishes seven nuclei grouped into rostral and caudal clusters relative to the pons. The boundaries of these nuclei have not been defined precisely enough, particularly with regard to developmental units, notably hindbrain rhombomeres. We hold that a developmental point of view considering rhombomeres may explain observed differences in connectivity and function. There are twelve rhombomeres characterized by particular genetic profiles, and each develops between one and four distinct serotonergic populations. We have studied the distribution of the conventional seven raphe nuclei among these twelve units. To this aim, we correlated 5-HT-immunoreacted neurons with rhombomeric boundary landmarks in sagittal mouse brain sections at different developmental stages. Furthermore, we performed a partial genoarchitectonic analysis of the developing raphe nuclei, mapping all known serotonergic differentiation markers, and compared these results, jointly with others found in the literature, with our map of serotonin-containing populations, in order to examine regional variations in correspondence. Examples of regionally selective gene patterns were identified. As a result, we produced a rhombomeric classification of some 45 serotonergic populations, and suggested a corresponding modified terminology. Only a minor rostral part of the dorsal raphe nucleus lies in the midbrain. Some serotonergic neurons were found in rhombomere 4, contrary to the conventional assumption that it lacks such neurons. We expect that our reclassification of raphe nuclei may be useful for causal analysis of their differential molecular specification, as well as for studies of differential connectivity and function.
Collapse
|
19
|
Yin P, Bousquet-Moore D, Annangudi SP, Southey BR, Mains RE, Eipper BA, Sweedler JV. Probing the production of amidated peptides following genetic and dietary copper manipulations. PLoS One 2011; 6:e28679. [PMID: 22194882 PMCID: PMC3241674 DOI: 10.1371/journal.pone.0028679] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 11/13/2011] [Indexed: 11/23/2022] Open
Abstract
Amidated neuropeptides play essential roles throughout the nervous and endocrine systems. Mice lacking peptidylglycine α-amidating monooxygenase (PAM), the only enzyme capable of producing amidated peptides, are not viable. In the amidation reaction, the reactant (glycine-extended peptide) is converted into a reaction intermediate (hydroxyglycine-extended peptide) by the copper-dependent peptidylglycine-α-hydroxylating monooxygenase (PHM) domain of PAM. The hydroxyglycine-extended peptide is then converted into amidated product by the peptidyl-α-hydroxyglycine α-amidating lyase (PAL) domain of PAM. PHM and PAL are stitched together in vertebrates, but separated in some invertebrates such as Drosophila and Hydra. In addition to its luminal catalytic domains, PAM includes a cytosolic domain that can enter the nucleus following release from the membrane by γ-secretase. In this work, several glycine- and hydroxyglycine-extended peptides as well as amidated peptides were qualitatively and quantitatively assessed from pituitaries of wild-type mice and mice with a single copy of the Pam gene (PAM+/−) via liquid chromatography-mass spectrometry-based methods. We provide the first evidence for the presence of a peptidyl-α-hydroxyglycine in vivo, indicating that the reaction intermediate becomes free and is not handed directly from PHM to PAL in vertebrates. Wild-type mice fed a copper deficient diet and PAM+/− mice exhibit similar behavioral deficits. While glycine-extended reaction intermediates accumulated in the PAM+/− mice and reflected dietary copper availability, amidated products were far more prevalent under the conditions examined, suggesting that the behavioral deficits observed do not simply reflect a lack of amidated peptides.
Collapse
Affiliation(s)
- Ping Yin
- Department of Chemistry, Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Danielle Bousquet-Moore
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Suresh P. Annangudi
- Department of Chemistry, Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Bruce R. Southey
- Department of Chemistry, Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Richard E. Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Betty A. Eipper
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- * E-mail: (JVS); (BAE)
| | - Jonathan V. Sweedler
- Department of Chemistry, Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail: (JVS); (BAE)
| |
Collapse
|
20
|
Alexandru A, Jagla W, Graubner S, Becker A, Bäuscher C, Kohlmann S, Sedlmeier R, Raber KA, Cynis H, Rönicke R, Reymann KG, Petrasch-Parwez E, Hartlage-Rübsamen M, Waniek A, Rossner S, Schilling S, Osmand AP, Demuth HU, von Hörsten S. Selective hippocampal neurodegeneration in transgenic mice expressing small amounts of truncated Aβ is induced by pyroglutamate-Aβ formation. J Neurosci 2011; 31:12790-801. [PMID: 21900558 PMCID: PMC6623394 DOI: 10.1523/jneurosci.1794-11.2011] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 07/09/2011] [Accepted: 07/12/2011] [Indexed: 12/24/2022] Open
Abstract
Posttranslational amyloid-β (Aβ) modification is considered to play an important role in Alzheimer's disease (AD) etiology. An N-terminally modified Aβ species, pyroglutamate-amyloid-β (pE3-Aβ), has been described as a major constituent of Aβ deposits specific to human AD but absent in normal aging. Formed via cyclization of truncated Aβ species by glutaminyl cyclase (QC; QPCT) and/or its isoenzyme (isoQC; QPCTL), pE3-Aβ aggregates rapidly and is known to seed additional Aβ aggregation. To directly investigate pE3-Aβ toxicity in vivo, we generated and characterized transgenic TBA2.1 and TBA2.2 mice, which express truncated mutant human Aβ. Along with a rapidly developing behavioral phenotype, these mice showed progressively accumulating Aβ and pE3-Aβ deposits in brain regions of neuronal loss, impaired long-term potentiation, microglial activation, and astrocytosis. Illustrating a threshold for pE3-Aβ neurotoxicity, this phenotype was not found in heterozygous animals but in homozygous TBA2.1 or double-heterozygous TBA2.1/2.2 animals only. A significant amount of pE3-Aβ formation was shown to be QC-dependent, because crossbreeding of TBA2.1 with QC knock-out, but not isoQC knock-out, mice significantly reduced pE3-Aβ levels. Hence, lowering the rate of QC-dependent posttranslational pE3-Aβ formation can, in turn, lower the amount of neurotoxic Aβ species in AD.
Collapse
MESH Headings
- Aging/pathology
- Aging/psychology
- Alzheimer Disease/pathology
- Amyloid beta-Protein Precursor/biosynthesis
- Animals
- Behavior, Animal
- Brain/pathology
- Enzyme-Linked Immunosorbent Assay
- Gliosis/pathology
- Heredodegenerative Disorders, Nervous System/genetics
- Heredodegenerative Disorders, Nervous System/pathology
- Heredodegenerative Disorders, Nervous System/psychology
- Hippocampus/pathology
- Humans
- Immunohistochemistry
- Kinetics
- Long-Term Potentiation/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Microscopy, Electron
- Neuronal Plasticity/genetics
- Neuronal Plasticity/physiology
- Phenotype
- Postural Balance/physiology
- Protein Processing, Post-Translational
- Pyrrolidonecarboxylic Acid/metabolism
- Reflex, Startle/physiology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kerstin A. Raber
- Experimental Therapy, Friedrich Alexander University Erlangen Nürnberg, 91054 Erlangen, Germany
| | | | - Raik Rönicke
- German Center of Neurodegenerative Diseases, 39120 Magdeburg, Germany
| | - Klaus G. Reymann
- German Center of Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | | | - Maike Hartlage-Rübsamen
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04109 Leipzig, Germany, and
| | - Alexander Waniek
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04109 Leipzig, Germany, and
| | - Steffen Rossner
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04109 Leipzig, Germany, and
| | | | - Alexander P. Osmand
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920
| | - Hans-Ulrich Demuth
- Ingenium Pharmaceuticals, 82152 Martinsried, Germany
- Probiodrug, 06120 Halle/Saale, Germany
| | - Stephan von Hörsten
- Experimental Therapy, Friedrich Alexander University Erlangen Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
21
|
Cynis H, Hoffmann T, Friedrich D, Kehlen A, Gans K, Kleinschmidt M, Rahfeld JU, Wolf R, Wermann M, Stephan A, Haegele M, Sedlmeier R, Graubner S, Jagla W, Müller A, Eichentopf R, Heiser U, Seifert F, Quax PHA, de Vries MR, Hesse I, Trautwein D, Wollert U, Berg S, Freyse EJ, Schilling S, Demuth HU. The isoenzyme of glutaminyl cyclase is an important regulator of monocyte infiltration under inflammatory conditions. EMBO Mol Med 2011; 3:545-58. [PMID: 21774078 PMCID: PMC3377097 DOI: 10.1002/emmm.201100158] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/01/2011] [Accepted: 06/08/2011] [Indexed: 02/06/2023] Open
Abstract
Acute and chronic inflammatory disorders are characterized by detrimental cytokine and chemokine expression. Frequently, the chemotactic activity of cytokines depends on a modified N-terminus of the polypeptide. Among those, the N-terminus of monocyte chemoattractant protein 1 (CCL2 and MCP-1) is modified to a pyroglutamate (pE-) residue protecting against degradation in vivo. Here, we show that the N-terminal pE-formation depends on glutaminyl cyclase activity. The pE-residue increases stability against N-terminal degradation by aminopeptidases and improves receptor activation and signal transduction in vitro. Genetic ablation of the glutaminyl cyclase iso-enzymes QC (QPCT) or isoQC (QPCTL) revealed a major role of isoQC for pE1-CCL2 formation and monocyte infiltration. Consistently, administration of QC-inhibitors in inflammatory models, such as thioglycollate-induced peritonitis reduced monocyte infiltration. The pharmacologic efficacy of QC/isoQC-inhibition was assessed in accelerated atherosclerosis in ApoE3*Leiden mice, showing attenuated atherosclerotic pathology following chronic oral treatment. Current strategies targeting CCL2 are mainly based on antibodies or spiegelmers. The application of small, orally available inhibitors of glutaminyl cyclases represents an alternative therapeutic strategy to treat CCL2-driven disorders such as atherosclerosis/restenosis and fibrosis.
