1
|
Herrera G, Scimonelli T, Lasaga M, Granero G, Onnainty R. Polysorbate 80 coated chitosan nanoparticles for delivery of α-melanocyte stimulating hormone analog (NDP-MSH) to the brain reverse cognitive impairment related to neuroinflammation produced by a high-fat diet (HFD). Neuropharmacology 2024; 253:109969. [PMID: 38688422 DOI: 10.1016/j.neuropharm.2024.109969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
This study aimed to develop polysorbate 80-coated chitosan nanoparticles (PS80/CS NPs) as a delivery system for improved brain targeting of α-Melanocyte Stimulating Hormone analog (NDP-MSH). Chitosan nanoparticles loaded with NDP-MSH were surface-modified with polysorbate 80 ([NDP-MSH]-PS80/CS NP), which formed a flattened layer on their surface. Nanoparticle preparation involved ionic gelation, followed by characterization using scanning electron microscopy (SEM) for morphology, dynamic light scattering (DLS) for colloidal properties, and ATR-FTIR spectroscopy for structure. Intraperitoneal injection of FITC-PS80/CS NPs and [NDP-MSH]-PS80/CS NP in rats demonstrated their ability to cross the blood-brain barrier, reach the brain, and accumulate in CA1 neurons of the dorsal hippocampus within 2 h. Two experimental models of neuroinflammation were employed with Male Wistar rats: a short-term model involving high-fat diet (HFD) consumption for 5 days followed by an immune stimulus with LPS, and a long-term model involving HFD consumption for 8 weeks. In both models, [NDP-MSH]-PS80/CS NPs could reverse the decreased expression of contextual fear memory induced by the diets. These findings suggest that [NDP-MSH]-PS80/CS NPs offer a promising strategy to overcome the limitations of NDP-MSH regarding pharmacokinetics and enzymatic stability. By facilitating NDP-MSH delivery to the hippocampus, these nanoparticles can potentially mitigate the cognitive impairments associated with HFD consumption and neuroinflammation.
Collapse
Affiliation(s)
- Guadalupe Herrera
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Teresa Scimonelli
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Gladys Granero
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), UNC-CONICET, Departamento de Ciencias Farmacéuticas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Renée Onnainty
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), UNC-CONICET, Departamento de Ciencias Farmacéuticas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
2
|
Valea A, Nistor C, Ciobica ML, Sima OC, Carsote M. Endocrine Petrified Ear: Associated Endocrine Conditions in Auricular Calcification/Ossification (A Sample-Focused Analysis). Diagnostics (Basel) 2024; 14:1303. [PMID: 38928718 PMCID: PMC11202653 DOI: 10.3390/diagnostics14121303] [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: 04/30/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Petrified ear (PE), an exceptional entity, stands for the calcification ± ossification of auricular cartilage (CAC/OAC); its pathogenic traits are still an open matter. Endocrine panel represents one of the most important; yet, no standard protocol of assessments is available. Our objective was to highlight most recent PE data and associated endocrine (versus non-endocrine) ailments in terms of presentation, imagery tools, hormonal assessments, biopsy, outcome, pathogenic features. This was a comprehensive review via PubMed search (January 2000-March 2024). A total of 75 PE subjects included: 46 case reports/series (N = 49) and two imagery-based retrospective studies (N = 26) with CAC/OAC prevalence of 7-23% (N = 251) amid routine head/temporal bone CT scans. Endocrine PE (EPE): N = 23, male/female ratio = 10.5; average age = 56.78, ranges: 22-79; non-EPE cohort: N = 26; male/female ratio = 1.88, mean age = 49.44; ranges: 18-75 (+a single pediatric case).The longest post-diagnosis follow-up was of 6-7 years. The diagnosis of PE and endocrine anomalies was synchronous or not (time gap of 10-20 years). A novel case in point (calcified EPE amid autoimmune poly-endocrine syndrome type 2 with a 10-year post-diagnosis documented follow-up) was introduced. We re-analyzed EPE and re-classified another five subjects as such. Hence, the final EPE cohort (N = 50) showed: adrenal insufficiency was the most frequent endocrine condition (36%) followed by hypopituitarism (22%) and hypothyroidism (18%); 39% of the patients with adrenal failure had Addison's disease; primary type represented 72% of all cases with hypothyroidism; an endocrine autoimmune (any type) component was diagnosed in 18%. We propose the term of "endocrine petrified ear" and a workflow algorithm to assess the potential hormonal/metabolic background in PE.
Collapse
Affiliation(s)
- Ana Valea
- Department of Endocrinology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Clinical County Hospital, 400347 Cluj-Napoca, Romania
| | - Claudiu Nistor
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
| | - Mihai-Lucian Ciobica
- “Dr. Carol Davila” Central Military Emergency University Hospital, 010242 Bucharest, Romania;
- Department of Internal Medicine and Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Oana-Claudia Sima
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- “C.I. Parhon” National Institute of Endocrinology, 011683 Bucharest, Romania;
| | - Mara Carsote
- “C.I. Parhon” National Institute of Endocrinology, 011683 Bucharest, Romania;
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
3
|
Chang CL, Cai Z, Hsu SYT. A gel-forming α-MSH analog promotes lasting melanogenesis. Eur J Pharmacol 2023; 958:176008. [PMID: 37673364 DOI: 10.1016/j.ejphar.2023.176008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
The α-MSH peptide plays a significant role in the regulation of pigmentation via the melanocortin 1 receptor (MC1R). It increases the DNA repair capacity of melanocytes and reduces the incidence of skin cancers. As such, α-MSH analogs could have the utility for protecting against UV-induced skin DNA damage in susceptible patients. Recently, α-MSH analogs have been approved for the treatment of erythropoietic protoporphyria, hypoactive sexual desire, or pediatric obesity. However, the delivery of these drugs requires inconvenient implants or frequent injections. We recently found that select palmitoylated melanocortin analogs such as afamelanotide and adrenocorticotropin peptides self-assemble to form liquid gels in situ. To explore the utility of these novel analogs, we studied their pharmacological characteristics in vitro and in vivo. Acylated afamelanotide (DDE 313) and ACTH1-24 (DDE314) analogs form liquid gels at 6-20% and have a significantly increased viscosity at >2.5% compared to original analogs. Using the DDE313 analog as a prototype, we showed gel-formation reduces the passage of DDE313 through Centricon filters, and subcutaneous injection of analog gel in rats leads to the sustained presence of the peptide in circulation for >12 days. In addition, DDE313 darkened the skin of frogs for >4 weeks, whereas those injected with an equivalent dose of afamelanotide lost the tanning response within a few days. Because self-assembled gels allow sustained activation of melanocortin receptors, further studies of these analogs may allow the development of effective and convenient tanning therapies to prophylactically protect against UV-induced malignant transformation of skin cells in susceptible patients.
