1
|
Gajbhiye DS, Fernandes GL, Oz I, Nahmias Y, Golan M. A transient neurohormonal circuit controls hatching in fish. Science 2024; 386:1173-1178. [PMID: 39636978 DOI: 10.1126/science.ado8929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/26/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
Hatching is a critical event in the life history of oviparous species. The decision to hatch is often carefully timed to coincide with favorable conditions that will improve survival through early life stages. However, how the relevant cues are relayed to trigger hatching remains unknown. In this work, we show that thyrotropin-releasing hormone (Trh) is the neuroendocrine activator of hatching in zebrafish. To elicit hatching, Trh neurons form a transient circuit that deposits the peptide into the embryo's circulation. Trh also activates hatching in a distantly related fish species that separated more than 200 million years ago. Our results reveal an evolutionarily conserved neuroendocrine circuit that controls a major life event in oviparous fish species.
Collapse
Affiliation(s)
- Deodatta S Gajbhiye
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Genevieve L Fernandes
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Itay Oz
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuni Nahmias
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Matan Golan
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
2
|
Vargas Y, Castro Tron AE, Rodríguez Rodríguez A, Uribe RM, Joseph-Bravo P, Charli JL. Thyrotropin-Releasing Hormone and Food Intake in Mammals: An Update. Metabolites 2024; 14:302. [PMID: 38921437 PMCID: PMC11205479 DOI: 10.3390/metabo14060302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Thyrotropin-releasing hormone (TRH; pGlu-His-Pro-NH2) is an intercellular signal produced mainly by neurons. Among the multiple pharmacological effects of TRH, that on food intake is not well understood. We review studies demonstrating that peripheral injection of TRH generally produces a transient anorexic effect, discuss the pathways that might initiate this effect, and explain its short half-life. In addition, central administration of TRH can produce anorexic or orexigenic effects, depending on the site of injection, that are likely due to interaction with TRH receptor 1. Anorexic effects are most notable when TRH is injected into the hypothalamus and the nucleus accumbens, while the orexigenic effect has only been detected by injection into the brain stem. Functional evidence points to TRH neurons that are prime candidate vectors for TRH action on food intake. These include the caudal raphe nuclei projecting to the dorsal motor nucleus of the vagus, and possibly TRH neurons from the tuberal lateral hypothalamus projecting to the tuberomammillary nuclei. For other TRH neurons, the anatomical or physiological context and impact of TRH in each synaptic domain are still poorly understood. The manipulation of TRH expression in well-defined neuron types will facilitate the discovery of its role in food intake control in each anatomical scene.
Collapse
Affiliation(s)
| | | | | | | | | | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Avenida Universidad 2001, Cuernavaca 62210, Mexico; (Y.V.); (A.E.C.T.); (A.R.R.); (R.M.U.); (P.J.-B.)
| |
Collapse
|
3
|
Trubacova R, Drastichova Z, Novotny J. Biochemical and physiological insights into TRH receptor-mediated signaling. Front Cell Dev Biol 2022; 10:981452. [PMID: 36147745 PMCID: PMC9485831 DOI: 10.3389/fcell.2022.981452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/29/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Thyrotropin-releasing hormone (TRH) is an important endocrine agent that regulates the function of cells in the anterior pituitary and the central and peripheral nervous systems. By controlling the synthesis and release of thyroid hormones, TRH affects many physiological functions, including energy homeostasis. This hormone exerts its effects through G protein-coupled TRH receptors, which signal primarily through Gq/11 but may also utilize other G protein classes under certain conditions. Because of the potential therapeutic benefit, considerable attention has been devoted to the synthesis of new TRH analogs that may have some advantageous properties compared with TRH. In this context, it may be interesting to consider the phenomenon of biased agonism and signaling at the TRH receptor. This possibility is supported by some recent findings. Although knowledge about the mechanisms of TRH receptor-mediated signaling has increased steadily over the past decades, there are still many unanswered questions, particularly about the molecular details of post-receptor signaling. In this review, we summarize what has been learned to date about TRH receptor-mediated signaling, including some previously undiscussed information, and point to future directions in TRH research that may offer new insights into the molecular mechanisms of TRH receptor-triggered actions and possible ways to modulate TRH receptor-mediated signaling.
Collapse
|
4
|
Yang F, Zhang H, Meng X, Li Y, Zhou Y, Ling S, Sun D, Lv P, Liu L, Shi P, Tian C. Structural insights into thyrotropin-releasing hormone receptor activation by an endogenous peptide agonist or its orally administered analogue. Cell Res 2022; 32:858-861. [PMID: 35352031 PMCID: PMC9437008 DOI: 10.1038/s41422-022-00646-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/12/2021] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Fan Yang
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Huanhuan Zhang
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianyu Meng
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Yingge Li
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Yingxin Zhou
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Shenglong Ling
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Demeng Sun
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Pei Lv
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Liu
- Tsinghua-Peking Joint Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China.
| | - Pan Shi
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China.
| | - Changlin Tian
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China.