Collapse
|
22
|
Abstract
Central hypothyroidism is defined as hypothyroidism due to insufficient stimulation by thyroid stimulating hormone (TSH) of an otherwise normal thyroid gland. It has an estimated prevalence of approximately 1 in 80,000 to 1 in 120,000. It can be secondary hypothyroidism (pituitary) or tertiary hypothyroidism (hypothalamus) in origin. In children, it is usually caused by craniopharyngiomas or previous cranial irradiation for brain tumors or hematological malignancies. In adults, it is usually due to pituitary macroadenomas, pituitary surgeries or post-irradiation. Fatigue and peripheral edema are the most specific clinical features. Diagnosis is established by the presence of normal to low-normal TSH on the background of low-normal thyroid hormones, confirmed by the thyrotropin releasing hormone stimulation test. Therapy includes use of levothyroxine titrated to improvement in symptomology and keeping free T4 in the upper limit of normal reference range.
Collapse
Affiliation(s)
- Vishal Gupta
- Department of Endocrinology, Jaslok Hospital and Research Centre, 15-Deshmukh Marg, Mumbai - 400 026, India
| | - Marilyn Lee
- Department of General Medicine/Endocrinology, Khoo Teck Puat Hospital, Singapore
| |
Collapse
|
23
|
Schilling S, Kohlmann S, Bäuscher C, Sedlmeier R, Koch B, Eichentopf R, Becker A, Cynis H, Hoffmann T, Berg S, Freyse EJ, von Hörsten S, Rossner S, Graubner S, Demuth HU. Glutaminyl cyclase knock-out mice exhibit slight hypothyroidism but no hypogonadism: implications for enzyme function and drug development. J Biol Chem 2011; 286:14199-208. [PMID: 21330373 PMCID: PMC3077621 DOI: 10.1074/jbc.m111.229385] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Indexed: 11/06/2022] Open
Abstract
Glutaminyl cyclases (QCs) catalyze the formation of pyroglutamate (pGlu) residues at the N terminus of peptides and proteins. Hypothalamic pGlu hormones, such as thyrotropin-releasing hormone and gonadotropin-releasing hormone are essential for regulation of metabolism and fertility in the hypothalamic pituitary thyroid and gonadal axes, respectively. Here, we analyzed the consequences of constitutive genetic QC ablation on endocrine functions and on the behavior of adult mice. Adult homozygous QC knock-out mice are fertile and behave indistinguishably from wild type mice in tests of motor function, cognition, general activity, and ingestion behavior. The QC knock-out results in a dramatic drop of enzyme activity in the brain, especially in hypothalamus and in plasma. Other peripheral organs like liver and spleen still contain QC activity, which is most likely caused by its homolog isoQC. The serum gonadotropin-releasing hormone, TSH, and testosterone concentrations were not changed by QC depletion. The serum thyroxine was decreased by 24% in homozygous QC knock-out animals, suggesting a mild hypothyroidism. QC knock-out mice were indistinguishable from wild type with regard to blood glucose and glucose tolerance, thus differing from reports of thyrotropin-releasing hormone knock-out mice significantly. The results suggest a significant formation of the hypothalamic pGlu hormones by alternative mechanisms, like spontaneous cyclization or conversion by isoQC. The different effects of QC depletion on the hypothalamic pituitary thyroid and gonadal axes might indicate slightly different modes of substrate conversion of both enzymes. The absence of significant abnormalities in QC knock-out mice suggests the presence of a therapeutic window for suppression of QC activity in current drug development.
Collapse
Affiliation(s)
| | | | | | | | - Birgit Koch
- From Probiodrug AG, Weinbergweg 22, 06120 Halle/Saale
| | | | | | - Holger Cynis
- From Probiodrug AG, Weinbergweg 22, 06120 Halle/Saale
| | | | - Sabine Berg
- the Institute of Diabetes, “Gerhardt Katsch,” 17495 Karlsburg
| | | | - Stephan von Hörsten
- the University of Erlangen-Nürnberg, Franz-Penzoldt-Center, Palmsanlage 5, 91054 Erlangen, and
| | - Steffen Rossner
- the Paul Flechsig Institute for Brain Research, University of Leipzig, Jahnallee 59, 04109 Leipzig, Germany
| | | | - Hans-Ulrich Demuth
- From Probiodrug AG, Weinbergweg 22, 06120 Halle/Saale
- Ingenium GmbH, Fraunhoferstrasse 13, 82152 Martinsried
| |
Collapse
|
24
|
Monga V, Meena CL, Rajput S, Pawar C, Sharma SS, Lu X, Gershengorn MC, Jain R. Synthesis, receptor binding, and CNS pharmacological studies of new thyrotropin-releasing hormone (TRH) analogues. ChemMedChem 2011; 6:531-43. [PMID: 21302359 DOI: 10.1002/cmdc.201000481] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 01/06/2011] [Indexed: 02/03/2023]
Abstract
As part of our search for selective and CNS-active thyrotropin-releasing hormone (TRH) analogues, we synthesized a set of 44 new analogues in which His and pGlu residues were modified or replaced. The analogues were evaluated as agonists at TRH-R1 and TRH-R2 in cells in vitro, and in vivo in mice for analeptic and anticonvulsant activities. Several analogues bound to TRH-R1 and TRH-R2 with good to moderate affinities, and are full agonists at both receptor subtypes. Specifically, analogue 21 a (R=CH3) exhibited binding affinities (Ki values) of 0.17 μM for TRH-R1 and 0.016 μM for TRH-R2; it is 10-fold less potent than TRH in binding to TRH-R1 and equipotent with TRH in binding to TRH-R2. Compound 21 a, the most selective agonist, activated TRH-R2 with a potency (EC50 value) of 0.0021 μM, but activated TRH-R1 at EC50=0.05 μM, and exhibited 24-fold selectivity for TRH-R2 over TRH-R1. The newly synthesized TRH analogues were also evaluated in vivo to assess their potencies in antagonism of barbiturate-induced sleeping time, and several analogues displayed potent analeptic activity. Specifically, analogues 21 a,b and 22 a,b decreased sleeping time by nearly 50% more than TRH. These analogues also displayed potent anticonvulsant activity and provided significant protection against PTZ-induced seizures, but failed to provide any protection in MES-induced seizures at 10 μmol kg(-1). The results of this study provide evidence that TRH analogues that show selectivity for TRH-R2 over TRH-R1 possess potent CNS activity.
