1
|
Sliwowska JH, Woods NE, Alzahrani AR, Paspali E, Tate RJ, Ferro VA. Kisspeptin a potential therapeutic target in treatment of both metabolic and reproductive dysfunction. J Diabetes 2024; 16:e13541. [PMID: 38599822 PMCID: PMC11006622 DOI: 10.1111/1753-0407.13541] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/21/2023] [Accepted: 02/03/2024] [Indexed: 04/12/2024] Open
Abstract
Kisspeptins (KPs) are proteins that were first recognized to have antimetastatic action. Later, the critical role of this peptide in the regulation of reproduction was proved. In recent years, evidence has been accumulated supporting a role for KPs in regulating metabolic processes in a sexual dimorphic manner. It has been proposed that KPs regulate metabolism both indirectly via gonadal hormones and/or directly via the kisspeptin receptor in the brain, brown adipose tissue, and pancreas. The aim of the review is to provide both experimental and clinical evidence indicating that KPs are peptides linking metabolism and reproduction. We propose that KPs could be used as a potential target to treat both metabolic and reproductive abnormalities. Thus, we focus on the consequences of disruptions in KPs and their receptors in metabolic conditions such as diabetes, undernutrition, obesity, and reproductive disorders (hypogonadotropic hypogonadism and polycystic ovary syndrome). Data from both animal models and human subjects indicate that alterations in KPs in the case of metabolic imbalance lead also to disruptions in reproductive functions. Changes both in the hypothalamic and peripheral KP systems in animal models of the aforementioned disorders are discussed. Finally, an overview of current clinical studies involving KP in fertility and metabolism show fewer studies on metabolism (15%) and only one to date on both. Presented data indicate a dynamic and emerging field of KP studies as possible therapeutic targets in treatments of both reproductive and metabolic dysfunctions.
Collapse
Affiliation(s)
- Joanna Helena Sliwowska
- Department of Veterinary Medicine and Animal Sciences, Laboratory of Neurobiology, Poznan University of Life Sciences, Poznan, Poland
| | - Nicola Elizabeth Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Abdullah Rzgallah Alzahrani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Elpiniki Paspali
- Department of Chemical Engineering, University of Strathclyde, Glasgow, UK
| | - Rothwelle Joseph Tate
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Valerie Anne Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
2
|
Hauser F, Al-Ribaty T, Stebegg M, Thygesen G, Grimmelikhuijzen CJP. Cloning and deorphanization of three inotocin (insect oxytocin/vasopressin-like) receptors and their ligand from the tick Ixodes scapularis. Biochem Biophys Res Commun 2023; 680:34-41. [PMID: 37716155 DOI: 10.1016/j.bbrc.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/18/2023]
Abstract
Many insects produce the cyclic neuropeptide inotocin (CLITNCPRGamide), which is the insect orthologue of the mammalian neuropeptides oxytocin and vasopressin. These insects also have one inotocin G protein-coupled receptor (GPCR), which is the orthologue of the mammalian oxytocin and vasopressin receptors. The tick Ixodes scapularis belongs to the subphylum Chelicerata, an arthropod taxon different from insects, to which also spiders, scorpions, and mites belong. I. scapularis is an ectoparasite and a health risk for humans, because it transfers pathogenic microorganisms to its human host during a blood meal, thereby causing serious neurological diseases, among them Lyme disease and tick-borne encephalitis (TBE). By annotating the genomic sequence of I. scapularis, we previously found one presumed tick inotocin preprohormone gene and, in contrast to insects, three genes coding for presumed inotocin GPCRs. We now find that these GPCR genes cluster on one genomic contig, suggesting that they originated by recent gene duplications. Closely located on the same contig are also four adipokinetic hormone/corazonin-related peptide (ACP) GPCR genes, and one crustacean cardioactive peptide (CCAP) GPCR gene, suggesting evolutionary relationships. These evolutionary relationships are confirmed by phylogenetic tree analyses of their gene products. We also cloned the tick inotocin preprohormone, which has a structural organization closely resembling mammalian oxytocin and vasopressin preprohormones, including the presence of a conserved neurophysin sequence, having seven cystine bridges. This neurophysin sequence has two cystine-knot domains, but in contrast to mammalian neurophysins, the tick neurophysin contains a canonical prohormone convertase cleavage signal and a peptide C-terminal amidation sequence (GKR), suggesting cleavage into two biologically active cystine-knot peptides. This cleavage/amidation sequence occurs in neurophysins from most hard tick species, but not in other chelicerates. Mature tick inotocin is different from insect inotocin and has the sequence CFITNCPPGamide. Finally, we cloned and stably expressed the three tick inotocin receptors in Chinese Hamster Ovary cells and found that each of them was activated by nanomolar concentrations of tick inotocin (EC50 for ITR1 = 1.6 × 10-8 M; EC50 for ITR2 = 5.8 × 10-9 M; EC50 for ITR3 = 9.3 × 10-9 M), thereby establishing that they are genuine tick inotocin receptors.
Collapse
Affiliation(s)
- Frank Hauser
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-2100, Copenhagen, Denmark
| | - Tara Al-Ribaty
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-2100, Copenhagen, Denmark
| | - Marisa Stebegg
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-2100, Copenhagen, Denmark
| | - Gedske Thygesen
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-2100, Copenhagen, Denmark
| | - Cornelis J P Grimmelikhuijzen
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
3
|
Silver R, Yao Y, Roy RK, Stern JE. Parallel trajectories in the discovery of the SCN-OVLT and pituitary portal pathways: Legacies of Geoffrey Harris. J Neuroendocrinol 2023; 35:e13245. [PMID: 36880566 PMCID: PMC10423749 DOI: 10.1111/jne.13245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/16/2023]
Abstract
A map of central nervous system organization based on vascular networks provides a layer of organization distinct from familiar neural networks or connectomes. As a well-established example, the capillary networks of the pituitary portal system enable a route for small amounts of neurochemical signals to reach local targets by traveling along specialized pathways, thereby avoiding dilution in the systemic circulation. The first evidence of such a pathway in the brain came from anatomical studies identifying a portal pathway linking the hypothalamus and the pituitary gland. Almost a century later, we demonstrated a vascular portal pathway that joined the capillary beds of the suprachiasmatic nucleus and a circumventricular organ, the organum vasculosum of the lamina terminalis, in a mouse brain. For each of these portal pathways, the anatomical findings opened many new lines of inquiry, including the determination of the direction of flow of information, the identity of the signal that flowed along this pathway, and the function of the signals that linked the two regions. Here, we review landmark steps to these discoveries and highlight the experiments that reveal the significance of portal pathways and more generally, the implications of morphologically distinct nuclei sharing capillary beds.
Collapse
Affiliation(s)
- Rae Silver
- Department of Neuroscience, Barnard College, 3009 Broadway, New York City, NY, 10027, USA
- Columbia University Department of Psychology, 1190 Amsterdam Avenue, New York City, NY, 10027, USA
- Department of Psychology, Barnard College, 3009 Broadway, New York City, NY, 10027, USA
| | - Yifan Yao
- Columbia University Department of Psychology, 1190 Amsterdam Avenue, New York City, NY, 10027, USA
| | - Ranjan K. Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, 30303, USA
| | - Javier E. Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, 30303, USA
| |
Collapse
|
4
|
Sheep as a model for neuroendocrinology research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:1-34. [PMID: 35595346 DOI: 10.1016/bs.pmbts.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Animal models remain essential to understand the fundamental mechanisms of physiology and pathology. Particularly, the complex and dynamic nature of neuroendocrine cells of the hypothalamus make them difficult to study. The neuroendocrine systems of the hypothalamus are critical for survival and reproduction, and are highly conserved throughout vertebrate evolution. Their roles in controlling body metabolism, growth and body composition, stress, electrolyte balance, and reproduction, have been intensively studied, and have yielded groundbreaking discoveries. Many of these discoveries would not have been feasible without the use of the domestic sheep (Ovis aries). The sheep has been used for decades to study the neuroendocrine systems of the hypothalamus and has become a model for human neuroendocrinology. The aim of this chapter is to review some of the profound biomedical discoveries made possible by the use of sheep. The advantages and limitations of sheep as a neuroendocrine model will be discussed. While no animal model can perfectly recapitulate a human disease or condition, sheep are invaluable for enabling manipulations not possible in human subjects and isolating physiologic variables to garner insight into neuroendocrinology and associated pathologies.
Collapse
|
5
|
Ozawa H. Kisspeptin neurons as an integration center of reproductive regulation: Observation of reproductive function based on a new concept of reproductive regulatory nervous system. Reprod Med Biol 2021; 21:e12419. [PMID: 34934400 PMCID: PMC8656200 DOI: 10.1002/rmb2.12419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
Background Regulation of the reproductive system has been explained by the actions and feedback of gonadotropin releasing hormone‐luteinizing hormone/follicle stimulating hormone (GnRH‐LH/FSH) ‐sex steroids; however, the discovery of kisspeptin neurons and a kisspeptin‐GnRH‐LH/FSH axis has prompted this regulation to be reviewed. Methods We investigated changes in kisspeptin neurons and associated changes in the hypothalamic‐pituitary‐gonadal (HPG) axis under various situations and experimental conditions using histochemical methods. Main findings (Results) Kisspeptin neurons play an important role in receiving and integrating information from internal and external environmental factors and communicating it to the conventional HPG axis. Conclusion The recently described Kisspeptin‐GnRH‐LH/FSH‐gonad system regulates reproductive function via mechanisms that until recently were not completely understood.