Collapse
Affiliation(s)
- Chia Lin Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Zheqing Cai
- CL Laboratory LLC, Gaithersburg, MD, 20878, United States
| | | |
Collapse
|
4
|
Shaughnessy CA, Le K, Myhre VD, Dores RM. Functional characterization of melanocortin 2 receptor (Mc2r) from a lobe-finned fish (Protopterus annectens) and insights into the molecular evolution of melanocortin receptors. Gen Comp Endocrinol 2023; 343:114356. [PMID: 37562700 DOI: 10.1016/j.ygcen.2023.114356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/25/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
Recent studies from our group on melanocortin 2 receptors (Mc2r) from basal families of actinopterygians have served to resolve that Mrap1 dependence and ACTH selectivity are features of even the most basal ray-finned fishes. However, there have been no studies on Mc2r function of the basal sarcopterygians, the lobe-finned fishes, represented by the extant members coelacanths and lungfishes. Here, we offer the first molecular and functional characterization of an Mc2r from a lobe-finned fish, the West African lungfish (Protopterus annectens). Plasmids containing cDNA constructs of lungfish (lf) Mc2r and Mrap1 were expressed in mammalian and zebrafish cell lines. Cells were then stimulated by human ACTH(1-24) and melanocyte stimulating hormone (α-MSH), as well as alanine-substituted analogs of hACTH(1-24) targeting residues within the H6F7R8W9 and K15K16R17R18P19 motifs. Activation of lfMc2r was assessed using a cAMP-responsive luciferase reporter gene assay. In these assays, lfMc2r required co-expression with lfMrap1, was selective for ACTH over α-MSH at physiological concentrations of the ligands, and was completely inhibited by multiple-alanine substitutions of the HFRW (A6-9) and KKRRP (A15-19) motifs. Single- and partial-alanine substitutions of the HFRW and KKRRP motifs varied in their impacts on receptor-ligand affinity from having no effect to completely inhibiting lfMc2r activation. This characterization of the Mc2r of a lobe-finned fish fulfills the last major extant vertebrate group for which Mc2r function had yet to be characterized. In doing so, we resolve that all basal bony vertebrate groups exhibit Mc2r function that substantially differs from that of the cartilaginous fishes, indicating that rapid and dramatic shift in Mc2r function occurred between the radiation of cartilaginous fishes and the emergence of bony fishes. We support this interpretation with a molecular clock analysis of the melanocortin receptors, which demonstrates the uniquely high rate of sequence divergence in Mc2r. Much remains to be understood regarding the molecular evolution of Mc2r during the early radiation of vertebrates that resulted in the derived functional characteristics of Mrap1 dependence and exclusive selectivity for ACTH.
Collapse
Affiliation(s)
| | - Khoa Le
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Valorie D Myhre
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| |
Collapse
|
5
|
Ruggiero-Ruff RE, Villa PA, Hijleh SA, Avalos B, DiPatrizio NV, Haga-Yamanaka S, Coss D. Increased body weight in mice with fragile X messenger ribonucleoprotein 1 (Fmr1) gene mutation is associated with hypothalamic dysfunction. Sci Rep 2023; 13:12666. [PMID: 37542065 PMCID: PMC10403586 DOI: 10.1038/s41598-023-39643-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
Mutations in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene are linked to Fragile X Syndrome, the most common monogenic cause of intellectual disability and autism. People affected with mutations in FMR1 have higher incidence of obesity, but the mechanisms are largely unknown. In the current study, we determined that male Fmr1 knockout mice (KO, Fmr1-/y), but not female Fmr1-/-, exhibit increased weight when compared to wild-type controls, similarly to humans with FMR1 mutations. No differences in food or water intake were found between groups; however, male Fmr1-/y display lower locomotor activity, especially during their active phase. Moreover, Fmr1-/y have olfactory dysfunction determined by buried food test, although they exhibit increased compulsive behavior, determined by marble burying test. Since olfactory brain regions communicate with hypothalamic regions that regulate food intake, including POMC neurons that also regulate locomotion, we examined POMC neuron innervation and numbers in Fmr1-/y mice. POMC neurons express Fmrp, and POMC neurons in Fmr1-/y have higher inhibitory GABAergic synaptic inputs. Consistent with increased inhibitory innervation, POMC neurons in the Fmr1-/y mice exhibit lower activity, based on cFOS expression. Notably, Fmr1-/y mice have fewer POMC neurons than controls, specifically in the rostral arcuate nucleus, which could contribute to decreased locomotion and increased body weight. These results suggest a role for Fmr1 in the regulation of POMC neuron function and the etiology of Fmr1-linked obesity.
Collapse
Affiliation(s)
- Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Sarah Abu Hijleh
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Bryant Avalos
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Sachiko Haga-Yamanaka
- Department of Molecular, Cell, and Systems Biology, College of Natural and Agricultural Sciences, University of California, Riverside, Riverside, USA
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
6
|
Feng W, Zhou Q, Chen X, Dai A, Cai X, Liu X, Zhao F, Chen Y, Ye C, Xu Y, Cong Z, Li H, Lin S, Yang D, Wang MW. Structural insights into ligand recognition and subtype selectivity of the human melanocortin-3 and melanocortin-5 receptors. Cell Discov 2023; 9:81. [PMID: 37524700 PMCID: PMC10390531 DOI: 10.1038/s41421-023-00586-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023] Open
Abstract
Members of the melanocortin receptor (MCR) family that recognize different melanocortin peptides mediate a broad spectrum of cellular processes including energy homeostasis, inflammation and skin pigmentation through five MCR subtypes (MC1R-MC5R). The structural basis of subtype selectivity of the endogenous agonist γ-MSH and non-selectivity of agonist α-MSH remains elusive, as the two agonists are highly similar with a conserved HFRW motif. Here, we report three cryo-electron microscopy structures of MC3R-Gs in complex with γ-MSH and MC5R-Gs in the presence of α-MSH or a potent synthetic agonist PG-901. The structures reveal that α-MSH and γ-MSH adopt a "U-shape" conformation, penetrate into the wide-open orthosteric pocket and form massive common contacts with MCRs via the HFRW motif. The C-terminus of γ-MSH occupies an MC3R-specific complementary binding groove likely conferring subtype selectivity, whereas that of α-MSH distances itself from the receptor with neglectable contacts. PG-901 achieves the same potency as α-MSH with a shorter length by rebalancing the recognition site and mimicking the intra-peptide salt bridge in α-MSH by cyclization. Solid density confirmed the calcium ion binding in MC3R and MC5R, and the distinct modulation effects of divalent ions were demonstrated. Our results provide insights into ligand recognition and subtype selectivity among MCRs, and expand the knowledge of signal transduction among MCR family members.
Collapse
Affiliation(s)
- Wenbo Feng
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qingtong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xianyue Chen
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
| | - Antao Dai
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoqing Cai
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fenghui Zhao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chenyu Ye
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yingna Xu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaotong Cong
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hao Li
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
| | - Shi Lin
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
| | - Dehua Yang
- Research Center for Deepsea Bioresources, Sanya, Hainan, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Ming-Wei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Research Center for Deepsea Bioresources, Sanya, Hainan, China.
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan.