Collapse
Affiliation(s)
- Vikramdeep Monga
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Perello M, Cakir I, Cyr NE, Romero A, Stuart RC, Chiappini F, Hollenberg AN, Nillni EA. Maintenance of the thyroid axis during diet-induced obesity in rodents is controlled at the central level. Am J Physiol Endocrinol Metab 2010; 299:E976-89. [PMID: 20858755 PMCID: PMC3006258 DOI: 10.1152/ajpendo.00448.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The hypothalamic-pituitary-thyroid (HPT) axis is a major contributor in maintaining energy expenditure and body weight, and the adipocyte hormone leptin regulates this axis by increasing TRH levels in the fed state. Leptin stimulates TRH directly in the hypothalamic paraventricular nucleus (PVN; direct pathway) and indirectly by regulating proopiomelnocortin neurons in the hypothalamic arcuate nucleus (ARC; indirect pathway). Whereas the indirect pathway is fully functional in lean animals, it is inactive during diet-induced obesity (DIO) because of the establishment of leptin resistance. Despite this, the HPT axis activity in obese humans and rodents remains within the normal levels or slightly higher. Therefore, in this study, we aimed to determine the mechanism(s) by which the HPT axis is still active despite leptin resistance. With a combination of using the Sprague-Dawley rat physiological model and the Zuker rat that bears a mutation in the leptin receptor, we were able to demonstrate that under DIO conditions the HPT axis is regulated at the central level, but only through the direct pathway of leptin action on TRH neurons. Deiodinase enzymes, which are present in many tissues and responsible for converting thyroid hormones, were not statistically different between lean and DIO animals. These data suggest that the increase in T(4/3) seen in obese animals is due mostly to central leptin action. We also found that T(3) feedback inhibition on the prepro-TRH gene is controlled partially by leptin-induced pSTAT3 signaling via the TRH promoter. This interactive relationship between T(3) and pSTAT3 signaling appears essential to maintain the HPT axis at normal levels in conditions such as obesity.
Collapse
Affiliation(s)
- Mario Perello
- Div. of Endocrinology, Brown Medical School, Providence, RI 02903, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Cantuti-Castelvetri I, Hernandez LF, Keller-McGandy CE, Kett LR, Landy A, Hollingsworth ZR, Saka E, Crittenden JR, Nillni EA, Young AB, Standaert DG, Graybiel AM. Levodopa-induced dyskinesia is associated with increased thyrotropin releasing hormone in the dorsal striatum of hemi-parkinsonian rats. PLoS One 2010; 5:e13861. [PMID: 21085660 PMCID: PMC2978093 DOI: 10.1371/journal.pone.0013861] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 10/07/2010] [Indexed: 11/25/2022] Open
Abstract
Background Dyskinesias associated with involuntary movements and painful muscle contractions are a common and severe complication of standard levodopa (L-DOPA, L-3,4-dihydroxyphenylalanine) therapy for Parkinson's disease. Pathologic neuroplasticity leading to hyper-responsive dopamine receptor signaling in the sensorimotor striatum is thought to underlie this currently untreatable condition. Methodology/Principal Findings Quantitative real-time polymerase chain reaction (PCR) was employed to evaluate the molecular changes associated with L-DOPA-induced dyskinesias in Parkinson's disease. With this technique, we determined that thyrotropin releasing hormone (TRH) was greatly increased in the dopamine-depleted striatum of hemi-parkinsonian rats that developed abnormal movements in response to L-DOPA therapy, relative to the levels measured in the contralateral non-dopamine-depleted striatum, and in the striatum of non-dyskinetic control rats. ProTRH immunostaining suggested that TRH peptide levels were almost absent in the dopamine-depleted striatum of control rats that did not develop dyskinesias, but in the dyskinetic rats, proTRH immunostaining was dramatically up-regulated in the striatum, particularly in the sensorimotor striatum. This up-regulation of TRH peptide affected striatal medium spiny neurons of both the direct and indirect pathways, as well as neurons in striosomes. Conclusions/Significance TRH is not known to be a key striatal neuromodulator, but intrastriatal injection of TRH in experimental animals can induce abnormal movements, apparently through increasing dopamine release. Our finding of a dramatic and selective up-regulation of TRH expression in the sensorimotor striatum of dyskinetic rat models suggests a TRH-mediated regulatory mechanism that may underlie the pathologic neuroplasticity driving dopamine hyper-responsivity in Parkinson's disease.
Collapse
Affiliation(s)
- Ippolita Cantuti-Castelvetri
- Neurology Department, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| | - Ledia F. Hernandez
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Christine E. Keller-McGandy
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Lauren R. Kett
- Neurology Department, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Alex Landy
- Neurology Department, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Zane R. Hollingsworth
- Neurology Department, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Esen Saka
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Jill R. Crittenden
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Eduardo A. Nillni
- Division of Endocrinology, Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Anne B. Young
- Neurology Department, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - David G. Standaert
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ann M. Graybiel
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| |
Collapse
|
27
|
Regulation of the hypothalamic thyrotropin releasing hormone (TRH) neuron by neuronal and peripheral inputs. Front Neuroendocrinol 2010; 31:134-56. [PMID: 20074584 PMCID: PMC2849853 DOI: 10.1016/j.yfrne.2010.01.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 12/29/2009] [Accepted: 01/06/2010] [Indexed: 12/29/2022]
Abstract
The hypothalamic-pituitary-thyroid (HPT) axis plays a critical role in mediating changes in metabolism and thermogenesis. Thus, the central regulation of the thyroid axis by Thyrotropin Releasing Hormone (TRH) neurons in the paraventricular nucleus of the hypothalamus (PVN) is of key importance for the normal function of the axis under different physiological conditions including cold stress and changes in nutritional status. Before the TRH peptide becomes biologically active, a series of tightly regulated processes occur including the proper folding of the prohormone for targeting to the secretory pathway, its post-translational processing, and targeting of the processed peptides to the secretory granules near the plasma membrane of the cell ready for secretion. Multiple inputs coming from the periphery or from neurons present in different areas of the brain including the hypothalamus are responsible for the activation or inhibition of the TRH neuron and in turn affect the output of TRH and the set point of the axis.
Collapse
|
28
|
Wittmann G, Füzesi T, Liposits Z, Lechan RM, Fekete C. Distribution and axonal projections of neurons coexpressing thyrotropin-releasing hormone and urocortin 3 in the rat brain. J Comp Neurol 2010; 517:825-40. [PMID: 19844978 DOI: 10.1002/cne.22180] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Thyrotropin-releasing hormone (TRH) decreases food intake when administered intracerebroventricularly or into the ventromedial hypothalamus. However, it is unknown which population of TRH neurons exerts this anorexigenic function. In the rostral perifornical area, the pattern of TRH-expressing neurons is reminiscent of the distribution of neurons expressing urocortin3 (Ucn3) that also inhibits feeding when injected into the hypothalamic ventromedial nucleus (VMN). Since colocalization of TRH and Ucn3 may help to identify feeding-related TRH neurons, the putative coexpression of the two peptides was examined using fluorescent in situ hybridization combined with immunofluorescence. Almost all (95.5 +/- 0.2%) Ucn3-immunoreactive neurons in the perifornical area expressed pro-TRH mRNA, while 50.2 +/- 1.6% Ucn3 neurons were double-labeled in the bed nucleus of the stria terminalis (BNST). Only a few Ucn3/pro-TRH neurons were found outside these two areas. The distribution of axons containing both Ucn3 and TRH was examined by dual immunofluorescence. Ucn3/TRH fibers heavily innervated the VMN. In addition, high densities of double-labeled axons were observed in the lateral septal nucleus, posterior division of the BNST, medial amygdaloid nucleus, amygdalohippocampal area, and ventral hippocampus, forebrain areas associated with psychological stress and anxiety. We conclude that Ucn3 and TRH are coexpressed in a discrete, continuous population of neurons in the perifornical area and BNST, making Ucn3 a neurochemical marker to define a distinct subset of TRH neurons. The distribution of their axons suggests that Ucn3/TRH neurons may coordinate feeding and behavioral responses to stressful stimuli.