Collapse
Affiliation(s)
- Hitoshi Ozawa
- Department of Anatomy and Neurobiology Graduate School of Medicine Nippon Medical School Tokyo Japan
| |
Collapse
|
6
|
Graceli JB, Dettogni RS, Merlo E, Niño O, da Costa CS, Zanol JF, Ríos Morris EA, Miranda-Alves L, Denicol AC. The impact of endocrine-disrupting chemical exposure in the mammalian hypothalamic-pituitary axis. Mol Cell Endocrinol 2020; 518:110997. [PMID: 32841708 DOI: 10.1016/j.mce.2020.110997] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022]
Abstract
The hypothalamic-pituitary axis (HP axis) plays a critical and integrative role in the endocrine system control to maintain homeostasis. The HP axis is responsible for the hormonal events necessary to regulate the thyroid, adrenal glands, gonads, somatic growth, among other functions. Endocrine-disrupting chemicals (EDCs) are a worldwide public health concern. There is growing evidence that exposure to EDCs such as bisphenol A (BPA), some phthalates, polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs) and biphenyls (PBBs), dichlorodiphenyltrichloroethane (DDT), tributyltin (TBT), and atrazine (ATR), is associated with HP axis abnormalities. EDCs act on hormone receptors and their downstream signaling pathways and can interfere with hormone synthesis, metabolism, and actions. Because the HP axis function is particularly sensitive to endogenous hormonal changes, disruptions by EDCs can alter HP axis proper function, leading to important endocrine irregularities. Here, we review the evidence that EDCs could directly affect the mammalian HP axis function.
Collapse
Affiliation(s)
- Jones B Graceli
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Raquel S Dettogni
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Eduardo Merlo
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Oscar Niño
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Charles S da Costa
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Jordana F Zanol
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Eduardo A Ríos Morris
- Laboratory of Experimental Endocrinology-LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil. Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil.
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology-LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil. Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil. Graduate Program in Pharmacology and Medicinal Chemistry, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil.
| | - Anna C Denicol
- Department of Animal Science, University of California, Davis, One Shields Avenue Davis, CA, 95616, USA.
| |
Collapse
|
7
|
Ijiro T, Yaguchi A, Yokoyama A, Abe Y, Kiguchi S. Ameliorating effect of rovatirelin on the ataxia in rolling mouse Nagoya. Eur J Pharmacol 2020; 882:173271. [PMID: 32534077 DOI: 10.1016/j.ejphar.2020.173271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 02/01/2023]
Abstract
Rovatirelin is a newly synthetized thyrotropin-releasing hormone (TRH) analog. This study aimed to investigate the effect of rovatirelin on motor function using rolling mouse Nagoya (RMN), a mouse model of hereditary ataxia, and compare it with that of taltirelin, which is clinically used to treat spinocerebellar degeneration in Japan. We also examined the effect of rovatirelin on glucose metabolism in various brain regions of RMN using autoradiography (ARG). Rovatirelin (1, 3, 10, and 30 mg/kg) dose-dependently reduced the fall index in RMN, and its effect was more potent than that of taltirelin (3, 10, 30, and 100 mg/kg). No attenuation of the effect was observed by repeated daily administration for 2 weeks. Furthermore, the reduction in the fall index by rovatirelin persisted for 2 weeks after completing treatment. In the ARG study, rovatirelin induced a significantly elevated uptake of glucose in the prefrontal cortex, nucleus accumbens shell, nucleus accumbens core, striatum, anterior cingulate cortex, secondary motor area, pretectal area, ventral tegmental area, black pars compacta, locus coeruleus, nucleus cerebellaris middle nucleus, medial nucleus of the vestibular nerve, fourth/fifth lobule, and third lobule. Furthermore, rovatirelin increased cerebellar mRNA level of brain derived neurotrophic factor. These results suggest that rovatirelin activates the cerebellum and other parts of the central nervous system to improve motor function in spinocerebellar ataxia (SCA) model animals, and its action is more potent than that of taltirelin. Therefore, rovatirelin can be a potential alternative to the traditionally used therapeutics for SCA.
Collapse
Affiliation(s)
- Tomoyuki Ijiro
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., 4365-1, Hotaka Kashiwabara, Azumino Nagano, 399-8304, Japan
| | - Atsushi Yaguchi
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., 4365-1, Hotaka Kashiwabara, Azumino Nagano, 399-8304, Japan
| | - Ayaka Yokoyama
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., 4365-1, Hotaka Kashiwabara, Azumino Nagano, 399-8304, Japan
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., 4365-1, Hotaka Kashiwabara, Azumino Nagano, 399-8304, Japan
| | - Sumiyoshi Kiguchi
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., 4365-1, Hotaka Kashiwabara, Azumino Nagano, 399-8304, Japan.
| |
Collapse
|
8
|
Kobayashi K, Abe Y, Kawai A, Furihata T, Endo T, Takeda H. Pharmacokinetic Drug Interactions of an Orally Available TRH Analog (Rovatirelin) With a CYP3A4/5 and P-Glycoprotein Inhibitor (Itraconazole). J Clin Pharmacol 2020; 60:1314-1323. [PMID: 32459872 DOI: 10.1002/jcph.1628] [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: 01/28/2020] [Accepted: 03/27/2020] [Indexed: 01/11/2023]
Abstract
The effects of itraconazole on the pharmacokinetics of rovatirelin were investigated in an open-label, single-sequence drug-drug interaction study in 16 healthy subjects. Subjects were administered a single oral dose of rovatirelin (1.6 mg) on day 1 and day 15. From day 8 through 16, subjects received daily oral doses of itraconazole (200 mg/day). Concentrations of rovatirelin and (thiazolylalanyl)methylpyrrolidine (TAMP), the major metabolite of rovatirelin formed by cytochrome P450 (CYP) 3A4/5, were determined in plasma and urine. Pharmacokinetic parameters were used to evaluate the drug-drug interaction potential of rovatirelin as a victim. With coadministration, maximum concentration (Cmax ) and area under the concentration-time curve extrapolated to infinity (AUCinf ) of rovatirelin increased 3.05-fold and 2.82-fold, respectively, and the 90% confidence intervals of the ratios for Cmax (2.64-3.52) and AUCinf (2.47-3.23) did not fall within the 0.8-1.25 boundaries. Urinary excretion of rovatirelin increased at almost the same ratio as the AUCinf ratio with coadministration; however, renal clearance did not change. Cmax , AUCinf , and urinary excretion of TAMP were decreased by coadministration. Itraconazole has the potential to inhibit drug transport via intestinal P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP); therefore, substrate assessments of rovatirelin for the 2 transporters were evaluated using Caco-2 cell monolayers. In vitro studies showed that rovatirelin is a substrate for P-gp but not for BCRP. The current study shows that itraconazole's effect on rovatirelin pharmacokinetics is mediated through inhibition of CYP3A4/5 and intestinal P-gp.
Collapse
Affiliation(s)
- Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., Azumino, Nagano, Japan
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., Azumino, Nagano, Japan
| | - Asuka Kawai
- Clinical Development Division, Kissei Pharmaceutical Co, Ltd., Bunkyo, Tokyo, Japan
| | - Takao Furihata
- Clinical Development Division, Kissei Pharmaceutical Co, Ltd., Bunkyo, Tokyo, Japan
| | - Takuro Endo
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., Azumino, Nagano, Japan
| | - Hiroo Takeda
- Central Research Laboratories, Kissei Pharmaceutical Co, Ltd., Azumino, Nagano, Japan
| |
Collapse
|
9
|
Wang Y, Sabbagh MF, Gu X, Rattner A, Williams J, Nathans J. Beta-catenin signaling regulates barrier-specific gene expression in circumventricular organ and ocular vasculatures. eLife 2019; 8:43257. [PMID: 30932813 PMCID: PMC6443350 DOI: 10.7554/elife.43257] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/17/2019] [Indexed: 12/29/2022] Open
Abstract
The brain, spinal cord, and retina are supplied by capillaries that do not permit free diffusion of molecules between serum and parenchyma, a property that defines the blood-brain and blood-retina barriers. Exceptions to this pattern are found in circumventricular organs (CVOs), small midline brain structures that are supplied by high permeability capillaries. In the eye and brain, high permeability capillaries are also present in the choriocapillaris, which supplies the retinal pigment epithelium and photoreceptors, and the ciliary body and choroid plexus, the sources of aqueous humor and cerebrospinal fluid, respectively. We show here that (1) endothelial cells in these high permeability vascular systems have very low beta-catenin signaling compared to barrier-competent endothelial cells, and (2) elevating beta-catenin signaling leads to a partial conversion of permeable endothelial cells to a barrier-type state. In one CVO, the area postrema, high permeability is maintained, in part, by local production of Wnt inhibitory factor-1.