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China.
| |
Collapse
|
5
|
β-Arrestin2 Is Critically Involved in the Differential Regulation of Phosphosignaling Pathways by Thyrotropin-Releasing Hormone and Taltirelin. Cells 2022; 11:cells11091473. [PMID: 35563779 PMCID: PMC9103620 DOI: 10.3390/cells11091473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
In recent years, thyrotropin-releasing hormone (TRH) and its analogs, including taltirelin (TAL), have demonstrated a range of effects on the central nervous system that represent potential therapeutic agents for the treatment of various neurological disorders, including neurodegenerative diseases. However, the molecular mechanisms of their actions remain poorly understood. In this study, we investigated phosphosignaling dynamics in pituitary GH1 cells affected by TRH and TAL and the putative role of β-arrestin2 in mediating these effects. Our results revealed widespread alterations in many phosphosignaling pathways involving signal transduction via small GTPases, MAP kinases, Ser/Thr- and Tyr-protein kinases, Wnt/β-catenin, and members of the Hippo pathway. The differential TRH- or TAL-induced phosphorylation of numerous proteins suggests that these ligands exhibit some degree of biased agonism at the TRH receptor. The different phosphorylation patterns induced by TRH or TAL in β-arrestin2-deficient cells suggest that the β-arrestin2 scaffold is a key factor determining phosphorylation events after TRH receptor activation. Our results suggest that compounds that modulate kinase and phosphatase activity can be considered as additional adjuvants to enhance the potential therapeutic value of TRH or TAL.
Collapse
|
6
|
Prokai-Tatrai K, Nguyen V, Prokai L. [β-Glu 2]TRH Is a Functional Antagonist of Thyrotropin-Releasing Hormone (TRH) in the Rodent Brain. Int J Mol Sci 2021; 22:ijms22126230. [PMID: 34207724 PMCID: PMC8226542 DOI: 10.3390/ijms22126230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/14/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
Selective antagonists of thyrotropin-releasing hormone (TRH; pGlu-His-Pro-NH2), in order to enable a better understanding of this peptide’s central functions, have not been identified. Using pGlu-Glu-Pro-NH2 ([Glu2]TRH) as a lead peptide and with modification at its central residue, our studies focused on some of its analogues synthesized as potential functional antagonists of TRH in the rodent brain. Among the peptides studied, the novel isomeric analogue [β-Glu2]TRH was found to suppress the analeptic and antidepressant-like pharmacological activities of TRH without eliciting intrinsic effects in these paradigms. [β-Glu2]TRH also completely reversed TRH’s stimulation of acetylcholine turnover in the rat hippocampus without a cholinergic activity of its own, which was demonstrated through in vivo microdialysis experiments. Altogether, [β-Glu2]TRH emerged as the first selective functional antagonist of TRH’s prominent cholinergic actions, by which this endogenous peptide elicits a vast array of central effects.
Collapse
|
7
|
Liu WY, Liu H, Aggarwal J, Huang ZL, Horner RL. Differential activating effects of thyrotropin-releasing hormone and its analog taltirelin on motor output to the tongue musculature in vivo. Sleep 2021; 43:5813557. [PMID: 32227104 PMCID: PMC7487885 DOI: 10.1093/sleep/zsaa053] [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] [Academic Contribution Register] [Received: 11/27/2019] [Revised: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Thyrotropin-releasing hormone (TRH) is produced by the hypothalamus but most brain TRH is located elsewhere where it acts as a neuromodulator. TRH-positive neurons project to the hypoglossal motoneuron pool where TRH receptor RNA shows a high degree of differential expression compared with the rest of the brain. Strategies to modulate hypoglossal motor activity are of physiological and clinical interest given the potential for pharmacotherapy for obstructive sleep apnea (OSA), a common and serious respiratory disorder. Here, we identified the effects on tongue motor activity of TRH and a specific analog (taltirelin) applied locally to the hypoglossal motoneuron pool and systemically in vivo. Studies were performed under isoflurane anesthesia and across sleep–wake states in rats. In anesthetized rats, microperfusion of TRH (n = 8) or taltirelin (n = 9) into the hypoglossal motoneuron pool caused dose-dependent increases in tonic and phasic tongue motor activity (both p < 0.001). However, the motor responses to TRH were biphasic, being significantly larger “early” in the response versus at the end of the intervention (p ≤ 0.022). In contrast, responses to taltirelin were similar “early” versus “late” (p ≥ 0.107); i.e. once elicited, the motor responses to taltirelin were sustained and maintained. In freely behaving conscious rats (n = 10), microperfusion of 10 μM taltirelin into the hypoglossal motoneuron pool increased tonic and phasic tongue motor activity in non-rapid-eye-movement (REM) sleep (p ≤ 0.038). Intraperitoneal injection of taltirelin (1 mg/kg, n = 16 rats) also increased tonic tongue motor activity across sleep–wake states (p = 0.010). These findings inform the studies in humans to identify the potential beneficial effects of taltirelin for breathing during sleep and OSA.