Collapse
Affiliation(s)
- Gábor Wittmann
- Department of Endocrine Neurobiology, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | |
Collapse
|
29
|
Hypothalamic Sirt1 regulates food intake in a rodent model system. PLoS One 2009; 4:e8322. [PMID: 20020036 PMCID: PMC2790615 DOI: 10.1371/journal.pone.0008322] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 11/20/2009] [Indexed: 01/11/2023] Open
Abstract
Sirt1 is an evolutionarily conserved NAD(+) dependent deacetylase involved in a wide range of processes including cellular differentiation, apoptosis, as well as metabolism, and aging. In this study, we investigated the role of hypothalamic Sirt1 in energy balance. Pharmacological inhibition or siRNA mediated knock down of hypothalamic Sirt1 showed to decrease food intake and body weight gain. Central administration of a specific melanocortin antagonist, SHU9119, reversed the anorectic effect of hypothalamic Sirt1 inhibition, suggesting that Sirt1 regulates food intake through the central melanocortin signaling. We also showed that fasting increases hypothalamic Sirt1 expression and decreases FoxO1 (Forkhead transcription factor) acetylation suggesting that Sirt1 regulates the central melanocortin system in a FoxO1 dependent manner. In addition, hypothalamic Sirt1 showed to regulate S6K signaling such that inhibition of the fasting induced Sirt1 activity results in up-regulation of the S6K pathway. Thus, this is the first study providing a novel role for the hypothalamic Sirt1 in the regulation of food intake and body weight. Given the role of Sirt1 in several peripheral tissues and hypothalamus, potential therapies centered on Sirt1 regulation might provide promising therapies in the treatment of metabolic diseases including obesity.
Collapse
|
30
|
Ishibashi H, Nakahata Y, Eto K, Nabekura J. Excitation of locus coeruleus noradrenergic neurons by thyrotropin-releasing hormone. J Physiol 2009; 587:5709-22. [PMID: 19840999 DOI: 10.1113/jphysiol.2009.181420] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Locus coeruleus (LC) noradrenergic neurons are implicated in a variety of functions including the regulation of vigilance and the modulation of sensory processing. Thyrotropin-releasing hormone (TRH) is an endogenous neuropeptide that induces a variety of behavioural changes including arousal and antinociception. In the present study, we explored whether the activity of LC noradrenergic neurons is modulated by TRH. Using current-clamp recording from isolated rat LC neurons, we found that TRH increased the firing rate of spontaneous action potentials. The TRH action was mimicked by TRH analogues including taltirelin and TRH-gly. In voltage-clamp recording at a holding potential of 50 mV, TRH produced an inward current associated with a decrease in the membrane K+ conductance. This current was inhibited by the TRH receptor antagonist chlordiazepoxide. Following inhibition of the pH-sensitive K+ conductance by extracellular acidification, the TRH response was fully inhibited. The TRH-induced current was also inhibited by the phospholipase C (PLC) inhibitor U-73122, but not by the protein kinase C inhibitor chelerythrine nor by chelation of intracellular Ca2+ by BAPTA. The recovery from the facilitatory action of TRH on the spike frequency was markedly inhibited by a high concentration of wortmannin. These results suggest that TRH activates LC noradrenergic neurons by decreasing an acid-sensitive K+ conductance via PLC-mediated hydrolysis of phosphatidylinositol 4,5-bisphosphate. The present findings demonstrate that TRH activates LC neurons and characterize the underlying signalling mechanisms. The action of TRH on LC neurons may influence a variety of CNS functions related to the noradrenergic system which include arousal and analgesia.
Collapse
Affiliation(s)
- Hitoshi Ishibashi
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan.
| | | | | | | |
Collapse
|
31
|
Wittmann G, Füzesi T, Singru PS, Liposits Z, Lechan RM, Fekete C. Efferent projections of thyrotropin-releasing hormone-synthesizing neurons residing in the anterior parvocellular subdivision of the hypothalamic paraventricular nucleus. J Comp Neurol 2009; 515:313-30. [PMID: 19425088 DOI: 10.1002/cne.22017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The anterior parvocellular subdivision of the PVN (aPVN) contains nonhypophysiotropic thyrotropin-releasing hormone (TRH) neurons that are densely innervated by feeding-related neuronal groups of the hypothalamic arcuate nucleus. To determine how these TRH neurons are integrated within the brain, the major projection fields of this cell group were studied by anterograde and retrograde tract-tracing methods. Projection sites were identified by injection of the anterograde tracer Phaseolus vulgaris leucoagglutinin (PHAL) into the aPVN, and subsequent double immunofluorescent staining was used to visualize axons containing both PHAL and pro-TRH. To distinguish between the projection sites of TRH neurons residing in the aPVN and the closely situated perifornical area, the retrograde tracer cholera toxin B subunit (CTB) was injected into regions where PHAL/pro-TRH-containing axons were densely accumulated. TRH neurons in the aPVN were found to project to the hypothalamic arcuate, dorsomedial and ventral premammillary nuclei, medial preoptic region, tuber cinereum area, paraventricular thalamic nucleus, bed nucleus of the stria terminalis, lateral septal nucleus, and central amygdaloid nucleus. Projection fields of perifornical TRH neurons were in partial overlap with those of the aPVN TRH cells. In addition, these neurons also innervated the hypothalamic ventromedial nucleus, the medial amygdaloid nucleus, and the amygdalohippocampal area. The data suggest that, through its efferent connections, aPVN TRH neurons may be involved in the regulation of energy homeostasis coordinately with effects on behavior, locomotor activity, and thermogenesis. In addition, the major differences in the projection fields of aPVN and perifornical TRH neurons suggest that these two TRH-synthesizing neuronal groups are functionally different.
Collapse
Affiliation(s)
- Gábor Wittmann
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary 1083
| | | | | | | | | | | |
Collapse
|
32
|
Myöhänen TT, García-Horsman JA, Tenorio-Laranga J, Männistö PT. Issues about the physiological functions of prolyl oligopeptidase based on its discordant spatial association with substrates and inconsistencies among mRNA, protein levels, and enzymatic activity. J Histochem Cytochem 2009; 57:831-48. [PMID: 19687473 DOI: 10.1369/jhc.2009.953711] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Prolyl oligopeptidase (POP) is a serine endopeptidase that hydrolyses proline-containing peptides shorter than 30 amino acids. POP may be associated with cognitive functions, possibly via the cleavage of neuropeptides. Recent studies have also suggested novel non-hydrolytic and non-catalytic functions for POP. Moreover, POP has also been proposed as a regulator of inositol 1,4,5-triphosphate signaling and several other functions such as cell proliferation and differentiation, as well as signal transduction in the central nervous system, and it is suspected to be involved in pathological conditions such as Parkinson's and Alzheimer's diseases and cancer. POP inhibitors have been developed to restore the depleted neuropeptide levels encountered in aging or in neurodegenerative disorders. These compounds have shown some antiamnesic effects in animal models. However, the mechanisms of these hypothesized actions are still far from clear. Moreover, the physiological role of POP has remained unknown, and a lack of basic studies, including its distribution, is obvious. The aim of this review is to gather information about POP and to propose some novel roles for this enzyme based on its distribution and its discordant spatial association with its best known substrates.
Collapse
Affiliation(s)
- Timo T Myöhänen
- Department of Pharmacology and Toxicology, University of Kuopio, PO Box 1627, FI-70211 Kuopio, Finland.
| | | | | | | |
Collapse
|
33
|
Parmentier R, Kolbaev S, Klyuch BP, Vandael D, Lin JS, Selbach O, Haas HL, Sergeeva OA. Excitation of histaminergic tuberomamillary neurons by thyrotropin-releasing hormone. J Neurosci 2009; 29:4471-83. [PMID: 19357273 PMCID: PMC3198719 DOI: 10.1523/jneurosci.2976-08.2009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 03/04/2009] [Accepted: 03/05/2009] [Indexed: 11/21/2022] Open
Abstract
The histaminergic tuberomamillary nucleus (TMN) controls arousal and attention, and the firing of TMN neurons is state-dependent, active during waking, silent during sleep. Thyrotropin-releasing hormone (TRH) promotes arousal and combats sleepiness associated with narcolepsy. Single-cell reverse-transcription-PCR demonstrated variable expression of the two known TRH receptors in the majority of TMN neurons. TRH increased the firing rate of most (ca 70%) TMN neurons. This excitation was abolished by the TRH receptor antagonist chlordiazepoxide (CDZ; 50 mum). In the presence of tetrodotoxin (TTX), TRH depolarized TMN neurons without obvious change of their input resistance. This effect reversed at the potential typical for nonselective cation channels. The potassium channel blockers barium and cesium did not influence the TRH-induced depolarization. TRH effects were antagonized by inhibitors of the Na(+)/Ca(2+) exchanger, KB-R7943 and benzamil. The frequency of GABAergic spontaneous IPSCs was either increased (TTX-insensitive) or decreased [TTX-sensitive spontaneous IPSCs (sIPSCs)] by TRH, indicating a heterogeneous modulation of GABAergic inputs by TRH. Facilitation but not depression of sIPSC frequency by TRH was missing in the presence of the kappa-opioid receptor antagonist nor-binaltorphimine. Montirelin (TRH analog, 1 mg/kg, i.p.) induced waking in wild-type mice but not in histidine decarboxylase knock-out mice lacking histamine. Inhibition of histamine synthesis by (S)-alpha-fluoromethylhistidine blocked the arousal effect of montirelin in wild-type mice. We conclude that direct receptor-mediated excitation of rodent TMN neurons by TRH demands activation of nonselective cation channels as well as electrogenic Na(+)/Ca(2+) exchange. Our findings indicate a key role of the brain histamine system in TRH-induced arousal.