Collapse
Affiliation(s)
- Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Mark F Sabbagh
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Xiaowu Gu
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
10
|
Kobayshi K, Abe Y, Kawai A, Furihata T, Harada H, Endo T, Takeda H. Human mass balance, pharmacokinetics and metabolism of rovatirelin and identification of its metabolic enzymes in vitro. Xenobiotica 2019; 49:1434-1446. [DOI: 10.1080/00498254.2019.1580796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Kaoru Kobayshi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Asuka Kawai
- Department of Clinical Projects Management, Kissei Pharmaceutical Co., Ltd, Bunkyo, Tokyo, Japan
| | - Takao Furihata
- Department of Clinical Projects Management, Kissei Pharmaceutical Co., Ltd, Bunkyo, Tokyo, Japan
| | - Hiroshi Harada
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takuro Endo
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Hiroo Takeda
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| |
Collapse
|
11
|
Hökfelt T. The new neuroendocrinology-a perspective. J Intern Med 2018; 284:563-567. [PMID: 30277630 DOI: 10.1111/joim.12835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- T Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Deussing JM, Chen A. The Corticotropin-Releasing Factor Family: Physiology of the Stress Response. Physiol Rev 2018; 98:2225-2286. [DOI: 10.1152/physrev.00042.2017] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1–3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jan M. Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
13
|
Kobayashi K, Abe Y, Harada H, Oota E, Endo T, Takeda H. Non-clinical pharmacokinetic profiles of rovatirelin, an orally available thyrotropin-releasing hormone analogue. Xenobiotica 2018; 49:106-119. [DOI: 10.1080/00498254.2017.1423130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan and
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan and
| | - Hiroshi Harada
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan and
| | - Emiko Oota
- Toxicological Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takuro Endo
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan and
| | - Hiroo Takeda
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan and
| |
Collapse
|
14
|
Vu M, Weiler B, Trudeau VL. Time- and dose-related effects of a gonadotropin-releasing hormone agonist and dopamine antagonist on reproduction in the Northern leopard frog (Lithobates pipiens). Gen Comp Endocrinol 2017; 254:86-96. [PMID: 28964731 DOI: 10.1016/j.ygcen.2017.09.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/23/2017] [Accepted: 09/24/2017] [Indexed: 11/25/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) stimulates luteinizing hormone release to control ovulation and spermiation in vertebrates. Dopamine (DA) has a clear inhibitory role in the control of reproduction in numerous teleosts, and emerging evidence suggests that similar mechanisms may exist in amphibians. The interactions between GnRH and DA on spawning success and pituitary gene expression in the Northern leopard frog (Lithobates pipiens) were therefore investigated. Frogs were injected during the natural breeding season with a GnRH agonist [GnRH-A; (Des-Gly10, D-Ala6, Pro-NHEt9)-LHRH; 0.1μg/g and 0.4μg/g] alone and in combination with the dopamine receptor D2 antagonist metoclopramide (MET; 5μg/g and 10μg/g). Injected animals were allowed to breed in outdoor mesocosms. Time to amplexus and oviposition were assessed, and egg mass release, incidences of amplexus, egg mass weight, total egg numbers and fertilization rates were measured. To examine gene expression, female pituitaries were sampled at 12, 24 and 36h following injection of GnRH-A (0.4μg/g) alone and in combination with MET (10μg/g). The mRNA levels of the genes lhb, fshb, gpha, drd2 and gnrhr1 were measured using quantitative real-time PCR. Data were analyzed by a two-way ANOVA. Both GnRH-A doses increased amplexus, oviposition and fertilization alone. Co-injection of MET with GnRH-A did not further enhance spawning success. Injection of GnRH-A alone time-dependently increased expression of lhb, fshb, gpha and gnrhr1. The major effect of MET alone was to decrease expression of drd2. Importantly, the stimulatory effects of GnRH-A on lhb, gpha and gnrhr1 were potentiated by the co-injection of MET at 36h. At this time, expression of fshb was increased only in animals injected with both GnRH-A and MET. Spawning success was primarily driven by the actions of GnRH-A. The hypothesized inhibitory action of DA was supported by pituitary gene expression analysis. The results from this study provide a fundamental framework for future time- and dose-response investigations to improve current spawning methods in amphibians.
Collapse
Affiliation(s)
- Maria Vu
- Department of Biology, University of Ottawa, 30 Marie-Curie Private, Ottawa, Ontario K1N 9B4, Canada.
| | - Bradley Weiler
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, 30 Marie-Curie Private, Ottawa, Ontario K1N 9B4, Canada.
| |
Collapse
|
15
|
Dubovy SR, Fernandez MP, Echegaray JJ, Block NL, Unoki N, Perez R, Vidaurre I, Lee RK, Nadji M, Schally AV. Expression of hypothalamic neurohormones and their receptors in the human eye. Oncotarget 2017; 8:66796-66814. [PMID: 28977997 PMCID: PMC5620137 DOI: 10.18632/oncotarget.18358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/20/2017] [Indexed: 12/27/2022] Open
Abstract
Extrapituitary roles for hypothalamic neurohormones have recently become apparent and clinically relevant, based on the use of synthetic peptide analogs for the treatment of multiple conditions including cancers, pulmonary edema and myocardial infarction. In the eye, it has been suggested that some of these hormones and their receptors may be present in the ciliary body, iris, trabecular meshwork and retina, but their physiological role has yet to be elucidated. Our study intends to comprehensively demonstrate the expression of some hypothalamic neuroendocrine hormones and their receptors within different retinal and extraretinal structures of the human eye. Immunofluorescence, Western blot analysis, and RT-PCR were used to evaluate the qualitative and quantitative expression of Luteinizing Hormone Releasing Hormone (LHRH), Growth Hormone Releasing Hormone (GHRH), Thyrotropin Releasing Hormone (TRH), Gastrin Releasing Peptide (GRP) and Somatostatin as well as their respective receptors (LHRH-R, GHRH-R, TRH-R, GRP-R, SST-R1) in cadaveric human eye tissue and in paraffinized human eye tissue sections. The hypothalamic hormones LHRH, GHRH, TRH, GRP and Somatostatin and their respective receptors (LHRH-R, GHRH-R, TRH-R, GRPR/BB2 and SST-R1), were expressed in the conjunctiva, cornea, trabecular meshwork, ciliary body, lens, retina, and optic nerve.
Collapse
Affiliation(s)
- Sander R Dubovy
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Maria P Fernandez
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Jose J Echegaray
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Ophthalmology, University of Puerto Rico School of Medicine, San Juan, PR, USA
| | - Norman L Block
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Noriyuki Unoki
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Roberto Perez
- Miami Veterans Affairs Medical Center, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | - Richard K Lee
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Mehrdad Nadji
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Andrew V Schally
- Miami Veterans Affairs Medical Center, Miami, Florida, USA.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
16
|
Romanov RA, Alpár A, Hökfelt T, Harkany T. Molecular diversity of corticotropin-releasing hormone mRNA-containing neurons in the hypothalamus. J Endocrinol 2017; 232:R161-R172. [PMID: 28057867 DOI: 10.1530/joe-16-0256] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023]
Abstract
Hormonal responses to acute stress rely on the rapid induction of corticotropin-releasing hormone (CRH) production in the mammalian hypothalamus, with subsequent instructive steps culminating in corticosterone release at the periphery. Hypothalamic CRH neurons in the paraventricular nucleus of the hypothalamus are therefore considered as 'stress neurons'. However, significant morphological and functional diversity among neurons that can transiently produce CRH in other hypothalamic nuclei has been proposed, particularly as histochemical and molecular biology evidence associates CRH to both GABA and glutamate neurotransmission. Here, we review recent advances through single-cell RNA sequencing and circuit mapping to suggest that CRH production reflects a state switch in hypothalamic neurons and thus confers functional competence rather than being an identity mark of phenotypically segregated neurons. We show that CRH mRNA transcripts can therefore be seen in GABAergic, glutamatergic and dopaminergic neuronal contingents in the hypothalamus. We then distinguish 'stress neurons' of the paraventricular nucleus that constitutively express secretagogin, a Ca2+ sensor critical for the stimulus-driven assembly of the molecular machinery underpinning the fast regulated exocytosis of CRH at the median eminence. Cumulatively, we infer that CRH neurons are functionally and molecularly more diverse than previously thought.