Collapse
Affiliation(s)
- Wen-Ying Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmacology, Institute of Brain Science, School of Basic Medical Science, Fudan University, Shanghai, China.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Hattie Liu
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jasmin Aggarwal
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Zhi-Li Huang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmacology, Institute of Brain Science, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Richard L Horner
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Charli JL, Rodríguez-Rodríguez A, Hernández-Ortega K, Cote-Vélez A, Uribe RM, Jaimes-Hoy L, Joseph-Bravo P. The Thyrotropin-Releasing Hormone-Degrading Ectoenzyme, a Therapeutic Target? Front Pharmacol 2020; 11:640. [PMID: 32457627 PMCID: PMC7225337 DOI: 10.3389/fphar.2020.00640] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/04/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
Thyrotropin releasing hormone (TRH: Glp-His-Pro-NH2) is a peptide mainly produced by brain neurons. In mammals, hypophysiotropic TRH neurons of the paraventricular nucleus of the hypothalamus integrate metabolic information and drive the secretion of thyrotropin from the anterior pituitary, and thus the activity of the thyroid axis. Other hypothalamic or extrahypothalamic TRH neurons have less understood functions although pharmacological studies have shown that TRH has multiple central effects, such as promoting arousal, anorexia and anxiolysis, as well as controlling gastric, cardiac and respiratory autonomic functions. Two G-protein-coupled TRH receptors (TRH-R1 and TRH-R2) transduce TRH effects in some mammals although humans lack TRH-R2. TRH effects are of short duration, in part because the peptide is hydrolyzed in blood and extracellular space by a M1 family metallopeptidase, the TRH-degrading ectoenzyme (TRH-DE), also called pyroglutamyl peptidase II. TRH-DE is enriched in various brain regions but is also expressed in peripheral tissues including the anterior pituitary and the liver, which secretes a soluble form into blood. Among the M1 metallopeptidases, TRH-DE is the only member with a very narrow specificity; its best characterized biological substrate is TRH, making it a target for the specific manipulation of TRH activity. Two other substrates of TRH-DE, Glp-Phe-Pro-NH2 and Glp-Tyr-Pro-NH2, are also present in many tissues. Analogs of TRH resistant to hydrolysis by TRH-DE have prolonged central efficiency. Structure-activity studies allowed the identification of residues critical for activity and specificity. Research with specific inhibitors has confirmed that TRH-DE controls TRH actions. TRH-DE expression by β2-tanycytes of the median eminence of the hypothalamus allows the control of TRH flux into the hypothalamus-pituitary portal vessels and may regulate serum thyrotropin secretion. In this review we describe the critical evidences that suggest that modification of TRH-DE activity in tanycytes, and/or in other brain regions, may generate beneficial consequences in some central and metabolic disorders and identify potential drawbacks and missing information needed to test these hypotheses.
Collapse
Affiliation(s)
- Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| | | | | | | | | | | | | |
Collapse
|
9
|
Evaluation of the effects of chemotherapy-induced fatigue and pharmacological interventions in multiple mouse behavioral assays. Behav Brain Res 2018; 360:255-261. [PMID: 30529403 DOI: 10.1016/j.bbr.2018.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/09/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
Fatigue is a common symptom in many diseases and disorders and can reduce quality of life, yet lacks an adequate pharmacological intervention. To identify and develop such interventions, and to better understand fatigue, additional preclinical research is necessary. However, despite numerous mouse behavioral assays reportedly detecting fatigue-like behavior, the assumption that fatigue-like behavior is detected in many assays has not been validated through a cross-assay study. Thus, we modeled fatigue in mice by administering 5-fluorouracil, a chemotherapy drug known to cause fatigue in humans and fatigue-like behavior in mice, then evaluated its effects via voluntary wheel running activity (VWRA), locomotor activity in the open field test (OFT), immobility in the forced swim test (FST), and distance run in the treadmill fatigue test (TFT) and treadmill exercise capacity test. Additionally, taltirelin or methylphenidate was administered to alleviate fatigue-like behavior. As a result of 5-fluorouracil treatment, VWRA and the TFT were markedly reduced, indicating fatigue. The OFT, FST, and treadmill exercise capacity test, however, failed to detect fatigue-like behavior. Interestingly, both taltirelin and methylphenidate alleviated fatigue-like behavior in TFT. These data suggest that, of the current assays, only the TFT and VWRA should be expected to detect fatigue-like behavior. Moreover, this study provides additional evidence that taltirelin may provide a novel treatment for chemotherapy-induced fatigue and warrants further evaluation as an anti-fatigue therapeutic.
Collapse
|
10
|
Zheng C, Chen G, Tan Y, Zeng W, Peng Q, Wang J, Cheng C, Yang X, Nie S, Xu Y, Zhang Z, Papa SM, Ye K, Cao X. TRH Analog, Taltirelin Improves Motor Function of Hemi-PD Rats Without Inducing Dyskinesia via Sustained Dopamine Stimulating Effect. Front Cell Neurosci 2018; 12:417. [PMID: 30555300 PMCID: PMC6282053 DOI: 10.3389/fncel.2018.00417] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2018] [Accepted: 10/25/2018] [Indexed: 01/08/2023] Open
Abstract
Thyrotropin-releasing hormone (TRH) and its analogs are able to stimulate the release of the endogenic dopamine (DA) in the central nervous system. However, this effect has not been tested in the Parkinson’s disease (PD), which is characterized by the DA deficiency due to the dopaminergic neurons loss in the substantia nigra. Here, we investigated the therapeutic effect of Taltirelin, a long-acting TRH analog on 6-hydroxydopamine-lesioned hemi-Parkinsonian rat model. 1–10 mg/kg Taltirelin i.p. administration significantly improved the locomotor function and halted the electrophysiological abnormities of PD animals without inducing dyskinesia even with high-dose for 7 days treatment. Microdialysis showed that Taltirelin gently and persistently promoted DA release in the cortex and striatum, while L-DOPA induced a sharp rise of DA especially in the cortex. The DA-releasing effect of Taltirelin was alleviated by reserpine, vanoxerine (GBR12909) or AMPT, indicating a mechanism involving vesicular monoamine transporter-2 (VMAT-2), dopamine transporter (DAT) and tyrosine hydroxylase (TH). The in vivo and in vitro experiments further supported that Taltirelin affected the regulation of TH expression in striatal neurons, which was mediated by p-ERK1/2. Together, this study demonstrated that Taltirelin improved motor function of hemi-PD rats without inducing dyskinesia, thus supporting a further exploration of Taltirelin for PD treatment.