Collapse
Affiliation(s)
- Regis Parmentier
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
- Inserm, U628, Physiologie Intégrée du Système d'Éveil, 69373 Lyon Cedex 08, France
| | - Sergej Kolbaev
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
| | - Boris P. Klyuch
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
| | - David Vandael
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
| | - Jian-Sheng Lin
- Inserm, U628, Physiologie Intégrée du Système d'Éveil, 69373 Lyon Cedex 08, France
| | - Oliver Selbach
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
| | - Helmut L. Haas
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
| | - Olga A. Sergeeva
- Department of Neurophysiology, Heinrich Heine University, D-40001 Duesseldorf, Germany, and
| |
Collapse
|
34
|
Kubek MJ, Domb AJ, Veronesi MC. Attenuation of kindled seizures by intranasal delivery of neuropeptide-loaded nanoparticles. Neurotherapeutics 2009; 6:359-71. [PMID: 19332331 PMCID: PMC5084215 DOI: 10.1016/j.nurt.2009.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Accepted: 02/03/2009] [Indexed: 10/21/2022] Open
Abstract
Thyrotropin-releasing hormone (TRH; Protirelin), an endogenous neuropeptide, is known to have anticonvulsant effects in animal seizure models and certain intractable epileptic patients. Its duration of action, however, is limited by rapid tissue metabolism and the blood-brain barrier. Direct nose-to-brain delivery of neuropeptides in sustained-release biodegradable nanoparticles (NPs) is a promising mode of therapy for enhancing CNS neuropeptide bioavailability. To provide proof of principle for this delivery approach, we used the kindling model of temporal lobe epilepsy to show that 1) TRH-loaded copolymer microdisks implanted in a seizure focus can attenuate kindling development in terms of behavioral stage, afterdischarge duration (ADD), and clonus duration; 2) intranasal administration of an unprotected TRH analog can acutely suppress fully kindled seizures in a concentration-dependent manner in terms of ADD and seizure stage; and 3) intranasal administration of polylactide nanoparticles (PLA-NPs) containing TRH (TRH-NPs) can impede kindling development in terms of behavioral stage, ADD, and clonus duration. Additionally, we used intranasal delivery of fluorescent dye-loaded PLA-NPs in rats and application of dye-loaded or dye-attached NPs to cortical neurons in culture to demonstrate NP uptake and distribution over time in vivo and in vitro respectively. Also, a nanoparticle immunostaining method was developed as a procedure for directly visualizing the tissue level and distribution of neuropeptide-loaded nanoparticles. Collectively, the data provide proof of concept for intranasal delivery of TRH-NPs as a viable means to 1) suppress seizures and perhaps epileptogenesis and 2) become the lead compound for intranasal anticonvulsant nanoparticle therapeutics.
Collapse
Affiliation(s)
- Michael J Kubek
- Department of Anatomy, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | |
Collapse
|
35
|
Prokai-Tatrai K, Prokai L. Prodrugs of thyrotropin-releasing hormone and related peptides as central nervous system agents. Molecules 2009; 14:633-54. [PMID: 19214153 PMCID: PMC6253886 DOI: 10.3390/molecules14020633] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2009] [Revised: 01/21/2009] [Accepted: 02/05/2009] [Indexed: 02/03/2023] Open
Abstract
Prodrug design for brain delivery of small- and medium-sized neuropeptides was reviewed, focusing on thyrotropin-releasing hormone and structurally related peptides as examples. We have summarized our most important advances in methodology, as well as assessed the benefits and limitations of bioreversible chemical manipulation techniques to achieve targeting of the parent molecules into the central nervous system. The value of prodrug-amenable analogues as potential drug-like central nervous systems agents was highlighted.
Collapse
Affiliation(s)
- Katalin Prokai-Tatrai
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Laszlo Prokai
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, USA; E-mail: (L.P.)
| |
Collapse
|
36
|
Romero A, Cakir I, Vaslet CA, Stuart RC, Lansari O, Lucero HA, Nillni EA. Role of a pro-sequence in the secretory pathway of prothyrotropin-releasing hormone. J Biol Chem 2008; 283:31438-48. [PMID: 18779326 DOI: 10.1074/jbc.m803413200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The biogenesis of rat thyrotropin releasing hormone (TRH) involves the processing of its precursor (proTRH) into five biologically active TRH peptides and several non-TRH peptides where two of them had been attributed potential biological functions. This process implicates 1) proper folding of proTRH in the endoplasmic reticulum after its biosynthesis and exit to the Golgi apparatus and beyond, 2) initial processing of proTRH in the trans Golgi network and, 3) sorting of proTRH-derived peptides to the regulated secretory pathway. Previous studies have focused on elucidating the processing and sorting determinants of proTRH. However, the role of protein folding in the sorting of proTRH remains unexplored. Here we have investigated the role in the secretion of proTRH of a sequence comprising 22 amino acid residues, located at the N-terminal region of proTRH, residues 31-52. Complete deletion of these 22 amino acids dramatically compromised the biosynthesis of proTRH, manifested as a severe reduction in the steady state level of proTRH in the endoplasmic reticulum. This effect was largely reproduced by the deletion of only three amino acid residues, 40PGL42, within the proTRH31-52 sequence. The decreased steady state level of the mutant DeltaPGL was due to enhanced endoplasmic reticulum-associated protein degradation. However, the remnant of DeltaPGL that escaped degradation was properly processed and sorted to secretory granules. Thus, these results suggest that the N-terminal domain within the prohormone sequence does not act as "sorting signal" in late secretion; instead, it seems to play a key role determining the proper folding pathway of the precursor and, thus, its stability.
Collapse
Affiliation(s)
- Amparo Romero
- Department of Medicine, Division of Endocrinology, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island 02903, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Espinosa VP, Liu Y, Ferrini M, Anghel A, Nie Y, Tripathi PV, Porche R, Jansen E, Stuart RC, Nillni EA, Lutfy K, Friedman TC. Differential regulation of prohormone convertase 1/3, prohormone convertase 2 and phosphorylated cyclic-AMP-response element binding protein by short-term and long-term morphine treatment: implications for understanding the "switch" to opiate addiction. Neuroscience 2008; 156:788-99. [PMID: 18771713 DOI: 10.1016/j.neuroscience.2008.07.063] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/21/2008] [Accepted: 07/22/2008] [Indexed: 10/21/2022]
Abstract
Drug addiction is a state of altered brain reward and self-regulation mediated by both neurotransmitter and hormonal systems. Although an organism's internal system attempts to maintain homeostasis when challenged by exogenous opiates and other drugs of abuse, it eventually fails, resulting in the transition from drug use to drug abuse. We propose that the attempted maintenance of hormonal homeostasis is achieved, in part, through alterations in levels of processing enzymes that control the ratio of active hormone to pro-hormone. Two pro-hormone convertases, PC1/3 and PC2 are believed to be responsible for the activation of many neurohormones and expression of these enzymes is dependent on the presence of a cyclic-AMP response element (CRE) in their promoters. Therefore, we studied the effects of short-term (24-h) and long-term (7-day) morphine treatment on the expression of hypothalamic PC1/3 and PC2 and levels of phosphorylated cyclic-AMP-response element binding protein (P-CREB). While short-term morphine exposure down-regulated, long-term morphine exposure up-regulated P-CREB, PC1/3 and PC2 protein levels in the rat hypothalamus as determined by Western blot analysis. Quantitative immunofluorescence studies confirmed these regulatory actions of morphine in the paraventricular and dorsomedial nucleus of the hypothalamus. Specific radioimmunoassays demonstrated that the increase in PC1/3 and PC2 levels following long-term morphine led to increased TRH biosynthesis as evidence by increased TRH/5.4 kDa C-terminal proTRH-derived peptide ratios in the median eminence. Promoter activity experiments in rat somatomammotrope GH3 cells containing the mu-opioid receptor demonstrated that the CRE(s) in the promoter of PC1/3 and PC2 is required for morphine-induced regulation of PC1/3 and PC2. Our data suggest that the regulation of the prohormone processing system by morphine may lead to alterations in the levels of multiple bioactive hormones and may be a compensatory mechanism whereby the organism tries to restore its homeostatic hormonal milieu. The down-regulation of PC1/3, PC2 and P-CREB by short-term morphine and up-regulation by long-term morphine treatment may be a signal mediating the switch from drug use to drug abuse.