- School of Pharmacy, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
7
|
Shaughnessy CA, Myhre VD, Hall DJ, McCormick SD, Dores RM. Hypothalamus-pituitary-interrenal (HPI) axis signaling in Atlantic sturgeon (Acipenser oxyrinchus) and sterlet (Acipenser ruthenus). Gen Comp Endocrinol 2023; 339:114290. [PMID: 37088167 DOI: 10.1016/j.ygcen.2023.114290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 04/25/2023]
Abstract
In vertebrates, the hypothalamic-pituitary-adrenal/interrenal (HPA/HPI) axis is a highly conserved endocrine axis that regulates glucocorticoid production via signaling by corticotropin releasing hormone (CRH) and adrenocorticotropic hormone (ACTH). Once activated by ACTH, Gs protein-coupled melanocortin 2 receptors (Mc2r) present in corticosteroidogenic cells stimulate expression of steroidogenic acute regulatory protein (Star), which initiates steroid biosynthesis. In the present study, we examined the tissue distribution of genes involved in HPI axis signaling and steroidogenesis in the Atlantic sturgeon (Acipenser oxyrinchus) and provided the first functional characterization of Mc2r in sturgeon. Mc2r of A. oxyrinchus and the sterlet sturgeon (Acipenser ruthenus) are co-dependent on interaction with the melanocortin receptor accessory protein 1 (Mrap1) and highly selective for human (h) ACTH over other melanocortin ligands. A. oxyrinchus expresses key genes involved in HPI axis signaling in a tissue-specific manner that is indicative of the presence of a complete HPI axis in sturgeon. Importantly, we co-localized mc2r, mrap1, and star mRNA expression to the head kidney, indicating that this is possibly a site of ACTH-mediated corticosteroidogenesis in sturgeon. Our results are discussed in the context of other studies on the HPI axis of basal bony vertebrates, which, when taken together, demonstrate a need to better resolve the evolution of HPI axis signaling in vertebrates.
Collapse
Affiliation(s)
- Ciaran A Shaughnessy
- Department of Biological Sciences, University of Denver, Denver, CO, United States.
| | - Valorie D Myhre
- Department of Biological Sciences, University of Denver, Denver, CO, United States
| | - Daniel J Hall
- U.S. Geological Survey, Eastern Ecological Science Center, S. O. Conte Anadromous Fish Research Laboratory, Turners Falls, MA, USA
| | - Stephen D McCormick
- U.S. Geological Survey, Eastern Ecological Science Center, S. O. Conte Anadromous Fish Research Laboratory, Turners Falls, MA, USA; Department of Biology, University of Massachusetts, Amherst, MA, USA
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, Denver, CO, United States
| |
Collapse
|
8
|
Li Q, Jiang B, Zhang Z, Huang Y, Xu Z, Chen X, Huang Y, Jian J, Yan Q. α-MSH is partially involved in the immunomodulation of Nile tilapia (Oreochromis niloticus) antibacterial immunity. FISH & SHELLFISH IMMUNOLOGY 2022; 131:929-938. [PMID: 36343851 DOI: 10.1016/j.fsi.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
α-Melanocyte-stimulating hormone (α-MSH) is a well-studied neuropeptide controlling skin and hair color. Besides, numerous immunomodulation roles of α-MSH were recorded in humans and mice. However, the regulatory effects of α-MSH in teleost immunity haven't been well elucidated. In this study, several precursor molecules of α-MSH (POMCs) and its receptors (MCRs) in Nile tilapia (Oreochromis niloticus) were characterized, and their expression characteristics and specific functions on antibacterial immunity were determined. Overall, POMCs and MCRs were principally detected in the brain, skin, and liver, and were remarkably promoted post Streptococcus agalactiae infection. However, tiny POMCs and MCRs were observed in tilapia immune organs (head kidney and spleen) or lymphocytes, and no evident immunomodulation effect was detected in vitro. Moreover, the in vivo challenge experiments revealed that α-MSH protects tilapia from bacterial infection by regulating responses in the brain and intestine. This study lays theoretical data for a deeper comprehension of the immunomodulation mechanisms of teleost α-MSH and the evolutional process of the vertebrate melanocortin system.
Collapse
Affiliation(s)
- Qi Li
- Fisheries College, Jimei University, Xiamen, China; College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Baijian Jiang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Zhiqiang Zhang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yongxiong Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Zhou Xu
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Xinjin Chen
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Qingpi Yan
- Fisheries College, Jimei University, Xiamen, China.
| |
Collapse
|
9
|
Shaughnessy CA, Jensen MF, Dores RM. A basal actinopterygian melanocortin receptor: Molecular and functional characterization of an Mc2r ortholog from the Senegal bichir (Polypterus senegalus). Gen Comp Endocrinol 2022; 328:114105. [PMID: 35973587 DOI: 10.1016/j.ygcen.2022.114105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022]
Abstract
In bony vertebrates, melanocortin 2 receptor (Mc2r) specifically binds adrenocorticotropic hormone (ACTH) and is responsible for mediating anterior pituitary signaling that stimulates corticosteroid production in the adrenal gland/interrenal cells. In bony fishes Mc2r requires the chaperoning of an accessory protein (Mrap1) to traffic to the membrane surface and bind ACTH. Here, we evaluated the structure and pharmacological properties of Mc2r from the Senegal bichir (Polypterus senegalus), which represents the most basal bony fish from which an Mc2r has been pharmacologically studied to date. In our experiments, cDNA constructs of the Mc2r from the Senegal bichir (sbMc2r) and various vertebrate Mrap1s were heterologously co-expressed in Chinese hamster ovary (CHO) cells, stimulated by ACTH or melanocyte-stimulating hormone (α-MSH) ligands, and assessed using a luciferase reporter gene assay. When expressed without an Mrap1, sbMc2r was not activated by ACTH. When co-expressed with Mrap1 from either chicken (Gallus gallus) or bowfin (Amia calva), sbMc2r could be activated in a dose-dependent manner by ACTH, but not α-MSH. Co-expression of sbMrap2 with sbMc2r resulted in no detectable activation of the receptor. Collectively, these results demonstrate that sbMc2r has pharmacological properties similar to those of Mc2rs of later-evolved bony fishes, such as Mrap1 dependence and ACTH selectivity, indicating that these qualities of Mc2r function are ancestral to all bony fish Mc2rs.
Collapse
Affiliation(s)
| | - Mary F Jensen
- Department of Biological Sciences, University of Denver, Denver, CO
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, Denver, CO
| |
Collapse
|
10
|
Zhang X, Su J, Huang T, Wang X, Wu C, Li J, Li J, Zhang J, Wang Y. Characterization of the chicken melanocortin 5 receptor and its potential role in regulating hepatic glucolipid metabolism. Front Physiol 2022; 13:917712. [PMID: 36277187 PMCID: PMC9583845 DOI: 10.3389/fphys.2022.917712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022] Open
Abstract
Melanocortin receptors (MC1R-MC5R) and their accessory proteins (MRAPs) are involved in a variety of physiological processes, including pigmentation, lipolysis, adrenal steroidogenesis, and immunology. However, the physiological roles of MC5R are rarely characterized in vertebrates, particularly in birds. In this work, we cloned the full-length cDNA of chicken MC5R and identified its core promoter region. Functional studies revealed that cMC5R was more sensitive to ACTH/α-MSH than β-MSH/γ-MSH, and was coupled to the cAMP/PKA signaling pathway. We demonstrated that MRAP2 decreased MC5R sensitivity to α-MSH, whereas MRAP1 did not have a similar effect, and that both MRAPs significantly reduced MC5R expression on the cell membrane surface. Transcriptome and qPCR data showed that both MRAP1 and MC5R were highly expressed in chicken liver. Additionally, we observed that ACTH might increase hepatic glucose production and decrease lipogenesis in primary hepatocytes, and dose-dependently downregulated the expression levels of ELOVL6 and THRSPA genes. These findings indicated that ACTH may act directly on hepatocytes to regulate glucolipid metabolism, which will help to understand the function of MC5R in avian.