Collapse
Affiliation(s)
- Roman A Romanov
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Alán Alpár
- MTA-SE NAP Research Group of Experimental Neuroanatomy and Developmental BiologyHungarian Academy of Sciences, Budapest, Hungary
- Department of AnatomySemmelweis University, Budapest, Hungary
| | - Tomas Hökfelt
- Department of NeuroscienceKarolinska Institutet, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of NeuroscienceKarolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Sui H, Wang Y, Zhang X, Wang X, Cheng W, Su H, Wang X, Sun X, Han XX, Zhao B, Ozaki Y. Ultrasensitive detection of thyrotropin-releasing hormone based on azo coupling and surface-enhanced resonance Raman spectroscopy. Analyst 2016; 141:5181-5188. [PMID: 27338554 DOI: 10.1039/c6an00884d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Surface-enhanced resonance Raman scattering (SERRS) has been used to establish a rapid and quantitative assay based on the diazotization coupling reaction for thyrotropin-releasing hormone (TRH). Ultrahigh sensitivity of this approach originates from two factors: changing TRH to an azo compound and the SERRS effect with the addition of silver nanoparticles (AgNPs) at 532 nm excitation wavelength. The lowest detectable concentration of TRH was found to be as low as 1 pg mL(-1), which is 10-fold lower than the lowest normal reference value in human serum reported in previous literature. The quantitative measurements in human serum based on this method were conducted, and the results showed its feasibility for detection in complex biological samples. In comparison with conventional TRH identification and quantification methodologies, radioimmunoassay (RIA) and subsequent various hyphenated techniques, the main advantages of this study are simplicity, rapidness (2 minutes), time effectiveness, no additional steps required to further characterize the immunogenic material, highest sensitivity (57.1 fg), high selectivity, practicality and reliability. Thus, this work puts forward a research tool that may be applied to the determination of TRH in practical assays.
Collapse
Affiliation(s)
- Huimin Sui
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Yue Wang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Xiaolei Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Xiaolei Wang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Weina Cheng
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Hongyang Su
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Xu Wang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Xiaoying Sun
- China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Bing Zhao
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Street, Changchun, P.R. China.
| | - Yukihiro Ozaki
- Department of Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo 669-1337, Japan
| |
Collapse
|
18
|
Uenoyama Y, Pheng V, Tsukamura H, Maeda KI. The roles of kisspeptin revisited: inside and outside the hypothalamus. J Reprod Dev 2016; 62:537-545. [PMID: 27478063 PMCID: PMC5177970 DOI: 10.1262/jrd.2016-083] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Kisspeptin, encoded by KISS1/Kiss1 gene, is now considered a master regulator of reproductive functions in mammals owing to
its involvement in the direct activation of gonadotropin-releasing hormone (GnRH) neurons after binding to its cognate receptor, GPR54. Ever since the discovery
of kisspeptin, intensive studies on hypothalamic expression of KISS1/Kiss1 and on physiological roles of hypothalamic
kisspeptin neurons have provided clues as to how the brain controls sexual maturation at the onset of puberty and subsequent reproductive performance in
mammals. Additionally, emerging evidence indicates the potential involvement of extra-hypothalamic kisspeptin in reproductive functions. Here, we summarize data
regarding kisspeptin inside and outside the hypothalamus and revisit the physiological roles of central and peripheral kisspeptins in the reproductive functions
of mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | |
Collapse
|
19
|
Jee C, Goncalves JF, LeBoeuf B, Garcia LR. CRF-like receptor SEB-3 in sex-common interneurons potentiates stress handling and reproductive drive in C. elegans. Nat Commun 2016; 7:11957. [PMID: 27321013 PMCID: PMC4915151 DOI: 10.1038/ncomms11957] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/13/2016] [Indexed: 02/08/2023] Open
Abstract
Environmental conditions can modulate innate behaviours. Although male Caenorhabditis elegans copulation can be perturbed in the presence of stress, the mechanisms underlying its decision to sustain copulation are unclear. Here we describe a mating interference assay, which quantifies the persistence of male C. elegans copulation in noxious blue light. We show that between copulations, the male escapes from blue light illumination at intensities over 370 μW mm−2. This response is attenuated in mutants with constitutive activation of the corticotropin-releasing factor receptor family homologue SEB-3. We show that activation of this receptor causes sex-common glutamatergic lumbar ganglion interneurons (LUA) to potentiate downstream male-specific reproduction circuits, allowing copulatory behaviours to partially override the light-induced escape responses in the male. SEB-3 activation in LUA also potentiates copulation during mild starvation. We suggest that SEB-3 activation allows C. elegans to acclimate to the environment and thus continue to execute innate behaviours even under non-optimal conditions. Innate animal behaviours can be negatively regulated by environmental stressors. Jee et al. show that suppression of male C. elegans copulation behaviour by noxious light can be overcome by activation of SEB-3, a homologue of the stress-associated mammalian corticotropin-releasing factor receptor family.
Collapse
Affiliation(s)
- Changhoon Jee
- Department of Biology, Howard Hughes Medical Institute, Texas A&M University, 3258 TAMU, College Station, Texas 77843-3258, USA
| | - Jimmy F Goncalves
- Department of Biology, Howard Hughes Medical Institute, Texas A&M University, 3258 TAMU, College Station, Texas 77843-3258, USA
| | - Brigitte LeBoeuf
- Department of Biology, Howard Hughes Medical Institute, Texas A&M University, 3258 TAMU, College Station, Texas 77843-3258, USA
| | - L Rene Garcia
- Department of Biology, Howard Hughes Medical Institute, Texas A&M University, 3258 TAMU, College Station, Texas 77843-3258, USA
| |
Collapse
|
20
|
In vitro digestion of luteinizing hormone releasing hormone analog (LHRHa) using simulated gastric conditions in assessing human food safety. Food Chem 2016; 192:409-14. [DOI: 10.1016/j.foodchem.2015.06.091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 06/18/2015] [Accepted: 06/26/2015] [Indexed: 11/18/2022]
|
21
|
Brown PJ, Cleghorn JM, Brown GM, Blackall MH. The Neuroendocrinology of Schizophrenia. INTERNATIONAL JOURNAL OF MENTAL HEALTH 2015. [DOI: 10.1080/00207411.1980.11448865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Ijiro T, Nakamura K, Ogata M, Inada H, Kiguchi S, Maruyama K, Nabekura J, Kobayashi M, Ishibashi H. Effect of rovatirelin, a novel thyrotropin-releasing hormone analog, on the central noradrenergic system. Eur J Pharmacol 2015; 761:413-22. [DOI: 10.1016/j.ejphar.2015.05.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/05/2023]
|
23
|
Romanov RA, Alpár A, Zhang MD, Zeisel A, Calas A, Landry M, Fuszard M, Shirran SL, Schnell R, Dobolyi Á, Oláh M, Spence L, Mulder J, Martens H, Palkovits M, Uhlen M, Sitte HH, Botting CH, Wagner L, Linnarsson S, Hökfelt T, Harkany T. A secretagogin locus of the mammalian hypothalamus controls stress hormone release. EMBO J 2015; 34:36-54. [PMID: 25430741 PMCID: PMC4291479 DOI: 10.15252/embj.201488977] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/07/2014] [Accepted: 10/21/2014] [Indexed: 11/09/2022] Open
Abstract
A hierarchical hormonal cascade along the hypothalamic-pituitary-adrenal axis orchestrates bodily responses to stress. Although corticotropin-releasing hormone (CRH), produced by parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) and released into the portal circulation at the median eminence, is known to prime downstream hormone release, the molecular mechanism regulating phasic CRH release remains poorly understood. Here, we find a cohort of parvocellular cells interspersed with magnocellular PVN neurons expressing secretagogin. Single-cell transcriptome analysis combined with protein interactome profiling identifies secretagogin neurons as a distinct CRH-releasing neuron population reliant on secretagogin's Ca(2+) sensor properties and protein interactions with the vesicular traffic and exocytosis release machineries to liberate this key hypothalamic releasing hormone. Pharmacological tools combined with RNA interference demonstrate that secretagogin's loss of function occludes adrenocorticotropic hormone release from the pituitary and lowers peripheral corticosterone levels in response to acute stress. Cumulatively, these data define a novel secretagogin neuronal locus and molecular axis underpinning stress responsiveness.
Collapse
Affiliation(s)
- Roman A Romanov
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alán Alpár
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ming-Dong Zhang
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Amit Zeisel
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - André Calas
- Laboratory for Central Mechanisms of Pain Sensitization, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 Université Bordeaux 2, Bordeaux, France
| | - Marc Landry
- Laboratory for Central Mechanisms of Pain Sensitization, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 Université Bordeaux 2, Bordeaux, France
| | - Matthew Fuszard
- School of Chemistry, University of St. Andrews, St. Andrews, UK
| | - Sally L Shirran
- School of Chemistry, University of St. Andrews, St. Andrews, UK
| | - Robert Schnell
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Árpád Dobolyi
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Márk Oláh
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Lauren Spence
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | | | - Miklós Palkovits
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, Hungary
| | - Mathias Uhlen
- Science for Life Laboratory, Albanova University Center, Royal Institute of Technology, Stockholm, Sweden
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology Medical University of Vienna, Vienna, Austria
| | | | - Ludwig Wagner
- University Clinic for Internal Medicine III General Hospital Vienna, Vienna, Austria
| | - Sten Linnarsson
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tibor Harkany
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
Related pituitary cell lineages develop into interdigitated 3D cell networks. Proc Natl Acad Sci U S A 2011; 108:12515-20. [PMID: 21746936 DOI: 10.1073/pnas.1105929108] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The pituitary gland has long been considered to be a random patchwork of hormone-producing cells. By using pituitary-scale tridimensional imaging for two of the least abundant cell lineages, the corticotropes and gonadotropes, we have now uncovered highly organized and interdigitated cell networks that reflect homotypic and heterotypic interactions between cells. Although newly differentiated corticotrope cells appear on the ventral surface of the gland, they rapidly form homotypic strands of cells that extend from the lateral tips of the anterior pituitary along its ventral surface and into the medial gland. As the corticotrope network is established away from the microvasculature, cell morphology changes from rounded, to polygonal, and finally to cells with long cytoplasmic processes or cytonemes that connect corticotropes to the perivascular space. Gonadotropes differentiate later and are positioned in close proximity to corticotropes and capillaries. Blockade of corticotrope terminal differentiation produced by knockout of the gene encoding the transcription factor Tpit results in smaller gonadotropes within an expanded cell network, particularly in the lateral gland. Thus, pituitary-scale tridimensional imaging reveals highly structured cell networks of unique topology for each pituitary lineage. The sequential development of interdigitated cell networks during organogenesis indicate that extensive cell:cell interactions lead to a highly ordered cell positioning rather than random patchwork.