Collapse
Affiliation(s)
- V Paez Espinosa
- Division of Endocrinology, Department of Medicine, The Charles Drew University of Medicine & Sciences-UCLA School of Medicine, 1731 East 120th Street, Los Angeles, CA 90059, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Perello M, Stuart R, Nillni EA. Prothyrotropin-releasing hormone targets its processing products to different vesicles of the secretory pathway. J Biol Chem 2008; 283:19936-47. [PMID: 18474603 DOI: 10.1074/jbc.m800732200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prothyrotropin-releasing hormone (pro-TRH) is initially cleaved by the prohormone convertase-1/3 (PC1/3) in the trans-Golgi network generating N- and C-terminal intermediate forms that are then packed into secretory vesicles. However, it is not known whether these peptides are differentially sorted within the secretory pathway. This is of key importance because the processing products of several prohormones fulfill different biological functions. Using AtT20 cells stably transfected with prepro-TRH cDNA, we found that two specific N- and C-terminal peptides were located in different vesicles. Furthermore, the C-terminal pro-TRH-derived peptides were more efficiently released in response to KCl and norepinephrine, a natural secretagogue of TRH. Similar sorting and secretion of N- and C-terminal peptides occurs in vivo. When we blocked the initial proteolytic processing by a mutagenic approach, the differential sorting and secretion of these peptides were prevented. In summary, our data show that pro-TRH-derived peptides are differentially sorted within the secretory pathway and that the initial cleavage in the trans-Golgi network is key to this process. This could be a common mechanism used by neuroendocrine cells to regulate independently the secretion of different bioactive peptides derived from the same gene product.
Collapse
Affiliation(s)
- Mario Perello
- Division of Endocrinology, Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | | | | |
Collapse
|
39
|
Gotoh K, Fukagawa K, Fukagawa T, Noguchi H, Kakuma T, Sakata T, Yoshimatsu H. Hypothalamic neuronal histamine mediates the thyrotropin-releasing hormone-induced suppression of food intake. J Neurochem 2007; 103:1102-10. [PMID: 17760865 PMCID: PMC2156111 DOI: 10.1111/j.1471-4159.2007.04802.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 03/14/2007] [Accepted: 06/21/2007] [Indexed: 11/28/2022]
Abstract
We examined the involvement of thyrotropin-releasing hormone (TRH) and TRH type 1 and 2 receptors (TRH-R1 and TRH-R2, respectively) in the regulation of hypothalamic neuronal histamine. Infusion of 100 nmol TRH into the rat third cerebroventricle (3vt) significantly decreased food intake (p < 0.05) compared to controls infused with phosphate- buffered saline. This TRH-induced suppression of food intake was attenuated partially in histamine-depleted rats pre-treated with alpha-fluoromethylhistidine (a specific suicide inhibitor of histidine decarboxylase) and in mice with targeted disruption of histamine H1 receptors. Infusion of TRH into the 3vt increased histamine turnover as assessed by pargyline-induced accumulation of tele-methylhistamine (t-MH, a major metabolite of neuronal histamine in the brain) in the tuberomammillary nucleus (TMN), the paraventricular nucleus, and the ventromedial hypothalamic nucleus in rats. In addition, TRH-induced decrease of food intake and increase of histamine turnover were in a dose-dependent manner. Microinfusion of TRH into the TMN increased t-MH content, histidine decarboxylase (HDC) activity and expression of HDC mRNA in the TMN. Immunohistochemical analysis revealed that TRH-R2, but not TRH-R1, was expressed within the cell bodies of histaminergic neurons in the TMN of rats. These results indicate that hypothalamic neuronal histamine mediates the TRH-induced suppression of feeding behavior.
Collapse
MESH Headings
- Animals
- Appetite Regulation/drug effects
- Appetite Regulation/physiology
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Histamine/metabolism
- Histamine H1 Antagonists/pharmacology
- Histidine Decarboxylase/antagonists & inhibitors
- Histidine Decarboxylase/genetics
- Histidine Decarboxylase/metabolism
- Hypothalamus/anatomy & histology
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Immunohistochemistry
- Injections, Intraventricular
- Male
- Methylhistamines/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons/drug effects
- Neurons/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Histamine H1/drug effects
- Receptors, Histamine H1/genetics
- Receptors, Histamine H1/metabolism
- Receptors, Thyrotropin-Releasing Hormone/agonists
- Receptors, Thyrotropin-Releasing Hormone/metabolism
- Thyrotropin-Releasing Hormone/metabolism
- Thyrotropin-Releasing Hormone/pharmacology
Collapse
Affiliation(s)
- Koro Gotoh
- Department of Internal Medicine 1, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Fekete C, Lechan RM. Negative feedback regulation of hypophysiotropic thyrotropin-releasing hormone (TRH) synthesizing neurons: role of neuronal afferents and type 2 deiodinase. Front Neuroendocrinol 2007; 28:97-114. [PMID: 17588648 PMCID: PMC2000455 DOI: 10.1016/j.yfrne.2007.04.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 04/01/2007] [Accepted: 04/23/2007] [Indexed: 11/26/2022]
Abstract
Hypophysiotropic thyrotropin-releasing hormone (TRH): synthesizing neurons reside in the hypothalamic paraventricular nucleus (PVN) and are the central regulators of the hypothalamic-pituitary-thyroid (HPT) axis. TRH synthesis and release from these neurons are primarily under negative feedback regulation by thyroid hormone. Under certain conditions such as cold exposure and fasting, however, inputs from neurons in the brainstem and hypothalamic arcuate and dorsomedial nuclei alter the set point for negative feedback through regulation of CREB phosphorylation. Thus, during cold exposure, adrenergic neurons stimulate the HPT axis, while fasting-induced central hypothyroidism is mediated through an arcuato-paraventricular pathway. Feedback regulation of TRH neurons may also be modified by local tissue levels of thyroid hormone regulated by the activation of type 2 iodothyronine deiodinase (D2), the primary enzyme in the brain that catalyzes T4 to T3 conversion. During infection, endotoxin or endotoxin induced cytokines increase D2 activity in the mediobasal hypothalamus, which by inducing local hyperthyroidism, may play an important role in infection-induced inhibition of hypophysiotropic TRH neurons.
Collapse
Affiliation(s)
- Csaba Fekete
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest 1083, Hungary.
| | | |
Collapse
|
41
|
Goldstein J, Perello M, Nillni EA. PreproThyrotropin-releasing hormone 178-199 affects tyrosine hydroxylase biosynthesis in hypothalamic neurons: a possible role for pituitary prolactin regulation. J Mol Neurosci 2007; 31:69-82. [PMID: 17416971 DOI: 10.1007/bf02686119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 08/04/2006] [Accepted: 08/04/2006] [Indexed: 02/02/2023]
Abstract
ProThyrotropin-releasing hormone (proTRH) is a prohormone widely distributed in many areas of the brain. After biosynthesis, proTRH is subjected to post-translational processing to generate TRH and seven non-TRH peptides. Among these non-TRH sequences, we found previously that preproTRH178-199 could regulate the secretion of prolactin in suckled rats by their pups. Dopamine (DA), the main regulator of prolactin secretion, is produced in dopaminergic tyrosine hydroxylase (TH)-positive neurons in the hypothalamic arcuate nucleus (ARC). In this study we investigated whether prolactin release during the estrous sexual cycle is regulated by preproTRH178-199 through its effect on DA neurons of the ARC. We observed that biotinylated preproTRH178-199 bound to neurons in the ARC; this was higher during proestrus than during diestrus. Binding of preproTRH178-199 to DA neurons was seen only during proestrus in the ARC. Using primary neuronal hypothalamic cultures we found that preproTRH178-199 peptide decreased TH levels in a dose-responsive manner, whereas intra-ARC administration of preproTRH178-199 induced a 20-fold increase in plasma prolactin levels. Together, these results suggest a potential role for preproTRH178-199 in regulating dopaminergic neurons involved in the inhibition of pituitary prolactin release.