Collapse
Affiliation(s)
- Xiao Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jiancheng Su
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Tianjiao Huang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xinglong Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Chenlei Wu
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jing Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jiannan Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yajun Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Kwok-Shing Wong M, Dores RM. Analyzing the Hypothalamus/Pituitary/Interrenal axis of the neopterygian fish, Lepisosteus oculatus: Co-localization of MC2R, MC5R, MRAP1, and MRAP2 in interrenal cells. Gen Comp Endocrinol 2022; 323-324:114043. [PMID: 35447133 DOI: 10.1016/j.ygcen.2022.114043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/10/2022] [Accepted: 04/14/2022] [Indexed: 11/19/2022]
Abstract
RT-PCR analysis indicated that steroidogenic tissues are located along the length of the kidney of the neopterygian fish, Lepisosteus oculatus (spotted gar; g). However, RT-PCR analysis of the distribution of mc2r mRNA and mrap1 mRNA, critical components of the gar hypothalamus/pituitary/interrenal (HPI) axis, was only associated with the anterior and medial regions of the kidney. Steroidogenic cells were designated as interrenal cells that possess star mRNA (in situ hybridization) and lipid vesicles (histological analysis) within the kidney. RT-PCR also detected mc5r mRNA along the length of the tissues associated with the kidney. In situ hybridization analysis of the putative interrenal cells revealed co-expression of mc2r, and mc5r mRNAs in the same steroidogenic cells. Co-expression of gar Mc2r (gMc2r) and Mrap1 (gMrap1) in Chinese Hamster Ovary (CHO) cells stimulated with ACTH(1-24) resulted in activation with an EC50 value of 1.0 × 10-11M +/- 4.6 × 10-11); whereas stimulation of CHO cells co-expressed with gar Mc5r (gMc5r) and gMrap1 and stimulated with ACTH(1-24) resulted in an EC50 value that was 3 orders of magnitude lower (2.1 × 10-8 M +/- 3.5 × 10-9). Interesting, when CHO cells were co-transfected with gMc2r, gMc5r, and gMrap1 there was a decline in activation as measured by the Vmax values for CHO cells stimulated with either ACTH(1-24) or α-MSH. These results suggest that some interaction may occur between gMc2r and gMc5r when both receptors are expressed in the same cells. Phylogenetic and selection pressure analyses of vertebrate mc2r and mc5r genes concluded that the two genes are evolving at different rates after duplication from a proposed common ancestral gene.
Collapse
Affiliation(s)
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, USA.
| |
Collapse
|
12
|
Van Baelen AC, Robin P, Kessler P, Maïga A, Gilles N, Servent D. Structural and Functional Diversity of Animal Toxins Interacting With GPCRs. Front Mol Biosci 2022; 9:811365. [PMID: 35198603 PMCID: PMC8859281 DOI: 10.3389/fmolb.2022.811365] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Peptide toxins from venoms have undergone a long evolutionary process allowing host defense or prey capture and making them highly selective and potent for their target. This has resulted in the emergence of a large panel of toxins from a wide diversity of species, with varied structures and multiple associated biological functions. In this way, animal toxins constitute an inexhaustible reservoir of druggable molecules due to their interesting pharmacological properties. One of the most interesting classes of therapeutic targets is the G-protein coupled receptors (GPCRs). GPCRs represent the largest family of membrane receptors in mammals with approximately 800 different members. They are involved in almost all biological functions and are the target of almost 30% of drugs currently on the market. Given the interest of GPCRs in the therapeutic field, the study of toxins that can interact with and modulate their activity with the purpose of drug development is of particular importance. The present review focuses on toxins targeting GPCRs, including peptide-interacting receptors or aminergic receptors, with a particular focus on structural aspects and, when relevant, on potential medical applications. The toxins described here exhibit a great diversity in size, from 10 to 80 amino acids long, in disulfide bridges, from none to five, and belong to a large panel of structural scaffolds. Particular toxin structures developed here include inhibitory cystine knot (ICK), three-finger fold, and Kunitz-type toxins. We summarize current knowledge on the structural and functional diversity of toxins interacting with GPCRs, concerning first the agonist-mimicking toxins that act as endogenous agonists targeting the corresponding receptor, and second the toxins that differ structurally from natural agonists and which display agonist, antagonist, or allosteric properties.
Collapse
Affiliation(s)
- Anne-Cécile Van Baelen
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Philippe Robin
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Pascal Kessler
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Arhamatoulaye Maïga
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
- CHU Sainte Justine, Université de Montréal, Montreal, QC, Canada
| | - Nicolas Gilles
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
- *Correspondence: Denis Servent,
| |
Collapse
|
13
|
Hoglin BE, Miner M, Dores RM. Pharmacological properties of whale shark (Rhincodon typus) melanocortin-2 receptor and melancortin-5 receptor: Interaction with MRAP1 and MRAP2. Gen Comp Endocrinol 2022; 315:113915. [PMID: 34607718 DOI: 10.1016/j.ygcen.2021.113915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
In the current study, the whale shark (ws; Rhincodon typus) melanocortin-2 receptor (MC2R) co-expressed with wsMRAP1 in Chinese Hamster Ovary (CHO) Cells could be stimulated in a dose dependent manner by ACTH(1-24) with an EC50 of 2.6 × 10-10 M ± 9.7 × 10-11. When the receptor was expressed alone, stimulation was only observed at [10-6 M]. A comparable increase in sensitivity to stimulation by srDes-Ac-αMSH was also observed when the receptor was co-expressed with wsMRAP1. Furthermore, co-expression with wsMRAP1 significantly increased the trafficking of wsMC2R to the plasma membrane of CHO cells. Surprisingly, co-expression with wsMRAP2 also increased sensitivity to stimulation by ACTH(1-24) and srDes-Ac-αMSH, and increased trafficking of the receptor to the plasma membrane. These observations are in sharp contrast to the response of MC2R orthologs of bony vertebrates which have an obligate requirement for co-expression with MRAP1 for both trafficking to the plasma membrane and activation, whereas, co-expression with MRAP2 increases trafficking, but has minimal effects on activation. In addition, when comparing the activation features of wsMC2R with those of the elephant shark MC2R and red stingray MC2R orthologs, both similarities and differences are observed. The spectrum of features for cartilaginous fish MC2R orthologs will be discussed. A second objective of this study was to determine whether wsMC5R has features in common with wsMC2R in terms of ligand selectivity and interaction with wsMRAP paralogs. While wsMC5R can be activated by either srACTH(1-24) or srDes-Ac-αMSH, and co-expression with wsMRAP1 enhances this activation, wsMRAP1 had no effect on the trafficking of wsMC5R. In addition, co-expression with wsMRAP2 had no positive or negative effect on either ligand sensitivity or trafficking of wsMC5R.