Collapse
|
25
|
|
26
|
Abstract
IMPORTANCE OF THE FIELD Luteinizing hormone releasing hormone (LH RH) agonists are the major agent for androgen deprivation therapy in advanced and metastatic prostate cancer. They also have a role in endometriosis, uterine fibroids and central precocious puberty. AREAS COVERED IN THE REVIEW Triptorelin embonate 22.5 mg is a new, sustained-release, 6-month formulation of an LH RH agonist. It possesses longer duration of action than the current standard 3-month preparation and appears to have similar efficacy and side effects. WHAT THE READER WILL GAIN The use of LH RH agonists for androgen deprivation in prostate cancer has increased considerably in the last 20 years. Recent work has shown that some of this usage has constituted overtreatment and it is within these newer paradigms of therapy that the new 6-month preparation is situated. TAKE HOME MESSAGE The new 6-month LH RH preparation - triptorelin embonate - will be of help in several key areas of therapy for prostate cancer, notably as an adjunct to radiation therapy and chemotherapy. It possesses a similar effect, but with fewer side effects, than those that are now commonly available.
Collapse
|
27
|
Watts AG. Structure and function in the conceptual development of mammalian neuroendocrinology between 1920 and 1965. ACTA ACUST UNITED AC 2010; 66:174-204. [PMID: 20637232 DOI: 10.1016/j.brainresrev.2010.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 06/20/2010] [Accepted: 07/07/2010] [Indexed: 11/28/2022]
Abstract
With the growing realization in the 1930s that the brain played a crucial role in regulating the secretions of the pituitary gland, neuroendocrinology as we now know it developed from two rather separate directions. One approach relied heavily on morphological techniques to define neurosecretion; a novel, but for many years flawed model that was originally developed to explain the presence of gland-like cells in the diencephalon. During its first 20 years neurosecretion, as a concept, made no significant contribution to our understanding of how the pituitary was controlled. Then, following the identification by Sanford Palay and Wolfgang Bargmann of a continuous neurosecretory pathway from the hypothalamus to the neural lobe, neurosecretion became incorporated into a more broadly based concept of pituitary function, particularly regarding the neural lobe. The second approach integrated structural and functional methods to investigate neural regulation of the pituitary. This work eventually explained how the pituitary was controlled by the brain. It led directly to our understanding of the control of vasopressin and oxytocin release by neuroendocrine terminals in the neural lobe, the neurohumoral control of the pars distalis, and eventually to a detailed description of the neural networks that control pituitary function. As increasingly sophisticated morphological, neurophysiological, and eventually molecular biological techniques were applied to the problem, the original notion of the diencephalic gland and neurosecretion became unsustainable. The gland-nerve cells of the 1930s became the neurosecretory cells of the 1940s and 1950s, and then finally neuroendocrine neurons in the 1960s. From then on neuroendocrinology developed into the more unified discipline we know today. The chronology of these two approaches will be examined here using examples from research that occurred approximately between 1920 and 1965. The goal is not to give a comprehensive history of pituitary function or neuroendocrinology. Instead, the focus will be to compare the rationales and effectiveness of two contrasting experimental approaches: predominantly structural analyses as opposed to more integrated approaches.
Collapse
Affiliation(s)
- Alan G Watts
- Department of Biological Sciences, The USC College, University of Southern California, Los Angeles, CA 90089-2520, USA.
| |
Collapse
|
28
|
Mylonas CC, Fostier A, Zanuy S. Broodstock management and hormonal manipulations of fish reproduction. Gen Comp Endocrinol 2010; 165:516-34. [PMID: 19318108 DOI: 10.1016/j.ygcen.2009.03.007] [Citation(s) in RCA: 205] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 03/12/2009] [Accepted: 03/16/2009] [Indexed: 11/17/2022]
Abstract
Control of reproductive function in captivity is essential for the sustainability of commercial aquaculture production, and in many fishes it can be achieved by manipulating photoperiod, water temperature or spawning substrate. The fish reproductive cycle is separated in the growth (gametogenesis) and maturation phase (oocyte maturation and spermiation), both controlled by the reproductive hormones of the brain, pituitary and gonad. Although the growth phase of reproductive development is concluded in captivity in most fishes-the major exemption being the freshwater eel (Anguilla spp.), oocyte maturation (OM) and ovulation in females, and spermiation in males may require exogenous hormonal therapies. In some fishes, these hormonal manipulations are used only as a management tool to enhance the efficiency of egg production and facilitate hatchery operations, but in others exogenous hormones are the only way to produce fertilized eggs reliably. Hormonal manipulations of reproductive function in cultured fishes have focused on the use of either exogenous luteinizing hormone (LH) preparations that act directly at the level of the gonad, or synthetic agonists of gonadotropin-releasing hormone (GnRHa) that act at the level of the pituitary to induce release of the endogenous LH stores, which, in turn act at the level of the gonad to induce steroidogenesis and the process of OM and spermiation. After hormonal induction of maturation, broodstock should spawn spontaneously in their rearing enclosures, however, the natural breeding behavior followed by spontaneous spawning may be lost in aquaculture conditions. Therefore, for many species it is also necessary to employ artificial gamete collection and fertilization. Finally, a common question in regards to hormonal therapies is their effect on gamete quality, compared to naturally maturing or spawning broodfish. The main factors that may have significant consequences on gamete quality-mainly on eggs-and should be considered when choosing a spawning induction procedure include (a) the developmental stage of the gonads at the time the hormonal therapy is applied, (b) the type of hormonal therapy, (c) the possible stress induced by the manipulation necessary for the hormone administration and (d) in the case of artificial insemination, the latency period between hormonal stimulation and stripping for in vitro fertilization.
Collapse
Affiliation(s)
- Constantinos C Mylonas
- Institute of Aquaculture, Hellenic Center for Marine Research, P.O. Box 2214, Heraklion, Crete 71003, Greece.
| | | | | |
Collapse
|
29
|
Hökfelt T. Looking at neurotransmitters in the microscope. Prog Neurobiol 2009; 90:101-18. [PMID: 19853008 DOI: 10.1016/j.pneurobio.2009.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 04/16/2009] [Accepted: 10/08/2009] [Indexed: 02/07/2023]
Abstract
This review article covers the early period of my career. I first summarize research initiated by the late Nils-Ake Hillarp, after his appointment in 1962 as professor in the Department of Histology at Karolinska Institutet. He only lived for three more years, but during this short period he started up a group of ten students who explored various aspects of the three monoamine transmitters, dopamine, noradrenaline and 5-hydroxytryptamine, using the new formaldehyde fluorescence method developed by Bengt Falck and Hillarp in Lund. This method allowed visualization of the cellular localization in the microscope of these monoamines, which introduced a new discipline in neurobiology-chemical neuroanatomy. I then deal with work aiming at localizing the monoamines at the ultrastructural level, as well as attempts to use radioactively labeled aminoacids, especially gamma-aminobutyric acid (GABA), and autoradiography, to identify, in the microscope, neurons using such transmitters. Finally, our immunohistochemical work together with Kjell Fuxe and the late Menek Goldstein, using antibodies to four monoamine-synthesizing enzymes is summarized, including some aspects on the adrenaline neurons, which had escaped detection with the Falck-Hillarp technique.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, S-17177 Stockholm, Sweden.
| |
Collapse
|
30
|
Thyrotropin-releasing hormone d,l polylactide nanoparticles (TRH-NPs) protect against glutamate toxicity in vitro and kindling development in vivo. Brain Res 2009; 1303:151-60. [PMID: 19766611 DOI: 10.1016/j.brainres.2009.09.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 09/03/2009] [Accepted: 09/11/2009] [Indexed: 11/22/2022]
Abstract
Thyrotropin-releasing hormone (TRH) is reported to have anticonvulsant effects in animal seizure models and certain intractable epileptic patients. However, its duration of action is limited by rapid tissue metabolism and the blood brain barrier. Direct nose-brain delivery of neuropeptides in sustained-release biodegradable nanoparticles (NPs) is a promising mode of therapy for enhancing CNS bioavailability. Bioactivity/neuroprotection of d,l polylactide nanoparticles containing TRH was assessed against glutamate toxicity in cultured rat fetal hippocampal neurons. Subsequently, we utilized the kindling model of temporal lobe epilepsy to determine if intranasal administration of nanoparticles containing TRH (TRH-NPs) could inhibit kindling development. Animals received daily treatments of either blank (control) or TRH-NPs for 7 days before initiation of kindling. On day 8 and each day thereafter until either fully kindled or until day 20, the animals received daily treatments before receiving a kindling stimulus 3 h later. Afterdischarge duration (ADD) was assessed via electroencephalographs recorded from electrodes in the basolateral amygdalae and behavioral seizure stereotypy was simultaneously recorded digitally. Intranasal application of TRH-NPs resulted in a significant reduction in seizure ADD as kindling progressed, while the number of stimulations required to reach stage V seizures and to become permanently kindled was significantly greater in TRH-NP-treated subjects. Additionally, delay to clonus was significantly prolonged while clonus duration was reduced indicating a less severe seizure in TRH-NP-treated subjects. Our results provide proof of principle that intranasal delivery of sustained-release TRH-NPs may be neuroprotective and can be utilized to suppress seizures and perhaps epileptogenesis.