Collapse
Affiliation(s)
- Jorge Goldstein
- Division of Endocrinology, Department of Medicine, Brown Medical School, Rhode Island Hospital, Providence, RI 02903, USA
| | | | | |
Collapse
|
42
|
Perello M, Stuart RC, Nillni EA. Differential effects of fasting and leptin on proopiomelanocortin peptides in the arcuate nucleus and in the nucleus of the solitary tract. Am J Physiol Endocrinol Metab 2007; 292:E1348-57. [PMID: 17227963 DOI: 10.1152/ajpendo.00466.2006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The alpha-melanocyte-stimulating hormone (alpha-MSH), derived from proopiomelanocortin (POMC), is generated by a posttranslational processing mechanism involving the prohormone convertases (PCs) PC1/3 and PC2. In the brain, alpha-MSH is produced in the arcuate nucleus (ARC) of the hypothalamus and in the nucleus of the solitary tract (NTS) of the medulla. This peptide is key in controlling energy balance, as judged by changes observed at transcriptional level. However, little information is available regarding the biosynthesis of the precursor POMC and the production of its processed peptides during feeding, fasting, and fasting plus leptin in the ARC compared with the NTS in conjunction with the PC activity. In this study we found that, in the ARC, pomc mRNA, POMC-derived peptides, and PC1/3 all decreased during fasting, and administration of leptin reversed these effects. In contrast, in the NTS, where there is a large amount of a 28.1-kDa peptide similar in size to POMC, the 28.1-kDa peptide and other POMC-derived peptides, including alpha-MSH, were further accumulated in fasting conditions, whereas pomc mRNA decreased. These changes were not reversed by leptin. We also observed that, during fasting, PC2 levels decreased in the NTS. These data suggest that, in the NTS, fasting induced changes in POMC biosynthesis, and processing is independent of leptin. These observations indicate that changes in energy status affect POMC in the brain in a tissue-specific manner. This represents a novel aspect in the regulation of energy balance and may have implications in the pathophysiology of obesity.
Collapse
Affiliation(s)
- Mario Perello
- Division of Endocrinology, Department of Medicine, Brown University, Rhode Island Hospital, Providence, Rhode Island 02903, USA
| | | | | |
Collapse
|
43
|
Teixidó M, Prokai-Tatrai K, Wang X, Nguyen V, Prokai L. Exploratory neuropharmacological evaluation of a conformationally constrained thyrotropin-releasing hormone analogue. Brain Res Bull 2007; 73:103-7. [PMID: 17499643 PMCID: PMC1950732 DOI: 10.1016/j.brainresbull.2007.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 02/19/2007] [Indexed: 11/29/2022]
Abstract
A conformationally constrained peptidomimetic derived from the endocrine and neuroactive tripeptide thyrotropin-releasing hormone (pGlu-His-Pro-NH(2)) was synthesized by convenient solid-phase organic chemistry and evaluated as a potential central nervous system agent. While this ethylene-bridged peptide analogue has been reported to lack the hormonal effect of the native peptide, we have shown in animal models that it possesses central nervous system activity characteristic of thyrotropin-releasing hormone. Compared to control, the peptidomimetic showed significant analeptic and antidepressant-like potencies. Moreover, an enhanced selectivity in antidepressant-like effect was measured when compared to that of the native peptide. Immobilized artificial membrane chromatography and in vitro metabolic stability studies also revealed that this constrained peptidomimetic has higher affinity to the blood-brain barrier than the native peptide and is metabolically stable. Consequently, this structure may be used as a template to design centrally selective and metabolically stable thyrotropin-releasing hormone analogues as potential neuropharmaceutical agents.
Collapse
Affiliation(s)
- Meritxell Teixidó
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Katalin Prokai-Tatrai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
- Corresponding author. Tel.: + 01 817 735 0617; fax: +01 817 735 2651. E-mail: (K. Prokai-Tatrai)
| | - Xiaoli Wang
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Vien Nguyen
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Laszlo Prokai
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| |
Collapse
|
44
|
Veronesi MC, Yard M, Jackson J, Lahiri DK, Kubek MJ. An analog of thyrotropin-releasing hormone (TRH) is neuroprotective against glutamate-induced toxicity in fetal rat hippocampal neurons in vitro. Brain Res 2007; 1128:79-85. [PMID: 17125753 PMCID: PMC2645863 DOI: 10.1016/j.brainres.2006.10.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 10/23/2006] [Accepted: 10/24/2006] [Indexed: 12/24/2022]
Abstract
TRH has been found to be efficacious in treating certain neurodegenerative disorders such as epilepsy, Alzheimer's disease, neurotrauma and depression, however, its mechanism of action is poorly understood. Since glutamate (Glu) toxicity has been implicated in these disorders, we utilized primary enriched cultures of rat fetal (E 17) hippocampal neurons to test the hypothesis that an analog of TRH, 3-Methyl-Histidine TRH (3Me-H TRH), given concurrently with Glu would protect such neurons against cell damage and cell death. Cell viability was assessed via Trypan Blue exclusion cell counts, and neuronal damage was determined by assaying lactic acid dehydrogenase (LDH) released in the conditioned media. Fetal hippocampal neurons were cultured in neurobasal media for 7 days. On day 7, neurons (10(6)/well) were treated with: control media, 10 microM 3Me-H TRH, 500 microM Glu or 500 microM Glu with either 10, 1, 0.1, 0.01 or 0.001 microM 3Me-H TRH. Both media and neurons were harvested 16 h after treatment. Prolonged exposure to 10 microM 3Me-H TRH was not toxic to the cells, whereas neurons exposed to 500 microM Glu resulted in maximal cell death. Notably, 10, 1 and 0.1 microM 3Me-H TRH, when co-treated with 500 microM Glu, protected fetal neurons against cell death in a concentration-dependent manner. These results provide support for an important neuroprotective effect of TRH/analogs against glutamate toxicity in primary hippocampal neuronal culture and implicate a potentially beneficial role of TRH/analogs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Michael C. Veronesi
- Program in Medical Neurobiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Michael Yard
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
- Program in Medical Neurobiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - James Jackson
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Debomoy K. Lahiri
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
- Program in Medical Neurobiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Michael J. Kubek
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Program in Medical Neurobiology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
45
|
Espinosa VP, Ferrini M, Shen X, Lutfy K, Nillni EA, Friedman TC. Cellular colocalization and coregulation between hypothalamic pro-TRH and prohormone convertases in hypothyroidism. Am J Physiol Endocrinol Metab 2007; 292:E175-86. [PMID: 16926379 DOI: 10.1152/ajpendo.00288.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The prohormone convertases (PCs), PC1/3 and PC2, are involved in the tissue-specific endoproteolytic posttranslational processing of many hormonal precursors within the secretory pathway. One important prohormone, pro-thyrotropin-releasing hormone (TRH), is expressed in both hypophysiotropic (where it regulates the secretion of thyroid-stimulating hormone) and nonhypophysiotropic regions of the brain. Pro-TRH is processed at specific sites in the secretory pathway, primarily by PC1/3 followed by PC2. We hypothesized that thyroid hormone status in specific nuclei of the brain would alter pro-TRH processing by inducing changes in PC1/3 and PC2 expression. Therefore, we examined pro-TRH, PC1/3, and PC2 coexpression and coregulation in the paraventricular nucleus (PVN), lateral hypothalamus (LH), and ventromedial nucleus (VMN) of hypothyroid and euthyroid rats. Our results show that 6-n-propyl-2-thiouracil (PTU) treatment producing hypothyroidism induced a significant increase in the expression of PC1/3, PC2, and pro-TRH in the PVN and LH, but not VMN. When confocal studies were performed, an increase in colocalization of PC1/3 or PC2 in pro-TRH was observed only in PVN, a response that was especially prominent in the ventral and medial areas of the PVN. PTU did not regulate colocalization in the VMH or LH. Regulation of colocalization of processing enzyme and prohormone expression is a novel mechanism to alter hormonal biosynthesis.