Collapse
Affiliation(s)
- Brianne E Hoglin
- University of Denver, Department of Biological Sciences, Denver, CO 80210, USA
| | - Marin Miner
- University of Denver, Department of Biological Sciences, Denver, CO 80210, USA
| | - Robert M Dores
- University of Denver, Department of Biological Sciences, Denver, CO 80210, USA.
| |
Collapse
|
14
|
Fischer NH, Fumi E, Oliveira MT, Thulstrup PW, Diness F. Tuning peptide structure and function through fluorobenzene stapling. Chemistry 2021; 28:e202103788. [PMID: 34897848 DOI: 10.1002/chem.202103788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Indexed: 11/09/2022]
Abstract
Cyclic peptides are promising next-generation therapeutics with improved biological stability and activity. A catalyst-free stapling method for cysteine-containing peptides was developed. This enables fine-tuning of the macrocycle by using the appropriate regioisomers of fluorobenzene linkers. Stapling was performed on the unprotected linear peptide or, more conveniently, directly on-resin after peptide synthesis. NMR spectroscopy and circular dichroism studies demonstrate that the type of stapling can tune the secondary structures of the peptides. The method was applied to a set of potential agonists for melanocortin receptors, generating a library of macrocyclic potent ligands with ortho , meta or para relationships between the thioethers. Their small but significant difference in potency and efficacy demonstrates how the method allows facile fine-tuning of macrocyclic peptides towards biological targets from the same linear precursor.
Collapse
Affiliation(s)
| | - Erik Fumi
- University of Copenhagen: Kobenhavns Universitet, Department of Chemistry, DENMARK
| | | | - Peter W Thulstrup
- University of Copenhagen: Kobenhavns Universitet, Department of Chemistry, DENMARK
| | - Frederik Diness
- University of Copenhagen, Department of Chemistry, Universitetsparken 5, DK2100, Copenhagen, DENMARK
| |
Collapse
|
15
|
Lindberg I, Fricker LD. Obesity, POMC, and POMC-processing Enzymes: Surprising Results From Animal Models. Endocrinology 2021; 162:6333651. [PMID: 34333593 PMCID: PMC8489426 DOI: 10.1210/endocr/bqab155] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Indexed: 11/19/2022]
Abstract
Peptides derived from proopiomelanocortin (POMC) are well-established neuropeptides and peptide hormones that perform multiple functions, including regulation of body weight. In humans and some animals, these peptides include α- and β-melanocyte-stimulating hormone (MSH). In certain rodent species, no β-MSH is produced from POMC because of a change in the cleavage site. Enzymes that convert POMC into MSH include prohormone convertases (PCs), carboxypeptidases (CPs), and peptidyl-α-amidating monooxygenase (PAM). Humans and mice with inactivating mutations in either PC1/3 or carboxypeptidase E (CPE) are obese, which was assumed to result from defective processing of POMC into MSH. However, recent studies have shown that selective loss of either PC1/3 or CPE in POMC-expressing cells does not cause obesity. These findings suggest that defects in POMC processing cannot alone account for the obesity observed in global PC1/3 or CPE mutants. We propose that obesity in animals lacking PC1/3 or CPE activity depends, at least in part, on deficient processing of peptides in non-POMC-expressing cells either in the brain and/or the periphery. Genetic background may also contribute to the manifestation of obesity.
Collapse
Affiliation(s)
- Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Correspondence: I. Lindberg, PhD, Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, Baltimore, MD 21201, USA.
| | - Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
16
|
Xu Y, Li L, Zheng J, Wang M, Jiang B, Zhai Y, Lu L, Zhang C, Kuang Z, Yang X, Jin LN, Lin G, Zhang C. Pharmacological modulation of the cAMP signaling of two isoforms of melanocortin-3 receptor by melanocortin receptor accessory proteins in the tetrapod Xenopus laevis. Endocr Connect 2021; 10:1477-1488. [PMID: 34678757 PMCID: PMC8630767 DOI: 10.1530/ec-21-0179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/22/2021] [Indexed: 11/29/2022]
Abstract
As a member of the seven-transmembrane rhodopsin-like G protein-coupled receptor superfamily, the melanocortin-3 receptor (MC3R) is vital for the regulation of energy homeostasis and rhythms synchronizing in mammals, and its pharmacological effect could be directly influenced by the presence of melanocortin receptor accessory proteins (MRAPs), MRAP1 and MRAP2. The tetrapod amphibian Xenopus laevis (xl) retains higher duplicated genome than extant teleosts and serves as an ideal model system for embryonic development and physiological studies. However, the melanocortin system of the Xenopus laevis has not yet been thoroughly evaluated. In this work, we performed sequence alignment, phylogenetic tree, and synteny analysis of two xlMC3Rs. Co-immunoprecipitation and immunofluorescence assay further confirmed the co-localization and in vitro interaction of xlMC3Rs with xlMRAPs on the plasma membrane. Our results demonstrated that xlMRAP2.L/S could improve α-MSH-stimulated xlMC3Rs signaling and suppress their surface expression. Moreover, xlMC3R.L showed a similar profile on the ligands and surface expression in the presence of xlMRAP1.L. Overall, the distinct pharmacological modulation of xlMC3R.L and xlMC3R.S by dual MRAP2 proteins elucidated the functional consistency of melanocortin system during genomic duplication of tetrapod vertebrates.
Collapse
Affiliation(s)
- Ying Xu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lei Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jihong Zheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bopei Jiang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yue Zhai
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Liumei Lu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cong Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhe Kuang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaomei Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Correspondence should be addressed to X Yang or L-N Jin or G Lin or C Zhang: or or or
| | - Li-Na Jin
- Department of Hematology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Correspondence should be addressed to X Yang or L-N Jin or G Lin or C Zhang: or or or
| | - Gufa Lin
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Correspondence should be addressed to X Yang or L-N Jin or G Lin or C Zhang: or or or
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Correspondence should be addressed to X Yang or L-N Jin or G Lin or C Zhang: or or or
| |
Collapse
|
17
|
Structural mechanism of calcium-mediated hormone recognition and Gβ interaction by the human melanocortin-1 receptor. Cell Res 2021; 31:1061-1071. [PMID: 34453129 PMCID: PMC8486761 DOI: 10.1038/s41422-021-00557-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
Melanocortins are peptide hormones critical for the regulation of stress response, energy homeostasis, inflammation, and skin pigmentation. Their functions are mediated by five G protein-coupled receptors (MC1R-MC5R), predominately through the stimulatory G protein (Gs). MC1R, the founding member of melanocortin receptors, is mainly expressed in melanocytes and is involved in melanogenesis. Dysfunction of MC1R is associated with the development of melanoma and skin cancer. Here we present three cryo-electron microscopy structures of the MC1R-Gs complexes bound to endogenous hormone α-MSH, a marketed drug afamelanotide, and a synthetic agonist SHU9119. These structures reveal the orthosteric binding pocket for the conserved HFRW motif among melanocortins and the crucial role of calcium ion in ligand binding. They also demonstrate the basis of differential activities among different ligands. In addition, unexpected interactions between MC1R and the Gβ subunit were discovered from these structures. Together, our results elucidate a conserved mechanism of calcium-mediated ligand recognition, a specific mode of G protein coupling, and a universal activation pathway of melanocortin receptors.