Collapse
|
31
|
|
32
|
Sower SA, Freamat M, Kavanaugh SI. The origins of the vertebrate hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-thyroid (HPT) endocrine systems: new insights from lampreys. Gen Comp Endocrinol 2009; 161:20-9. [PMID: 19084529 DOI: 10.1016/j.ygcen.2008.11.023] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 11/04/2008] [Accepted: 11/20/2008] [Indexed: 11/30/2022]
Abstract
The acquisition of a hypothalamic-pituitary axis was a seminal event in vertebrate evolution leading to the neuroendocrine control of many complex functions including growth, reproduction, osmoregulation, stress and metabolism. Lampreys as basal vertebrates are the earliest evolved vertebrates for which there are demonstrated functional roles for two gonadotropin-releasing hormones (GnRHs) that act via the hypothalamic-pituitary-gonadal axis controlling reproductive processes. With the availability of the lamprey genome, we have identified a novel GnRH form (lamprey GnRH-II) and a novel glycoprotein hormone receptor, lGpH-R II (thyroid-stimulating hormone-like receptor). Based on functional studies, in situ hybridization and phylogenetic analysis, we hypothesize that the newly identified lamprey GnRH-II is an ancestral GnRH to the vertebrate GnRHs. This finding opens a new understanding of the GnRH family and can help to delineate the evolution of the complex neuro/endocrine axis of reproduction. A second glycoprotein hormone receptor (lGpH-R II) was also identified in the sea lamprey. The existing data suggest the existence of a primitive, overlapping yet functional HPG and HPT endocrine systems in this organism, involving one possibly two pituitary glycoprotein hormones and two glycoprotein hormone receptors as opposed to three or four glycoprotein hormones interacting specifically with three receptors in gnathostomes. We hypothesize that the glycoprotein hormone/glycoprotein hormone receptor systems emerged as a link between the neuro-hormonal and peripheral control levels during the early stages of gnathostome divergence. The significance of the results obtained by analysis of the HPG/T axes in sea lamprey may transcend the limited scope of the corresponding physiological compartments by providing important clues in respect to the interplay between genome-wide events (duplications), coding sequence (mutation) and expression control level evolutionary mechanisms in definition of the chemical control pathways in vertebrates.
Collapse
Affiliation(s)
- Stacia A Sower
- Center for Molecular and Comparative Endocrinology and Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA.
| | | | | |
Collapse
|
33
|
Bauce LG, Goren HJ. Synthesis of a series of residue 1 (pyroglutamic acid) analogs of thyrotrophin releasing hormone. INTERNATIONAL JOURNAL OF PEPTIDE AND PROTEIN RESEARCH 2009; 14:216-26. [PMID: 118133 DOI: 10.1111/j.1399-3011.1979.tb01928.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This report describes the synthesis of 23 analogs of thyrotrophin releasing hormone, L-pyroglutamic acid-L-histidyl-L-prolineamide, where only the pyroglutamic acid moiety is modified. Twelve of the analogs contain different heterocyclic rings or are derivatized pyrrolidone rings. The syntheses of these pyroglutamic acid analogs are also described. Peptide bond formation was generally achieved by catalyzing carbodiimide coupling with 1-hydroxybenzotriazole. The histidine side chain was protected with the 2,4-dinitrophenyl group during di- and tripeptide synthesis and was removed with benzenethiol Final purification of tripeptides involved passage of synthetic material over a Dowex 1--X4 (bicarbonate) column. The route for synthesis of thyrotrophin releasing hormone and its analogs is highly efficient, since the native hormone may be obtained with 90% efficiency.
Collapse
|
34
|
Autonomous Rhythmic Drug Delivery Systems Based on Chemical and Biochemomechanical Oscillators. CHEMOMECHANICAL INSTABILITIES IN RESPONSIVE MATERIALS 2009. [DOI: 10.1007/978-90-481-2993-5_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
López JM, Domínguez L, González A. Immunohistochemical localization of thyrotropin-releasing hormone in the brain of reptiles. J Chem Neuroanat 2008; 36:251-63. [DOI: 10.1016/j.jchemneu.2008.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 06/26/2008] [Accepted: 06/26/2008] [Indexed: 01/31/2023]
|
36
|
Carroll AR, Urban S, Lamb J, Moni R, Guymer GP, Forster PI, Quinn RJ. Corymbones A and B, phloroglucinols with thyrotropin releasing hormone receptor 2 binding affinity from the flowers of Corymbia peltata. JOURNAL OF NATURAL PRODUCTS 2008; 71:881-883. [PMID: 18412395 DOI: 10.1021/np0706567] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
High-throughput screeing of a plant and marine invertebrate extract library to find natural products with rat thytotropin releasing hormone receptor 2 binding affinity led to the isolation of two new active acylphloroglucinols, corymbones A and B (1 and 2) from flowers of the Queensland tree Corymbia peltata. Their structures were assigned from interpretation of 2D NMR and high-resolution ESIMS data. Compounds 1 and 2 showed rat TRH receptor 2 binding affinity with IC 50 values of 23 and 19 microM, respectively.
Collapse
Affiliation(s)
- Anthony R Carroll
- Eskitis Institute, Griffith University, Brisbane, Queensland, Australia
| | | | | | | | | | | | | |
Collapse
|
37
|
Domínguez L, López JM, González A. Distribution of Thyrotropin-Releasing Hormone (TRH) Immunoreactivity in the Brain of Urodele Amphibians. BRAIN, BEHAVIOR AND EVOLUTION 2008; 71:231-46. [DOI: 10.1159/000122835] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 12/13/2007] [Indexed: 01/28/2023]
|
38
|
Aoki Y, Ono H, Yasuo S, Masuda T, Yoshimura T, Ebihara S, Iigo M, Yanagisawa T. Molecular Evolution of Prepro-Thyrotropin-Releasing Hormone in the Chicken (Gallus gallus) and Its Expression in the Brain. Zoolog Sci 2007; 24:686-92. [PMID: 17824776 DOI: 10.2108/zsj.24.686] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 01/28/2007] [Indexed: 11/17/2022]
Abstract
A cDNA encoding prepro-thyrotropin-relaesing hormone (ppTRH) in chicken (Gallus gallus) was isolated and the sites of expression in the brain were determined. The chicken ppTRH cDNA encodes 260 amino acids, including four TRH progenitor sequences (-Lys/Arg-Arg-Gln-His-Pro-Gly-Lys/Arg-Arg-). It is interesting to note that chicken ppTRH harbors four TRH progenitor-like sequences. According to the hydropathy profile of chicken ppTRH, not only the TRH progenitor sequences but also the TRH progenitor-like sequences are localized in hydrophilic regions. The TRH progenitor-like sequences might be related to structural conservation in the evolution of ppTRH, although they cannot be processed into TRH due to the mutation of several amino acids. According to the alignment of the deduced amino-acid sequences of known vertebrate ppTRHs and the molecular phylogenetic tree we constructed, we speculate on the molecular evolution of ppTRH in vertebrates. In situ hybridization demonstrated experession of the ppTRH gene in the nucleus preopticus periventricularis, nucleus preopticus medialis, regio lateralis hypothalami, paraventricular nucleus, nucleus periventricularis hypothalami, and nucleus ventromedialis hypothalami in the chicken brain.
Collapse
Affiliation(s)
- Yasuhiro Aoki
- Department of Biotechnology, United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Aoki Y, Masuda T, Iigo M, Yanagisawa T. Molecular cloning of prepro-thyrotropin-releasing hormone cDNA from medaka (Oryzias latipes). Gen Comp Endocrinol 2007; 150:364-70. [PMID: 17098236 DOI: 10.1016/j.ygcen.2006.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 09/25/2006] [Accepted: 09/27/2006] [Indexed: 11/18/2022]
Abstract
The cDNA encoding prepro-thyrotropin-releasing hormone (ppTRH) in a teleost, medaka (Oryzias latipes) was isolated and characterized. The medaka ppTRH cDNA codes for 270 amino acid residues including eight TRH progenitor sequences (-Lys/Arg-Arg-Gln-His-Pro-Gly-Lys/Arg-Arg-). In silico analyses of the medaka genome database predicted that the structure of the medaka ppTRH gene is similar to the ppTRH genes of the other vertebrate species studied to date; consisting of three exons and two introns. Identity of the medaka ppTRH with the other vertebrates is rather low except the sockeye salmon. A molecular phylogenic tree showed that the ppTRH sequences reflected the predicted pattern of species classification. RT-PCR analysis demonstrated ppTRH gene expression in the brain and retina. These results gave some insight into the molecular evolution of ppTRH and physiological functions of TRH in vertebrates.