Collapse
Affiliation(s)
- Cong Zheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guiqin Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Tan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiqi Zeng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiwei Peng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chi Cheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuke Nie
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, United States.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Kobayashi N, Sato N, Fujimura Y, Kihara T, Sugita K, Takahashi K, Koike K, Sugawara T, Tada Y, Nakai H, Yoshikawa T. Discovery of the Orally Effective Thyrotropin-Releasing Hormone Mimetic: 1-{ N-[(4 S,5 S)-(5-Methyl-2-oxooxazolidine-4-yl)carbonyl]-3-(thiazol-4-yl)-l-alanyl}-(2 R)-2-methylpyrrolidine Trihydrate (Rovatirelin Hydrate). ACS OMEGA 2018; 3:13647-13666. [PMID: 30411045 PMCID: PMC6217654 DOI: 10.1021/acsomega.8b01481] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/02/2018] [Accepted: 09/25/2018] [Indexed: 06/01/2023]
Abstract
We have explored orally effective thyrotropin-releasing hormone (TRH) mimetics, showing oral bioavailability and brain penetration by structure-activity relationship (SAR) study on the basis of in vivo antagonistic activity on reserpine-induced hypothermia in mice. By primary screening of the synthesized TRH mimetics, we found a novel TRH mimetic: l-pyroglutamyl-[3-(thiazol-4-yl)-l-alanyl]-l-prolinamide with a high central nervous system effect compared with TRH as a lead compound. Further SAR optimization studies of this lead compound led to discovery of a novel orally effective TRH mimetic: 1-{N-[(4S,5S)-(5-methyl-2-oxooxazolidine-4-yl)carbonyl]-3-(thiazol-4-yl)-l-alanyl}-(2R)-2-methylpyrrolidine trihydrate (rovatirelin hydrate), which was selected as a candidate for clinical trials.
Collapse
Affiliation(s)
- Naotake Kobayashi
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Norihito Sato
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Yuko Fujimura
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Tsuyoshi Kihara
- Business
Search & Evaluation, Shionogi &
Co., Ltd., 3-1-8, Doshomachi, Chuo-ku, Osaka-shi, Osaka 541-0045, Japan
| | - Katsuji Sugita
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Kouji Takahashi
- DMPK
Services, Shionogi Techno Advance Research
Co., Ltd., 3-1-1, Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Katsumi Koike
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Tamio Sugawara
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Yukio Tada
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Hiroshi Nakai
- Medicinal
Chemistry Research Laboratory, Research Laboratory for Development, and Drug Discovery
& Disease Research Laboratory, Shionogi
& Co., Ltd., 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Takayoshi Yoshikawa
- Pharmacovigilance
Japan, Allergan Japan K.K., 4-20-3-35, Ebisu, Shibuya-ku, Tokyo 150-6035, Japan
| |
Collapse
|
12
|
Zhang Y, Wang C, Zhang L. The potential role of thyrotropin-releasing hormone in colonic dysmotility induced by water avoidance stress in rats. Neuropeptides 2018; 70:47-54. [PMID: 29803395 DOI: 10.1016/j.npep.2018.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/22/2017] [Revised: 05/13/2018] [Accepted: 05/13/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study sought to investigate the effect and underlying mechanism of thyrotrophin releasing hormone (TRH) on colonic contractile disorders induced by chronic water avoidance stress (WAS). METHODS Male SD rats were exposed to daily 1-h WAS or sham WAS for 10 consecutive days. The presence of TRH in the serum and colonic mucosa were determined using enzyme immunoassay kits. Immunohistochemistry and western blotting were performed to detect the expression of TRH receptor 1 (TRH-R1). The contractions of proximal colonic smooth muscle were studied in an organ bath system. The whole-cell patch-clamp technique was used to record the currents of both L-type calcium currents (ICa,L) and large conductance Ca2+-activated K+ (BKCa) channels in colonic smooth muscle cells (SMCs) isolated from adult rats. RESULTS Enzyme immunoassay revealed that TRH was present in both serum and colonic mucosa and that this expression increased in the WAS group. Immunohistochemistry revealed that the TRH-R1 level increased in colons devoid of mucosa and submucosa from the stressed rats as compared with the control group. TRH increased the spontaneous contractions of the longitudinal muscle and circular muscle strips in a dose-dependent manner in vitro. The effect was also confirmed in an vivo experiment, where an intraperitoneal injection of TRH in rats significantly increased fecal pellet output during a 24-h period as compared with the control group. Furthermore, intraperitoneal injection of a non-specific TRH receptor antagonist, chlordiazepoxide and a TRH-R1 antibody, partially decreased the fecal pellets of WAS rats during the 10-day stress period. Furthermore, TRH increased the peak current of L-type channels in colonic smooth muscle cells (SMCs) at a membrane potential of 0 mV, while the current of large conductance Ca2+-activated K+ (BKCa) channels was not changed following the addition of TRH. CONCLUSION TRH may be involved in the dysmotility induced by chronic stress and may have some potential clinical therapeutic use in regulating gut motility.