Collapse
|
18
|
Antistress Action of Melanocortin Derivatives Associated with Correction of Gene Expression Patterns in the Hippocampus of Male Rats Following Acute Stress. Int J Mol Sci 2021; 22:ijms221810054. [PMID: 34576218 PMCID: PMC8469576 DOI: 10.3390/ijms221810054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/19/2023] Open
Abstract
Natural melanocortins (MCs) have been used in the successful development of drugs with neuroprotective properties. Here, we studied the behavioral effects and molecular genetic mechanisms of two synthetic MC derivatives-ACTH(4-7)PGP (Semax) and ACTH(6-9)PGP under normal and acute restraint stress (ARS) conditions. Administration of Semax or ACTH(6-9)PGP (100 μg/kg) to rats 30 min before ARS attenuated ARS-induced behavioral alterations. Using high-throughput RNA sequencing (RNA-Seq), we identified 1359 differentially expressed genes (DEGs) in the hippocampus of vehicle-treated rats subjected to ARS, using a cutoff of >1.5 fold change and adjusted p-value (Padj) < 0.05, in samples collected 4.5 h after the ARS. Semax administration produced > 1500 DEGs, whereas ACTH(6-9)PGP administration led to <400 DEGs at 4.5 h after ARS. Nevertheless, ~250 overlapping DEGs were identified, and expression of these DEGs was changed unidirectionally by both peptides under ARS conditions. Modulation of the expression of genes associated with biogenesis, translation of RNA, DNA replication, and immune and nervous system function was produced by both peptides. Furthermore, both peptides upregulated the expression levels of many genes that displayed decreased expression after ARS, and vice versa, the MC peptides downregulated the expression levels of genes that were upregulated by ARS. Consequently, the antistress action of MC peptides may be associated with a correction of gene expression patterns that are disrupted during ARS.
Collapse
|
19
|
Ancient fishes and the functional evolution of the corticosteroid stress response in vertebrates. Comp Biochem Physiol A Mol Integr Physiol 2021; 260:111024. [PMID: 34237466 DOI: 10.1016/j.cbpa.2021.111024] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022]
Abstract
The neuroendocrine mechanism underlying stress responses in vertebrates is hypothesized to be highly conserved and evolutionarily ancient. Indeed, elements of this mechanism, from the brain to steroidogenic tissue, are present in all vertebrate groups; yet, evidence of the function and even identity of some elements of the hypothalamus-pituitary-adrenal/interrenal (HPA/I) axis is equivocal among the most basal vertebrates. The purpose of this review is to discuss the functional evolution of the HPA/I axis in vertebrates with a focus on our understanding of this neuroendocrine mechanism in the most ancient vertebrates: the agnathan (i.e., hagfish and lamprey) and chondrichthyan fishes (i.e., sharks, rays, and chimeras). A review of the current literature presents evidence of a conserved HPA/I axis in jawed vertebrates (i.e., gnathostomes); yet, available data in jawless (i.e., agnathan) and chondrichthyan fishes are limited. Neuroendocrine regulation of corticosteroidogenesis in agnathans and chondrichthyans appears to function through similar pathways as in bony fishes and tetrapods; however, key elements have yet to be identified and the involvement of melanotropins and gonadotropin-releasing hormone in the stress axis in these ancient fishes warrants further investigation. Further, the identities of physiological glucocorticoids are uncertain in hagfishes, chondrichthyans, and even coelacanths. Resolving these and other knowledge gaps in the stress response of ancient fishes will be significant for advancing knowledge of the evolutionary origins of the vertebrate stress response.
Collapse
|
20
|
The Characterization of Sex Differences in Hypoglycemia-Induced Activation of HPA Axis on the Transcriptomic Level. Cell Mol Neurobiol 2021; 42:1523-1542. [PMID: 33544274 DOI: 10.1007/s10571-021-01043-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/11/2021] [Indexed: 11/27/2022]
Abstract
Activation of the hypothalamic-pituitary-adrenal (HPA) axis using an insulin tolerance test (ITT) is a medical diagnostic procedure that is frequently used in humans to assess the HPA and growth-hormone (GH) axes. Whether sex differences exist in the response to ITT stress is unknown. Thus, investigations into the analysis of transcripts during activation of the HPA axis in response to hypoglycemia have revealed the underlying influences of sex in signaling pathways that stimulate the HPA axis. We assessed four time points of ITT application in Balb/c mice. After insulin injection, expression levels of 192 microRNAs and 41 mRNAs associated with the HPA, GH and hypothalamic-pituitary-gonadal (HPG) axes were determined by real-time RT-PCR in the hypothalamus, pituitary and adrenal tissues, as well as blood samples (Raw data accession: https://drive.google.com/drive/folders/10qI00NAtjxOepcNKxSJnQbJeBFa6zgHK?usp=sharing ). Although the ITT is commonly used as a gold standard for evaluating the HPA axis, we found completely different responses between males and females with respect to activation of the HPA axis. While activation of several transcripts in the hypothalamus and pituitary was observed after performing the ITT in males within 10 min, females responded via the pituitary and adrenal immediately and durably over 40 min. Additionally, we found that microRNA alterations precede mRNA responses in the HPA axis. Furthermore, robust changes in the levels of several transcripts including Avpr1b and Avpr2 observed at all time points strongly suggest that transcriptional control of these genes occurs mostly via differential signaling in pituitary and blood between males and females. Male and female HPA axis responses to ITT involve a number of sophisticated regulatory signaling pathways of miRNAs and mRNAs. Our results highlight the first robust markers in several layers of HPA, HPG and GH axis involved in ITT/hypoglycemia stress-induced dynamics.
Collapse
|
21
|
Dinparastisaleh R, Mirsaeidi M. Antifibrotic and Anti-Inflammatory Actions of α-Melanocytic Hormone: New Roles for an Old Player. Pharmaceuticals (Basel) 2021; 14:ph14010045. [PMID: 33430064 PMCID: PMC7827684 DOI: 10.3390/ph14010045] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
The melanocortin system encompasses melanocortin peptides, five receptors, and two endogenous antagonists. Besides pigmentary effects generated by α-Melanocytic Hormone (α-MSH), new physiologic roles in sexual activity, exocrine secretion, energy homeostasis, as well as immunomodulatory actions, exerted by melanocortins, have been described recently. Among the most common and burdensome consequences of chronic inflammation is the development of fibrosis. Depending on the regenerative capacity of the affected tissue and the quality of the inflammatory response, the outcome is not always perfect, with the development of some fibrosis. Despite the heterogeneous etiology and clinical presentations, fibrosis in many pathological states follows the same path of activation or migration of fibroblasts, and the differentiation of fibroblasts to myofibroblasts, which produce collagen and α-SMA in fibrosing tissue. The melanocortin agonists might have favorable effects on the trajectories leading from tissue injury to inflammation, from inflammation to fibrosis, and from fibrosis to organ dysfunction. In this review we briefly summarized the data on structure, receptor signaling, and anti-inflammatory and anti-fibrotic properties of α-MSH and proposed that α-MSH analogues might be promising future therapeutic candidates for inflammatory and fibrotic diseases, regarding their favorable safety profile.