Collapse
Affiliation(s)
- Yasuhiro Aoki
- Department of Biotechnology, United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | | | | | | |
Collapse
|
40
|
Abstract
Considerable changes are afoot in the practice of assisted human conception. Doubts about its methods, especially over endocrinology, concern its complexity and its expense. IVF has spread worldwide since its beginnings in the UK, but its current practice, termed routine IVF, is being challenged by simpler routines. These include natural cycle IVF, which has been in the background for many years, minimal stimulation IVF, where doses of hormones are reduced, and the in-vitro maturation of human oocytes ready for fertilization in vitro (IVM). These three approaches are now practised in increasing numbers of IVF clinics, and may well replace routine IVF. The events leading to current interest in these methods will be discussed briefly in this review.
Collapse
Affiliation(s)
- R G Edwards
- Reproductive BioMedicine Online, Duck End Farm, Dry Drayton, Cambridge CB23 8DB, UK.
| |
Collapse
|
41
|
Role of liquid membranes in drug action – Experimental studies. STUDIES IN INTERFACE SCIENCE 2005:124-218. [DOI: 10.1016/s1383-7303(05)80045-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
42
|
Rabeler R, Mittag J, Geffers L, Rüther U, Leitges M, Parlow AF, Visser TJ, Bauer K. Generation of Thyrotropin-Releasing Hormone Receptor 1-Deficient Mice as an Animal Model of Central Hypothyroidism. Mol Endocrinol 2004; 18:1450-60. [PMID: 14988432 DOI: 10.1210/me.2004-0017] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
AbstractTo provide an animal model of central hypothyroidism, mice deficient in the TRH-receptor 1 (TRH-R1) gene were generated by homologous recombination. The pituitaries of TRH-R1−/− mice are devoid of any TRH-binding capacity, demonstrating that TRH-R1 is the only receptor localized on TRH target cells of the pituitary. With the exception of some retardation in growth rate, TRH-R1−/− mice appear normal, but compared with control animals they exhibit a considerable decrease in serum T3, T4, and prolactin (PRL) levels but not in serum TSH levels. In situ hybridization histochemistry and real-time RT-PCR analysis revealed that in adult TRH-R1−/− animals TSHβ-mRNA expression is not impaired whereas PRL mRNA and GH mRNA levels are considerably reduced compared with control mice. The numbers of thyrotropes, somatotropes, and lactotropes, however, are not affected by the deletion of the TRH-R1 gene. The mutant mice are fertile, and the dams nourish their pups well, indicating that TRH is not a decisive factor for suckling-induced PRL release. In situ hybridization and quantitative RT-PCR analysis, furthermore, revealed that, as in control animals, pituitary PRL-mRNA expression in TRH-R1−/− is considerably increased during lactation, albeit strongly reduced as compared with lactating control animals.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Southern
- Disease Models, Animal
- Female
- Genetic Vectors
- Genotype
- Growth Hormone/metabolism
- Hypothyroidism/genetics
- Hypothyroidism/pathology
- In Situ Hybridization
- Lac Operon
- Lactation
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Models, Genetic
- Mutation
- Pituitary Gland/metabolism
- Pituitary Hormones/metabolism
- Polymerase Chain Reaction
- Prolactin/blood
- RNA, Messenger/metabolism
- Receptors, Thyrotropin-Releasing Hormone/genetics
- Receptors, Thyrotropin-Releasing Hormone/physiology
- Recombination, Genetic
- Reverse Transcriptase Polymerase Chain Reaction
- Thyrotropin-Releasing Hormone/blood
- Thyroxine/blood
- Time Factors
- Transcription, Genetic
- Triiodothyronine/blood
Collapse
Affiliation(s)
- Roland Rabeler
- Max-Planck-Institut für experimentelle Endokrinologie, Feodor-Lynen-Strasse 7, D-30625 Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The role of peptides as signalling molecules in the nervous system has been studied for more than 30 years. Neuropeptides and their G-protein-coupled receptors are widely distributed throughout the body and they commonly occur with, and are complementary to, classic neurotransmitters. The functions of neuropeptides range from neurotransmitter to growth factor. They are present in glial cells, are hormones in the endocrine system, and are messengers in the immune system. Much evidence indicates that neuropeptides are of particular importance when the nervous system is challenged (eg, by stress, injury, or drug abuse). These features and the large number of neuropeptides and neuropeptide receptors provide many opportunities for the discovery of new drug targets for the treatment of nervous-system disorders. In fact, receptor-subtype-selective antagonists and agonists have been developed, and recently a substance P receptor (neurokinin 1) antagonist has been shown to have clinical efficacy in the treatment of major depression and chemotherapy-induced emesis. Several other neuropeptide receptor ligands are in clinical trials for various indications.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | | | | |
Collapse
|
44
|
Teijido O, Manso MJ, Anadón R. Distribution of thyrotropin-releasing hormone immunoreactivity in the brain of the dogfish Scyliorhinus canicula. J Comp Neurol 2002; 454:65-81. [PMID: 12410619 DOI: 10.1002/cne.10431] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
To improve knowledge of the peptidergic systems of elasmobranch brains, the distribution of thyrotropin-releasing hormone-immunoreactive (TRHir) neurons and fibers was studied in the brain of the small-spotted dogfish (Scyliorhinus canicula L.). In the olfactory bulbs, small granule neurons richly innervated the olfactory glomeruli. In the telencephalic hemispheres, small TRHir neurons were observed in the superficial dorsal pallium, whereas TRHir fibers were widely distributed in pallial and subpallial regions. In the preoptic region, TRHir neurons formed a caudal ventrolateral group in the preoptic nucleus. In the hypothalamus, the most conspicuous TRHir populations were associated with the lateral hypothalamic recess, but small TRHir populations were found in the posterior tubercle and ventral wall of the posterior recess. The preoptic region and hypothalamus exhibited rich innervation by TRHir fibers. TRHir fibers were observed coursing to the neurohypophysis and the neuroepithelium of the saccus vasculosus, but not to the neurohemal region of the median eminence. Some stellate-like TRHir cells were observed in a few cell cords of the neurointermediate lobe of the hypophysis. The thalamus, pretectum, and midbrain lacked TRHir neurons. Further TRHir neuronal populations were observed in the central gray and superior raphe nucleus of the isthmus, and a few TRHir cells were located in the nucleus of the trigeminal descending tract at the level of the rostral spinal cord. In the brainstem, the central gray, interpeduncular nucleus, secondary visceral region of the isthmus, rhombencephalic raphe, inferior olive, vagal lobe, and Cajal's commissural nucleus were all richly TRHir-innervated. Comparison of the distribution of TRHir neurons observed in the dogfish brain with that observed in teleosts and tetrapods reveals strong resemblance but also interesting differences, indicating the presence of both a conserved basic vertebrate pattern and a number of derived characters.
Collapse
Affiliation(s)
- Oscar Teijido
- Department of Cell and Molecular Biology, Faculty of Sciences, University of A Coruña, 15071-A Coruña, Spain
| | | | | |
Collapse
|
45
|
Del Carmen De Andrés M, Anadón R, Manso MJ, González MJ. Distribution of thyrotropin-releasing hormone immunoreactivity in the brain of larval and adult sea lampreys, Petromyzon marinus L. J Comp Neurol 2002; 453:323-35. [PMID: 12389205 DOI: 10.1002/cne.10385] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This study investigated the distribution of thyrotropin-releasing hormone-immunoreactive (TRHir) neurons and fibers in the brain and retina of lampreys. Our results in the brains of large larvae and upstream-migrating adults of the sea lamprey showed the presence of TRHir neurons mainly in the preoptic region and the hypothalamus. A few TRHir neurons were also found in the striatum. The number and staining intensity of TRHir neurons increased from larval stages to adulthood, and the distribution of TRHir populations was wider in adults. The TRHir fibers were more easily traced in adults. Some TRHir fibers entered the neurohypophysis, although most fibers coursed in the different regions of the brain, mostly in the basal region, from the forebrain to the hindbrain. The presence of TRHir stellate cells was observed in the adenohypophysis. In the retina of adult lampreys, but not in that of larvae, TRHir amacrine cells are present.