Collapse
Affiliation(s)
- Yanzhen Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Chunfeng Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lianfeng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
13
|
Boghosian JD, Luethy A, Cotten JF. Intravenous and Intratracheal Thyrotropin Releasing Hormone and Its Analog Taltirelin Reverse Opioid-Induced Respiratory Depression in Isoflurane Anesthetized Rats. J Pharmacol Exp Ther 2018; 366:105-112. [PMID: 29674333 DOI: 10.1124/jpet.118.248377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2018] [Accepted: 04/18/2018] [Indexed: 01/14/2023] Open
Abstract
Thyrotropin releasing hormone (TRH) is a tripeptide hormone and a neurotransmitter widely expressed in the central nervous system that regulates thyroid function and maintains physiologic homeostasis. Following injection in rodents, TRH has multiple effects including increased blood pressure and breathing. We tested the hypothesis that TRH and its long-acting analog, taltirelin, will reverse morphine-induced respiratory depression in anesthetized rats following intravenous or intratracheal (IT) administration. TRH (1 mg/kg plus 5 mg/kg/h, i.v.) and talitrelin (1 mg/kg, i.v.), when administered to rats pretreated with morphine (5 mg/kg, i.v.), increased ventilation from 50% ± 6% to 131% ± 7% and 45% ± 6% to 168% ± 13%, respectively (percent baseline; n = 4 ± S.E.M.), primarily through increased breathing rates (from 76% ± 9% to 260% ± 14% and 66% ± 8% to 318% ± 37%, respectively). By arterial blood gas analysis, morphine caused a hypoxemic respiratory acidosis with decreased oxygen and increased carbon dioxide pressures. TRH decreased morphine effects on arterial carbon dioxide pressure, but failed to impact oxygenation; taltirelin reversed morphine effects on both arterial carbon dioxide and oxygen. Both TRH and talirelin increased mean arterial blood pressure in morphine-treated rats (from 68% ± 5% to 126% ± 12% and 64% ± 7% to 116% ± 8%, respectively; n = 3 to 4). TRH, when initiated prior to morphine (15 mg/kg, i.v.), prevented morphine-induced changes in ventilation; and TRH (2 mg/kg, i.v.) rescued all four rats treated with a lethal dose of morphine (5 mg/kg/min, until apnea). Similar to intravenous administration, both TRH (5 mg/kg, IT) and taltirelin (2 mg/kg, IT) reversed morphine effects on ventilation. TRH or taltirelin may have clinical utility as an intravenous or inhaled agent to antagonize opioid-induced cardiorespiratory depression.
Collapse
Affiliation(s)
- James D Boghosian
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.D.B., A.L., J.F.C.); and Department of Anesthesia, Kantonsspital Aarau, Aarau, Switzerland (A.L.)
| | - Anita Luethy
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.D.B., A.L., J.F.C.); and Department of Anesthesia, Kantonsspital Aarau, Aarau, Switzerland (A.L.)
| | - Joseph F Cotten
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts (J.D.B., A.L., J.F.C.); and Department of Anesthesia, Kantonsspital Aarau, Aarau, Switzerland (A.L.)
| |
Collapse
|
14
|
Swarnalatha NB, Roy N, Gouda MM, Moger R, Abraham A. High-fat, simple-carbohydrate diet intake induces hypothalamic-pituitary-thyroid axis dysregulation in C57BL/6J male mice. Appl Physiol Nutr Metab 2017; 43:371-380. [PMID: 29099999 DOI: 10.1139/apnm-2017-0410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022]
Abstract
Given the association between subclinical hypothyroidism and metabolic syndrome, we wanted to explore if high-fat, simple-carbohydrate (HFSC) diet affects hypothalamus-pituitary-thyroid axis. One-month-old male C57BL/6J mice were fed with control (C) and HFSC (T) feed (n = 18 each), respectively, for 5 months. There was a significant increase in triiodothyronine in the T group (13.5%) compared with the age-matched C group by the fifth month. Thyroid-stimulating hormone was significantly higher (1 month: 1.9-fold; 3 months: 2.66-fold; 5 months: 3.5-fold) from the first to fifth months in the T group compared with age-matched C group. Thyrotropin-releasing hormone (TRH) gene expression showed significant decrease (1 month: 83.2%; 5 months: 40.7%) in the T group compared with the age-matched C group. TRHR1 showed significant decrease in the T group compared with the age-matched C group throughout the study (1 month: 82.8%; 3 months: 45.7%; 5 months: 75.2%). However, TRHR2 showed dynamic change during the study. Initially there was significant (1 month: 0.104-fold) downregulation, followed by significant upregulation (3 months: 3.6-fold) and downregulation (0.73-fold) by the fifth month in the T group compared with the age-matched C group. There was marked depletion of functional follicular cells and colloid substance in the thyroid glands of the T group by the fifth month compared with the C group. Leptin receptors ObRa (1 month: 48.25%; 5 months: 88%) and ObRb (1 month: 46.9%; 5 months: 63.3%) were significantly downregulated in the T group compared with the age-matched C group in the first and fifth months of feeding the respective diets. The expression of p-STAT3, a transcription factor known to have a role in energy balance, intermediate metabolism, and leptin signalling was seen to decrease significantly (6.25-fold) in the hypothalamus of the T group compared with the age-matched C group. In conclusion, HFSC feed disrupts the hypothalamus-pituitary-thyroid axis in male C57BL/6J mice.