Collapse
Affiliation(s)
- Roshan Dinparastisaleh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL 33146, USA
- Correspondence: ; Tel.: +1-305-243-1377
| |
Collapse
|
22
|
Rousseau K, Prunet P, Dufour S. Special features of neuroendocrine interactions between stress and reproduction in teleosts. Gen Comp Endocrinol 2021; 300:113634. [PMID: 33045232 DOI: 10.1016/j.ygcen.2020.113634] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/10/2020] [Accepted: 09/20/2020] [Indexed: 02/08/2023]
Abstract
Stress and reproduction are both essential functions for vertebrate survival, ensuring on one side adaptative responses to environmental changes and potential life threats, and on the other side production of progeny. With more than 25,000 species, teleosts constitute the largest group of extant vertebrates, and exhibit a large diversity of life cycles, environmental conditions and regulatory processes. Interactions between stress and reproduction are a growing concern both for conservation of fish biodiversity in the frame of global changes and for the development of sustainability of aquaculture including fish welfare. In teleosts, as in other vertebrates, adverse effects of stress on reproduction have been largely documented and will be shortly overviewed. Unexpectedly, stress notably via cortisol, may also facilitate reproductive function in some teleost species in relation to their peculiar life cyles and this review will provide some examples. Our review will then mainly address the neuroendocrine axes involved in the control of stress and reproduction, namely the corticotropic and gonadotropic axes, as well as their interactions. After reporting some anatomo-functional specificities of the neuroendocrine systems in teleosts, we will describe the major actors of the corticotropic and gonadotropic axes at the brain-pituitary-peripheral glands (interrenals and gonads) levels, with a special focus on the impact of teleost-specific whole genome duplication (3R) on the number of paralogs and their potential differential functions. We will finally review the current knowledge on the neuroendocrine mechanisms of the various interactions between stress and reproduction at different levels of the two axes in teleosts in a comparative and evolutionary perspective.
Collapse
Affiliation(s)
- Karine Rousseau
- Muséum National d'Histoire Naturelle, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, SU, UCN, UA, Paris, France
| | - Patrick Prunet
- INRAE, UR1037, Laboratoire de Physiologie et de Génomique des Poissons (LPGP), Rennes, France
| | - Sylvie Dufour
- Muséum National d'Histoire Naturelle, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, SU, UCN, UA, Paris, France.
| |
Collapse
|
23
|
Dores RM, Chapa E. Hypothesis and Theory: Evaluating the Co-Evolution of the Melanocortin-2 Receptor and the Accessory Protein MRAP1. Front Endocrinol (Lausanne) 2021; 12:747843. [PMID: 34790168 PMCID: PMC8591103 DOI: 10.3389/fendo.2021.747843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/20/2021] [Indexed: 11/24/2022] Open
Abstract
The melanocortin receptors (MCRs) and the MRAP accessory proteins belong to distinct gene families that are unique to the chordates. During the radiation of the chordates, the melancortin-2 receptor paralog (MC2R) and the MRAP1 paralog (melanocortin-2 receptor accessory protein 1) have co-evolved to form a heterodimer interaction that can influence the ligand selectivity and trafficking properties of MC2R. This apparently spontaneous interaction may have begun with the ancestral gnathostomes and has persisted in both the cartilaginous fishes and the bony vertebrates. The ramifications of this interaction had profound effects on the hypothalamus/anterior pituitary/adrenal-interrenal axis of bony vertebrates resulting in MC2R orthologs that are exclusively selective for the anterior pituitary hormone, ACTH, and that are dependent on MRAP1 for trafficking to the plasma membrane. The functional motifs within the MRAP1 sequence and their potential contact sites with MC2R are discussed. The ramifications of the MC2R/MRAP1 interaction for cartilaginous fishes are also discussed, but currently the effects of this interaction on the hypothalamus/pituitary/interrenal axis is less clear. The cartilaginous fish MC2R orthologs have apparently retained the ability to be activated by either ACTH or MSH-sized ligands, and the effect of MRAP1 on trafficking varies by species. In this regard, the possible origin of the dichotomy between cartilaginous fish and bony vertebrate MC2R orthologs with respect to ligand selectivity and trafficking properties is discussed in light of the evolution of functional amino acid motifs within MRAP1.
Collapse
|
24
|
Berruien NNA, Smith CL. Emerging roles of melanocortin receptor accessory proteins (MRAP and MRAP2) in physiology and pathophysiology. Gene 2020; 757:144949. [PMID: 32679290 DOI: 10.1016/j.gene.2020.144949] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 01/07/2023]
Abstract
Melanocortin-2 receptor accessory protein (MRAP) has an unusual dual topology and influences the expression, localisation, signalling and internalisation of the melanocortin receptor 2 (MC2); the adrenocorticotropic hormone (ACTH) receptor. Mutations in MRAP are associated with familial glucocorticoid deficiency type-2 and evidence is emerging of the importance of MRAP in adrenal development and ACTH signalling. Human MRAP has two functional splice variants: MRAP-α and MRAP-β, unlike MRAP-β, MRAP-α has little expression in brain but is highly expressed in ovary. MRAP2, identified through whole human genome sequence analysis, has approximately 40% sequence homology to MRAP. MRAP2 facilitates MC2 localisation to the cell surface but not ACTH signalling. MRAP and MRAP2 have been found to regulate the surface expression and signalling of all melanocortin receptors (MC1-5). Additionally, MRAP2 moderates the signalling of the G-protein coupled receptors (GCPRs): orexin, prokineticin and GHSR1a; the ghrelin receptor. Whilst MRAP appears to be mainly involved in glucocorticoid synthesis, an important role is emerging for MRAP2 in regulating appetite and energy homeostasis. Transgenic models indicate the importance of MRAP in adrenal gland formation. Like MC3R and MC4R knockout mice, MRAP2 knockout mice have an obese phenotype. In vitro studies indicate that MRAP2 enhances the MC3 and MC4 response to the agonist αMSH, which, like ACTH, is produced through precursor polypeptide proopiomelanocortin (POMC) cleavage. Analysis of cohorts of individuals with obesity have revealed several MRAP2 genetic variants with loss of function mutations which are causative of monogenic hyperphagic obesity with hyperglycaemia and hypertension. MRAP2 may also be associated with female infertility. This review summarises current knowledge of MRAP and MRAP2, their influence on GPCR signalling, and focusses on pathophysiology, particularly familial glucocorticoid deficiency type-2 and obesity.
Collapse
Affiliation(s)
- Nasrin N A Berruien
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK.
| | - Caroline L Smith
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK.
| |
Collapse
|
25
|
Yang Y, Xu Y. The central melanocortin system and human obesity. J Mol Cell Biol 2020; 12:785-797. [PMID: 32976556 PMCID: PMC7816681 DOI: 10.1093/jmcb/mjaa048] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity and the associated comorbidities highlight the importance of understanding the regulation of energy homeostasis. The central melanocortin system plays a critical role in controlling body weight balance. Melanocortin neurons sense and integrate the neuronal and hormonal signals, and then send regulatory projections, releasing anorexigenic or orexigenic melanocortin neuropeptides, to downstream neurons to regulate the food intake and energy expenditure. This review summarizes the latest progress in our understanding of the role of the melanocortin pathway in energy homeostasis. We also review the advances in the identification of human genetic variants that cause obesity via mechanisms that affect the central melanocortin system, which have provided rational targets for treatment of genetically susceptible patients.