Collapse
Affiliation(s)
- María Del Carmen De Andrés
- Department of Cell and Molecular Biology, Faculty of Sciences, University of A Coruña, 15071-A Coruña, Spain
| | | | | | | |
Collapse
|
46
|
Díaz ML, Becerra M, Manso MJ, Anadón R. Distribution of thyrotropin-releasing hormone (TRH) immunoreactivity in the brain of the zebrafish (Danio rerio). J Comp Neurol 2002; 450:45-60. [PMID: 12124766 DOI: 10.1002/cne.10300] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The distribution of thyrotropin-releasing hormone (TRH) in the brain of the adult zebrafish was studied with immunohistochemical techniques. In the telencephalon, abundant TRH-immunoreactive (TRHir) neurons were observed in the central, ventral, and supra- and postcommissural regions of the ventral telencephalic area. In the diencephalon, TRHir neurons were observed in the anterior parvocellular preoptic nucleus, the suprachiasmatic nucleus, the lateral hypothalamic nucleus, the rostral parts of the anterior tuberal nucleus and torus lateralis, and the posterior tuberal nucleus. Some TRHir neurons were also observed in the central posterior thalamic nucleus and in the habenula. The mesencephalon contained TRHir cells in the rostrodorsal tegmentum, the Edinger-Westphal nucleus, the torus semicircularis, and the nucleus of the lateral lemniscus. Further TRHir neurons were observed in the interpeduncular nucleus. In the rhombencephalon, TRHir cells were observed in the nucleus isthmi and the locus coeruleus, rostrally, and in the vagal lobe and vagal motor nucleus, caudally. In the forebrain, TRHir fibers were abundant in several regions, including the medial and caudodorsal parts of the dorsal telencephalic area, the ventral and commissural parts of the ventral telencephalic area, the preoptic area, the posterior tubercle, the anterior tuberal nucleus, and the posterior hypothalamic lobe. The dorsal thalamus exhibited moderate TRHir innervation. In the mesencephalon, the optic tectum received a rich TRHir innervation between the periventricular gray zone and the stratum griseum centrale. A conspicuous TRHir longitudinal tract traversed the tegmentum and extended to the rhombencephalon. The medial and lateral mesencephalic reticular areas and the interpeduncular nucleus were richly innervated by TRHir fibers. In the rhombencephalon, the secondary gustatory nucleus received abundant TRHir fibers. TRHir fibers moderately innervated the ventrolateral and ventromedial reticular area and richly innervated the vagal lobe and Cajal's commissural nucleus. Some TRHir fibers coursed in the lateral funiculus of the spinal cord. Some TRHir amacrine cells were observed in the retina. The wide distribution of TRHir neurons and fibers observed in the zebrafish brain suggests that TRH plays different roles. These results in the adult zebrafish reveal a number of differences with respect to the TRHir systems reported in other adult teleosts but were similar to those found during late developmental stages of trout (Díaz et al., 2001).
Collapse
Affiliation(s)
- María Luz Díaz
- Department of Cell and Molecular Biology, Faculty of Sciences, University of A Coruña, 15071 A Coruña, Spain
| | | | | | | |
Collapse
|
47
|
Schmitmeier S, Thole H, Bader A, Bauer K. Purification and characterization of the thyrotropin-releasing hormone (TRH)-degrading serum enzyme and its identification as a product of liver origin. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:1278-86. [PMID: 11856362 DOI: 10.1046/j.1432-1033.2002.02768.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Previous biochemical studies have indicated that the membrane-bound thyrotropin-releasing hormone (TRH)-degrading enzyme (TRH-DE) from brain and liver and the serum TRH-DE are derived from the same gene. These studies also suggested that the serum enzyme is of liver origin. The present study was undertaken to verify these hypotheses. In different species, a close relationship between the activities of the serum enzyme and the particulate liver enzyme was noticed. The activity of the serum enzyme decreased when rats were treated with thioacetamide, a known hepatotoxin. With hepatocytes cultured in a sandwich configuration, release of the TRH-DE into the culture medium could also be demonstrated. The trypsin-solubilized particulate liver TRH-DE and the serum TRH-DE were purified to electrophoretic homogeneity. Both enzymes and the brain TRH-DE were recognized by a monoclonal antibody generated with the purified brain enzyme as antigen. Lectin blot analysis indicated that the serum enzyme and the liver enzyme are glycoproteins containing a sugar structure of the complex type, whereas the brain enzyme exhibits an oligomannose/hybrid glycostructure. A molecular mass of 97 000 Da could be estimated for all three enzymes after deglycosylation and SDS/PAGE followed by Western blotting. Fragment analysis of the serum TRH-DE revealed that the peptide sequences correspond to the cDNA deduced amino-acid sequences of the membrane-bound brain TRH-DE, whereby two peptides were identified that are encoded by exon 1. These data strongly support the hypothesis that the TRH-DEs are all derived from the same gene, whereby the serum enzyme is generated by proteolytic cleavage of the particulate liver enzyme.
Collapse
|
48
|
Schally AV, Comaru-Schally AM, Nagy A, Kovacs M, Szepeshazi K, Plonowski A, Varga JL, Halmos G. Hypothalamic hormones and cancer. Front Neuroendocrinol 2001; 22:248-91. [PMID: 11587553 DOI: 10.1006/frne.2001.0217] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The use of peptide analogs for the therapy of various cancers is reviewed. Inhibition of the pituitary-gonadal axis forms the basis for oncological applications of luteinizing hormone-releasing hormone (LH-RH) agonists and antagonists, but direct effects on tumors may also play a role. Analogs of somatostatin are likewise used for treatment of various tumors. Radiolabeled somatostatin analogs have been successfully applied for the localization of tumors expressing somatostatin receptors. Studies on the role of tumoral LH-RH, growth hormone-releasing hormone (GH-RH), and bombesin/GRP and their receptors in the proliferation of various tumors are summarized, but the complete elucidation of all the mechanisms involved will require much additional work. Human tumors producing hypothalamic hormones are also discussed. Treatment of many cancers remains a major challenge, but new therapeutic modalities are being developed based on antagonists of GH-RH and bombesin, which inhibit growth factors or their receptors. Other approaches consist of the use of cytotoxic analogs of LH-RH, bombesin, and somatostatin, which can be targeted to receptors for these peptides in various cancers and their metastases. These new classes of peptide analogs should lead to a more effective treatment for various cancers.
Collapse
Affiliation(s)
- A V Schally
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, 1601 Perdido Str., New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The role of progesterone (P) in the mechanism of ovulation is controversial at best. The contraceptive application of P was established in rodents in 1936 and with orally absorbed progestogenes was put to human use. There were hints on the proovulatory actions of P administered before the time of ovulation in rats by 1948. Similarly, in 1954 the observation of high P level in the preovulatory follicle pointed to a role in ovulation. Neither of these two observations was further investigated and the positive feedback effect of P exerted on gonadotropins was described in 1968. Still the positive feedback between P and gonadotropins were not recognized as a physiologic mechanism, much less utilized pharmacologically. The apparent contradiction between these two different actions of P continues upto now. The paper sets out to expose this controversy and tries to resolve it using extensive literary data and the author's experiences with synthetic progestogenes in contraception, in the treatment of infertility and with the antigestagen mifepristone in blocking ovulation. The precise mechanisms lying behind these applications are explored and discussed in detail. The putative role of oestradiol (E2) in the mechanism of eliciting the gonadotropin surge is extensively discussed but refuted as the ovulatory signal. The time sequence between the rise of P and gonadotropins contradicts the common wisdom of LH causing luteinization. The positive feedback effect of P on the E2 sensitized ovulatory axis on the hypothalamic and pituitary level is discussed and its local role in the mechanism of follicular rupture is also taken into account. The final proof seems to be the antiovulatory effect of mifepristone, which blocked both GnRH pulsatility, pituitary sensitivity to GnRH and follicular rupture in several experiments. Thus, the dogma of LH peak causing follicular rupture and subsequent luteinization seems questionable, the putative role of E2 to initiate the ovulatory cascade has to be discarded and P's role as a trigger of the physiological mechanisms leading to ovulation should be firmly recognized.
Collapse
Affiliation(s)
- S Zalányi
- MAV-TEK, 6826 Szeged, Csanádi u. 34/a., Hungary
| |
Collapse
|
50
|
Papadopoulos T, Kelly JA, Bauer K. Mutational analysis of the thyrotropin-releasing hormone-degrading ectoenzyme. similarities and differences with other members of the M1 family of aminopeptidases and thermolysin. Biochemistry 2001; 40:9347-55. [PMID: 11478903 DOI: 10.1021/bi010695w] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thyrotropin-releasing hormone-degrading ectoenzyme (TRH-DE) is a TRH-specific peptidase which catalyzes the inactivation of the peptidergic signal substance TRH. As indicated by sequence alignment, TRH-DE and the other members of the M1 family of aminopeptidases have a distinct set of conserved amino acid residues in common. By replacing amino acid residues that are putatively involved in catalysis, we could demonstrate that the enzymatic activities of the mutants E408D, E442D, E464Q, E464D, Y528F, H507R, and H507F are dramatically decreased, essentially due to the changes of V(max). The mutant enzymes E408Q and E442Q are inactive, whereas the specific enzymatic activity of the mutants R488Q, R488A, and Y554F are similar to that of the wild-type enzyme. These data strongly suggest that E408, E442, Y528, and H507 are involved in the catalytic process of TRH-DE while E464 presumably represents the third zinc-coordinating residue and may be equivalent to E166 in thermolysin. In contrast, amino acid residues R488 and Y554 seem not to be involved in the catalytic mechanism of TRH-DE.
Collapse
Affiliation(s)
- T Papadopoulos
- Max-Planck-Institut für Experimentelle Endokrinologie, 30603 Hannover, Germany
| | | | | |
Collapse
|