Collapse
Affiliation(s)
- Nagaraj Banavara Swarnalatha
- a Father George Albuquerque Pai Cell and Molecular Biology Laboratory, Department of Postgraduate Studies and Research in Biotechnology, St Aloysius College (Autonomous), Mangaluru-575003, India.,b PG Department of Biochemistry, St Aloysius College (Autonomous), Mangaluru-575003, India
| | - Neena Roy
- a Father George Albuquerque Pai Cell and Molecular Biology Laboratory, Department of Postgraduate Studies and Research in Biotechnology, St Aloysius College (Autonomous), Mangaluru-575003, India
| | | | - Rajeish Moger
- d Department Fisheries Microbiology, College of Fisheries, Mangaluru-575002, India
| | - Asha Abraham
- a Father George Albuquerque Pai Cell and Molecular Biology Laboratory, Department of Postgraduate Studies and Research in Biotechnology, St Aloysius College (Autonomous), Mangaluru-575003, India
| |
Collapse
|
15
|
Tanycytes control the hormonal output of the hypothalamic-pituitary-thyroid axis. Nat Commun 2017; 8:484. [PMID: 28883467 PMCID: PMC5589884 DOI: 10.1038/s41467-017-00604-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/13/2017] [Accepted: 07/13/2017] [Indexed: 12/29/2022] Open
Abstract
The hypothalamic–pituitary–thyroid (HPT) axis maintains circulating thyroid hormone levels in a narrow physiological range. As axons containing thyrotropin-releasing hormone (TRH) terminate on hypothalamic tanycytes, these specialized glial cells have been suggested to influence the activity of the HPT axis, but their exact role remained enigmatic. Here, we demonstrate that stimulation of the TRH receptor 1 increases intracellular calcium in tanycytes of the median eminence via Gαq/11 proteins. Activation of Gαq/11 pathways increases the size of tanycyte endfeet that shield pituitary vessels and induces the activity of the TRH-degrading ectoenzyme. Both mechanisms may limit the TRH release to the pituitary. Indeed, blocking TRH signaling in tanycytes by deleting Gαq/11 proteins in vivo enhances the response of the HPT axis to the chemogenetic activation of TRH neurons. In conclusion, we identify new TRH- and Gαq/11-dependent mechanisms in the median eminence by which tanycytes control the activity of the HPT axis. The hypothalamic-pituitary-thyroid (HPT) axis regulates a wide range of physiological processes. Here the authors show that hypothalamic tanycytes play a role in the homeostatic regulation of the HPT axis; activation of TRH signaling in tanycytes elevates their intracellular Ca2+ via Gαq/11 pathway, ultimately resulting in reduced TRH release into the pituitary vessels.
Collapse
|
16
|
Dougherty JP, Wolff BS, Cullen MJ, Saligan LN, Gershengorn MC. Taltirelin alleviates fatigue-like behavior in mouse models of cancer-related fatigue. Pharmacol Res 2017; 124:1-8. [PMID: 28720519 DOI: 10.1016/j.phrs.2017.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/09/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 02/08/2023]
Abstract
Fatigue affects most cancer patients and has numerous potential causes, including cancer itself and cancer treatment. Cancer-related fatigue (CRF) is not relieved by rest, can decrease quality of life, and has no FDA-approved therapy. Thyrotropin-releasing hormone (TRH) has been proposed as a potential novel treatment for CRF, but its efficacy against CRF remains largely untested. Thus, we tested the TRH analog, taltirelin (TAL), in mouse models of CRF. To model fatigue, we used a mouse model of chemotherapy, a mouse model of radiation therapy, and mice bearing colon 26 carcinoma tumors. We used the treadmill fatigue test to assess fatigue-like behavior after treatment with TAL. Additionally, we used wild-type and TRH receptor knockout mice to determine which TRH receptor was necessary for the actions of TAL. Tumor-bearing mice displayed muscle wasting and all models caused fatigue-like behavior, with mice running a shorter distance in the treadmill fatigue test than controls. TAL reversed fatigue-like behavior in all three models and the mouse TRH1 receptor was necessary for the effects of TAL. These data suggest that TAL may be useful in alleviating fatigue in all cancer patients and provide further support for evaluating TAL as a potential therapy for CRF in humans.
Collapse
Affiliation(s)
- John P Dougherty
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Brian S Wolff
- National Institute of Nursing Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Mary J Cullen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Leorey N Saligan
- National Institute of Nursing Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA,.
| | - Marvin C Gershengorn
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
17
|
Aguirre TAS, Teijeiro-Osorio D, Rosa M, Coulter IS, Alonso MJ, Brayden DJ. Current status of selected oral peptide technologies in advanced preclinical development and in clinical trials. Adv Drug Deliv Rev 2016; 106:223-241. [PMID: 26921819 DOI: 10.1016/j.addr.2016.02.004] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2016] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 01/12/2023]
Abstract
The development of oral dosage forms that allows absorption of therapeutic peptides to the systemic circulation is one of the greatest challenges for the pharmaceutical industry. Currently, a number of technologies including either mixtures of penetration enhancers or protease inhibitors and/or nanotechnology-based products are under clinical development. Typically, these formulations are presented in the form of enteric-coated tablets or capsules. Systems undergoing preclinical investigation include further advances in nanotechnology, including intestinal microneedle patches, as well as their combination with regional delivery to the colon. This review critically examines four selected promising oral peptide technologies at preclinical stage and the twelve that have progressed to clinical trials, as indicated in www.clinicaltrials.gov. We examined these technologies under the criteria of peptide selection, formulation design, system components and excipients, intestinal mechanism of action, efficacy in man, and safety issues. The conclusion is that most of the technologies in clinical trials are incremental rather than paradigm-shifting and that even the more clinically advanced oral peptide drugs examples of oral bioavailability appear to yield oral bioavailability values of only 1-2% and are, therefore, only currently suitable for a limited range of peptides.