Collapse
Affiliation(s)
- Veronica Paez Espinosa
- Charles R. Drew Univ. of Medicine & Sciences, Division of Endocrinology, 1731 E. 120th St., Los Angeles, CA 90059, USA
| | | | | | | | | | | |
Collapse
|
46
|
Deng PY, Porter JE, Shin HS, Lei S. Thyrotropin-releasing hormone increases GABA release in rat hippocampus. J Physiol 2006; 577:497-511. [PMID: 16990402 PMCID: PMC1890442 DOI: 10.1113/jphysiol.2006.118141] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Thyrotropin-releasing hormone (TRH) is a tripeptide that is widely distributed in the brain including the hippocampus where TRH receptors are also expressed. TRH has anti-epileptic effects and regulates arousal, sleep, cognition, locomotion and mood. However, the cellular mechanisms underlying such effects remain to be determined. We examined the effects of TRH on GABAergic transmission in the hippocampus and found that TRH increased the frequency of GABAA receptor-mediated spontaneous IPSCs in each region of the hippocampus but had no effects on miniature IPSCs or evoked IPSCs. TRH increased the action potential firing frequency recorded from GABAergic interneurons in CA1 stratum radiatum and induced membrane depolarization suggesting that TRH increases the excitability of interneurons to facilitate GABA release. TRH-induced inward current had a reversal potential close to the K+ reversal potential suggesting that TRH inhibits resting K+ channels. The involved K+ channels were sensitive to Ba2+ but resistant to other classical K+ channel blockers, suggesting that TRH inhibits the two-pore domain K+ channels. Because the effects of TRH were mediated via Galphaq/11, but were independent of its known downstream effectors, a direct coupling may exist between Galphaq/11 and K+ channels. Inhibition of the function of dynamin slowed the desensitization of TRH responses. TRH inhibited seizure activity induced by Mg2+ deprivation, but not that generated by picrotoxin, suggesting that TRH-mediated increase in GABA release contributes to its anti-epileptic effects. Our results demonstrate a novel mechanism to explain some of the hippocampal actions of TRH.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | | | | | | |
Collapse
|
47
|
Matlhagela K, Taub M. Regulation of the Na-K-ATPase beta(1)-subunit promoter by multiple prostaglandin-responsive elements. Am J Physiol Renal Physiol 2006; 291:F635-46. [PMID: 16478973 DOI: 10.1152/ajprenal.00452.2005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal prostaglandins modulate the activity of a number of the transport systems in the kidney, including the Na-K-ATPase. Not only do prostaglandins have acute affects on renal Na-K-ATPase, but in addition prostaglandins have chronic affects, which include regulation at the transcriptional level. Previously, we have presented evidence that one such prostaglandin, PGE(1), stimulates the transcription of the human Na-K-ATPase beta(1)-subunit gene in Madin-Darby canine kidney cells via cAMP- and Ca(2+)-mediated pathways (Taub M, Borsick M, Geisel J, Matlhagela K, Rajkhowa T, and Allen C. Exp Cell Res 299: 1-14, 2004; Matlhagela K, Borsick M, Rajkhowa T, and Taub M. J Biol Chem 280: 334-346, 2005). Evidence was presented indicating that PGE(1) stimulation was mediated through the binding of cAMP-regulatory element binding protein (CREB) to a prostaglandin-responsive element (PGRE) as well as Sp1 binding to an adjacent Sp1 site. In this report, we present evidence from EMSAs and DNA affinity precipitation studies that another PGRE present in the Na-K-ATPase beta(1)-subunit promoter similarly binds CREB and Sp1. The evidence that indicates a requirement for CREB as well as Sp1 for gene activation through both PGREs (PGRE1 and PGRE3) includes studies with a dominant negative CREB (KCREB), Drosophila SL2 cells, and PGRE mutants. The results of these studies are indicative of a synergism between Sp1 and CREB in mediating regulation by PGRE3; while regulation occurring through PGRE1 also involves Sp1 and CREB, the mechanism appears to be distinct.
Collapse
Affiliation(s)
- Keikantse Matlhagela
- Biochemistry Department, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | | |
Collapse
|
48
|
Kelly JA, Scalabrino GA, Slator GR, Cullen AA, Gilmer JF, Lloyd DG, Bennett GW, Bauer K, Tipton KF, Williams CH. Structure-activity studies with high-affinity inhibitors of pyroglutamyl-peptidase II. Biochem J 2005; 389:569-76. [PMID: 15799721 PMCID: PMC1175135 DOI: 10.1042/bj20041722] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Inhibitors of PPII (pyroglutamyl-peptidase II) (EC 3.4.19.6) have potential applications as investigative and therapeutic agents. The rational design of inhibitors is hindered, however, by the lack of an experimental structure for PPII. Previous studies have demonstrated that replacement of histidine in TRH (thyrotropin-releasing hormone) with asparagine produces a competitive PPII inhibitor (Ki 17.5 microM). To gain further insight into which functional groups are significant for inhibitory activity, we investigated the effects on inhibition of structural modifications to Glp-Asn-ProNH2 (pyroglutamyl-asparaginyl-prolineamide). Synthesis and kinetic analysis of a diverse series of carboxamide and C-terminally extended Glp-Asn-ProNH2 analogues were undertaken. Extensive quantitative structure-activity relationships were generated, which indicated that key functionalities in the basic molecular structure of the inhibitors combine in a unique way to cause PPII inhibition. Data from kinetic and molecular modelling studies suggest that hydrogen bonding between the asparagine side chain and PPII may provide a basis for the inhibitory properties of the asparagine-containing peptides. Prolineamide appeared to be important for interaction with the S2' subsite, but some modifications were tolerated. Extension of Glp-Asn-ProNH2 with hydrophobic amino acids at the C-terminus led to a novel set of PPII inhibitors active in vitro at nanomolar concentrations. Such inhibitors were shown to enhance recovery of TRH released from rat brain slices. Glp-Asn-Pro-Tyr-Trp-Trp-7-amido-4-methylcoumarin displayed a Ki of 1 nM, making it the most potent competitive PPII inhibitor described to date. PPII inhibitors with this level of potency should find application in exploring the biological functions of TRH and PPII, and potentially provide a basis for development of novel therapeutics.
Collapse
Affiliation(s)
- Julie A Kelly
- Department of Biochemistry, Trinity College Dublin, Dublin 2, Ireland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sanchez VC, Goldstein J, Stuart RC, Hovanesian V, Huo L, Munzberg H, Friedman TC, Bjorbaek C, Nillni EA. Regulation of hypothalamic prohormone convertases 1 and 2 and effects on processing of prothyrotropin-releasing hormone. J Clin Invest 2004; 114:357-69. [PMID: 15286802 PMCID: PMC484982 DOI: 10.1172/jci21620] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2004] [Accepted: 06/15/2004] [Indexed: 01/19/2023] Open
Abstract
Regulation of energy balance by leptin involves regulation of several neuropeptides, including thyrotropin-releasing hormone (TRH). Synthesized from a larger inactive precursor, its maturation requires proteolytic cleavage by prohormone convertases 1 and 2 (PC1 and PC2). Since this maturation in response to leptin requires prohormone processing, we hypothesized that leptin might regulate hypothalamic PC1 and PC2 expression, ultimately leading to coordinated processing of prohormones into mature peptides. Using hypothalamic neurons, we found that leptin stimulated PC1 and PC2 mRNA and protein expression and also increased PC1 and PC2 promoter activities in transfected 293T cells. Starvation of rats, leading to low serum leptin levels, decreased PC1 and PC2 gene and protein expression in the paraventricular nucleus (PVN) of the hypothalamus. Exogenous administration of leptin to fasted animals restored PC1 levels in the median eminence (ME) and the PVN to approximately the level found in fed control animals. Consistent with this regulation of PCs in the PVN, concentrations of TRH in the PVN and ME were substantially reduced in the fasted animals relative to the fed animals, and leptin reversed this decrease. Further analysis showed that proteolytic cleavage of pro-thyrotropin-releasing hormone (proTRH) at known PC cleavage sites was reduced by fasting and increased in animals given leptin. Combined, these findings suggest that leptin-dependent stimulation of hypothalamic TRH expression involves both activation of trh transcription and stimulation of PC1 and PC2 expression, which lead to enhanced processing of proTRH into mature TRH.
Collapse
Affiliation(s)
- Vanesa C Sanchez
- Division of Endocrinology, Department of Medicine, Brown Medical School, Rhode Island Hospital, Providence, Rhode Island 02903, USA
| | | | | | | | | | | | | | | | | |
Collapse
|