Collapse
Affiliation(s)
- T A S Aguirre
- Centro de Ciências Exatas e Tecnologia, Universidade de Caxias do Sul (UCS), Caxias do Sul, Brazil
| | - D Teijeiro-Osorio
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - M Rosa
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - I S Coulter
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - M J Alonso
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - D J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
18
|
Effect of L-pGlu-(1-benzyl)-l-His-l-Pro-NH 2 against in-vitro and in-vivo models of cerebral ischemia and associated neurological disorders. Biomed Pharmacother 2016; 84:1256-1265. [PMID: 27810782 DOI: 10.1016/j.biopha.2016.10.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 01/28/2023] Open
Abstract
Central nervous system plays a vital role in regulation of most of biological functions which are abnormally affected in various disorders including cerebral ischemia, Alzheimer's and Parkinson's (AD and PD) worldwide. Cerebral stroke is an extremely fatal and one of the least comprehensible neurological disorders due to limited availability of prospective clinical approaches and therapeutics. Since, some endogenous peptides like thyrotropin-releasing hormone have shown substantial neuroprotective potential, hence present study evaluates the newer thyrotropin-releasing hormone (TRH) analogue L-pGlu-(1-benzyl)-l-His-l-Pro-NH2 for its neuroprotective effects against oxygen glucose deprivation (OGD), glutamate and H2O2 induced injury in pheochromocytoma cell lines (PC-12 cells) and in-vivo ischemic injury in mice. Additionally, the treatment was further analyzed with respect to models of AD and PD in mice. Cerebral ischemia was induced by clamping both bilateral common carotid arteries for ten minutes. Treatment was administered to the mice five minute after restoration of blood supply to brain. Consequential changes in neurobehavioural, biochemical and histological parameters were assessed after a week. L-pGlu-(1-benzyl)-l-His-l-Pro-NH2 showed significant reduction in glutamate, H2O2 and OGD -induced cell death in concentration and time dependent manner. Moreover, L-pGlu-(1-benzyl)-l-His-l-Pro-NH2 resulted in a substantial reduction in CA1 (Cornus Ammonis 1) hippocampal neuronal cell death, inflammatory cytokines, TNF-α, IL-6 and oxidative stress in hippocampus. In addition, L-pGlu-(1-benzyl)-l-His-l-Pro-NH2 was found to be protective in two acute models of AD and PD as well these findings demonstrate the neuroprotective potential of L-pGlu-(1-benzyl)-l-His-l-Pro-NH2 in cerebral ischemia and other diseases, which may be mediated through reduction of excitotoxicity, oxidative stress and inflammation.
Collapse
|
19
|
Meena CL, Thakur A, Nandekar PP, Sharma SS, Sangamwar AT, Jain R. Synthesis and biology of ring-modified l-Histidine containing thyrotropin-releasing hormone (TRH) analogues. Eur J Med Chem 2016; 111:72-83. [PMID: 26854379 DOI: 10.1016/j.ejmech.2016.01.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 12/18/2022]
Abstract
Thyrotropin-releasing hormone (TRH) analogues bearing halogen groups (Cl, Br and I) at the C-2 and/or C-5 position, and the alkyl group (CH3, C2H5, C3H7, CH2C6H5) at the N-1 position of the imidazole ring of the central histidine residue were synthesized and evaluated for the receptor binding, calcium mobilization (FLIPR), and IP-1 assay at the HEK mTRHR1 and HEK mTRHR2 expressing cell lines. The most promising analogue 7k showed 925-fold selectivity for HEK mTRH-R2 receptor subtype in the IP-1 assay, 272-fold selectivity for HEK mTRH-R2 receptor subtype in the FLIPR assay, and 21-fold receptor binding specificity at HEK TRH-R2 receptor subtype. The peptide 7k was evaluated in vitro in a brain membrane competitive binding assay, and for stability analysis in the presence of TRH-DE, in vivo. The analogue 7k showed decrease in the sleeping time by more than 76% in a pentobarbital-induced sleeping assay, and showed comparatively less elevation in the TSH level in the blood, in vivo. The computational homology modeling of TRH-R1 and TRH-R2 and docking study with the most potent peptide 7k provide impetus to design CNS specific TRH analogues.
Collapse
Affiliation(s)
- Chhuttan L Meena
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Avinash Thakur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Prajwal P Nandekar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Abhay T Sangamwar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India.
| |
Collapse
|
20
|
Meena CL, Ingole S, Rajpoot S, Thakur A, Nandeker PP, Sangamwar AT, Sharma SS, Jain R. Discovery of a low affinity thyrotropin-releasing hormone (TRH)-like peptide that exhibits potent inhibition of scopolamine-induced memory impairment in mice. RSC Adv 2015; 5:56872-56884. [PMID: 26191403 PMCID: PMC4501038 DOI: 10.1039/c5ra06935a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
TRH-like peptides were synthesized in which the critical N-terminus residue L-pGlu was replaced with various heteroaromatic rings, and the central residue histidine with 1-alkyl-L-histidines. All synthesized TRH-like peptides were evaluated in vitro as agonists in HEK mTRH-R1 and HEK mTRH-R2 cell lines, an expressing receptor binding assay (IC50), and cell signaling assay (EC50). The analeptic potential of the synthesized peptides was evaluated in vivo by using the antagonism of a pentobarbital-induced sleeping time. The peptides 6a, 6c and 6e were found to activate TRH-R2 with potencies (EC50) of 0.002 μM, 0.28 μM and 0.049 μM, respectively. In contrast, for signaling activation of TRH-R1, the same peptides required higher concentration of 0.414 μM, 50 μM and 19.1 μM, respectively in the FLIPR assay. The results showed that these peptides were 207, 178 and 389-fold selective towards TRH-R2 receptor subtype. In the antagonism of a pentobarbital-induced sleeping time assay, peptide 6c showed a 58.5% reduction in sleeping time. The peptide 6c exhibited high stability in rat blood plasma, a superior effect on the scopolamine-induced cognition impairment mice model, safe effects on the cardiovascular system, and general behavior using a functional observation battery (FOB).
Collapse
Affiliation(s)
- Chhuttan L. Meena
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Shubdha Ingole
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Satyendra Rajpoot
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Avinash Thakur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Prajwal P. Nandeker
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Abhay T. Sangamwar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Shyam S. Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, 160 062, Punjab, India
| |
Collapse
|
21
|
Abstract
The treatment options currently available for narcolepsy are often unsatisfactory due to suboptimal efficacy, troublesome side effects, development of drug tolerance, and inconvenience. Our understanding of the neurobiology of narcolepsy has greatly improved over the last decade. This knowledge has not yet translated into additional therapeutic options for patients, but progress is being made. Some compounds, such as histaminergic H3 receptor antagonists, may prove useful in symptom control of narcolepsy. The prospect of finding a cure still seems distant, but hypocretin replacement therapy offers some promise. In this narrative review, we describe these developments and others which may yield more effective narcolepsy treatments in the future.
Collapse
Affiliation(s)
- Dariusz R Wozniak
- Respiratory Support and Sleep Centre, Papworth Hospital, Cambridge, UK
| | | |
Collapse
|
22
|
First-in-class thyrotropin-releasing hormone (TRH)-based compound binds to a pharmacologically distinct TRH receptor subtype in human brain and is effective in neurodegenerative models. Neuropharmacology 2014; 89:193-203. [PMID: 25281210 DOI: 10.1016/j.neuropharm.2014.09.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/14/2014] [Revised: 08/26/2014] [Accepted: 09/18/2014] [Indexed: 12/13/2022]
Abstract
JAK4D, a first-in-class thyrotropin-releasing hormone (TRH)-based compound, is a prospective therapeutic candidate offering a multifaceted approach to treating neurodegeneration and other CNS conditions. The purpose of these studies was to determine the ability of JAK4D to bind to TRH receptors in human brain and to evaluate its neuropharmacological effects in neurodegenerative animal models. Additionally, JAK4D brain permeation was examined in mouse, and initial toxicology was assessed in vivo and in vitro. We report that JAK4D bound selectively with nanomolar affinity to native TRH receptors in human hippocampal tissue and showed for the first time that these receptors are pharmacologically distinct from TRH receptors in human pituitary, thus revealing a new TRH receptor subtype which represents a promising neurotherapeutic target in human brain. Systemic administration of JAK4D elicited statistically significant and clinically-relevant neuroprotective effects in three established neurodegenerative animal models: JAK4D reduced cognitive deficits when administered post-insult in a kainate (KA)-induced rat model of neurodegeneration; it protected against free radical release and neuronal damage evoked by intrastriatal microdialysis of KA in rat; and it reduced motor decline, weight loss, and lumbar spinal cord neuronal loss in G93A-SOD1 transgenic Amyotrophic Lateral Sclerosis mice. Ability to cross the blood-brain barrier and a clean initial toxicology profile were also shown. In light of these findings, JAK4D is an important tool for investigating the hitherto-unidentified central TRH receptor subtype reported herein and an attractive therapeutic candidate for neurodegenerative disorders.
Collapse
|
23
|
Rodríguez-Molina V, Patiño J, Vargas Y, Sánchez-Jaramillo E, Joseph-Bravo P, Charli JL. TRH regulates action potential shape in cerebral cortex pyramidal neurons. Brain Res 2014; 1571:1-11. [PMID: 24842001 DOI: 10.1016/j.brainres.2014.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/02/2013] [Revised: 05/03/2014] [Accepted: 05/08/2014] [Indexed: 11/28/2022]
Abstract
Thyrotropin releasing hormone (TRH) is a neuropeptide with a wide neural distribution and a variety of functions. It modulates neuronal electrophysiological properties, including resting membrane potential, as well as excitatory postsynaptic potential and spike frequencies. We explored, with whole-cell patch clamp, TRH effect on action potential shape in pyramidal neurons of the sensorimotor cortex. TRH reduced spike and after hyperpolarization amplitudes, and increased spike half-width. The effect varied with dose, time and cortical layer. In layer V, 0.5µM of TRH induced a small increase in spike half-width, while 1 and 5µM induced a strong but transient change in spike half-width, and amplitude; after hyperpolarization amplitude was modified at 5µM of TRH. Cortical layers III and VI neurons responded intensely to 0.5µM TRH; layer II neurons response was small. The effect of 1µM TRH on action potential shape in layer V neurons was blocked by G-protein inhibition. Inhibition of the activity of the TRH-degrading enzyme pyroglutamyl peptidase II (PPII) reproduced the effect of TRH, with enhanced spike half-width. Many cortical PPII mRNA+ cells were VGLUT1 mRNA+, and some GAD mRNA+. These data show that TRH regulates action potential shape in pyramidal cortical neurons, and are consistent with the hypothesis that PPII controls its action in this region.
Collapse
Affiliation(s)
- Víctor Rodríguez-Molina
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), AP 70250, México, D.F. 04510, México; Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Javier Patiño
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Yamili Vargas
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Ave. Universidad 2001, Chamilpa, Cuernavaca, Morelos 62210, México
| | - Edith Sánchez-Jaramillo
- Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría, Ramón de la Fuente Muñíz, México D.F., México
| | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Ave. Universidad 2001, Chamilpa, Cuernavaca, Morelos 62210, México
| | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Ave. Universidad 2001, Chamilpa, Cuernavaca, Morelos 62210, México.
| |
Collapse
|