Collapse
Affiliation(s)
- Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
Fricker LD, Margolis EB, Gomes I, Devi LA. Five Decades of Research on Opioid Peptides: Current Knowledge and Unanswered Questions. Mol Pharmacol 2020; 98:96-108. [PMID: 32487735 DOI: 10.1124/mol.120.119388] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
In the mid-1970s, an intense race to identify endogenous substances that activated the same receptors as opiates resulted in the identification of the first endogenous opioid peptides. Since then, >20 peptides with opioid receptor activity have been discovered, all of which are generated from three precursors, proenkephalin, prodynorphin, and proopiomelanocortin, by sequential proteolytic processing by prohormone convertases and carboxypeptidase E. Each of these peptides binds to all three of the opioid receptor types (μ, δ, or κ), albeit with differing affinities. Peptides derived from proenkephalin and prodynorphin are broadly distributed in the brain, and mRNA encoding all three precursors are highly expressed in some peripheral tissues. Various approaches have been used to explore the functions of the opioid peptides in specific behaviors and brain circuits. These methods include directly administering the peptides ex vivo (i.e., to excised tissue) or in vivo (in animals), using antagonists of opioid receptors to infer endogenous peptide activity, and genetic knockout of opioid peptide precursors. Collectively, these studies add to our current understanding of the function of endogenous opioids, especially when similar results are found using different approaches. We briefly review the history of identification of opioid peptides, highlight the major findings, address several myths that are widely accepted but not supported by recent data, and discuss unanswered questions and future directions for research. SIGNIFICANCE STATEMENT: Activation of the opioid receptors by opiates and synthetic drugs leads to central and peripheral biological effects, including analgesia and respiratory depression, but these may not be the primary functions of the endogenous opioid peptides. Instead, the opioid peptides play complex and overlapping roles in a variety of systems, including reward pathways, and an important direction for research is the delineation of the role of individual peptides.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Elyssa B Margolis
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Ivone Gomes
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Lakshmi A Devi
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| |
Collapse
|
27
|
Parween S, Rihs S, Flück CE. Metformin inhibits the activation of melanocortin receptors 2 and 3 in vitro: A possible mechanism for its anti-androgenic and weight balancing effects in vivo? J Steroid Biochem Mol Biol 2020; 200:105684. [PMID: 32360359 DOI: 10.1016/j.jsbmb.2020.105684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Metformin is recommended as one of the first-line drugs for the treatment of type 2 diabetes and the metabolic syndrome. In addition to its insulin sensitizing effects, it has been shown to attenuate androgen excess in women with polycystic ovary syndrome (PCOS) or congenital adrenal hyperplasia (CAH), as well as to ameliorate obesity. The mechanisms of metformin action seem manifold. Preclinical studies suggest that it inhibits the cellular stress response at the level of the mitochondrial OXPHOS system and through AMPK dependent and independent mechanisms. Recent studies have shown that metformin decreases ACTH secretion from pituitary and reduces ACTH-stimulated adrenal secretion. In this study we investigated its specific effect through the melanocortin receptor 2 (MC2R) on signaling targeting adrenal steroidogenesis. To assess this effect, we used mouse adrenal OS3 cells, which do not express the MC2R. Cells were transfected with the MC2R and stimulated by ACTH. Downstream cyclic AMP production was then assessed by a co-transfected cAMP-responsive vector producing luciferase that was measured by a dual luciferase assay. The amount of luciferase produced in this assay corresponds to the amount of receptor activation with varying amount of ACTH. The effect of metformin was then tested in this system. We found a significant inhibition of ACTH induced MC2R activation and signaling with 10 mM metformin. The ACTH concentration response curve (CRC) was half-log shifted and a ∼30 % reduction in maximum receptor response (Rmax) to ACTH in presence of metformin was observed. This effect was dose dependent with an IC50 of 4.2 mM. qRT-PCR analyses showed that metformin decreased ACTH induced MC2R expression. Metformin did not affect cell viability and basal cAMP levels. We also tested the effect of metformin on homologous melanocortin receptors (MCRs). No significant effect was found on MC1R and MC4R activity. However, a log shift of EC50 of ACTH stimulation on MC3R was observed with metformin treatment. Metformin also inhibited melanocortin stimulating hormone (αMSH) induced MC3R activity. In conclusion, we show that metformin acts on MC2R and MC3R signaling directly. The role of MC2R for steroidogenesis is well established. MC3R is involved in energy balance and seems to act as a rheostat when the metabolism is challenged. Our study may explain how metformin helps in weight loss and attenuates the excess response to ACTH in androgen excess disorders such as PCOS and CAH.
Collapse
MESH Headings
- Adrenocorticotropic Hormone/pharmacology
- Androgen Antagonists/pharmacology
- Animals
- Cell Line
- Cell Survival/drug effects
- Hypoglycemic Agents/pharmacology
- Metformin/pharmacology
- Mice
- Receptor, Melanocortin, Type 2/antagonists & inhibitors
- Receptor, Melanocortin, Type 2/genetics
- Receptor, Melanocortin, Type 2/metabolism
- Receptor, Melanocortin, Type 3/antagonists & inhibitors
- Receptor, Melanocortin, Type 3/metabolism
- Weight Loss
Collapse
Affiliation(s)
- Shaheena Parween
- Pediatric Endocrinology, Diabetology, and Metabolism, Department of Pediatrics, University Children's Hospital Bern, 3010, Bern, Switzerland; Department of Biomedical Research, University of Bern, 3010, Bern, Switzerland
| | - Silvia Rihs
- Pediatric Endocrinology, Diabetology, and Metabolism, Department of Pediatrics, University Children's Hospital Bern, 3010, Bern, Switzerland; Department of Biomedical Research, University of Bern, 3010, Bern, Switzerland
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology, and Metabolism, Department of Pediatrics, University Children's Hospital Bern, 3010, Bern, Switzerland; Department of Biomedical Research, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
28
|
Delery EC, Edwards S. Neuropeptide and cytokine regulation of pain in the context of substance use disorders. Neuropharmacology 2020; 174:108153. [PMID: 32470337 DOI: 10.1016/j.neuropharm.2020.108153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Substance use disorders (SUDs) are frequently accompanied by affective symptoms that promote negative reinforcement mechanisms contributing to SUD maintenance or progression. Despite their widespread use as analgesics, chronic or excessive exposure to alcohol, opioids, and nicotine produces heightened nociceptive sensitivity, termed hyperalgesia. This review focuses on the contributions of neuropeptide (CRF, melanocortin, opioid peptide) and cytokine (IL-1β, TNF-α, chemokine) systems in the development and maintenance of substance-induced hyperalgesia. Few effective therapies exist for either chronic pain or SUD, and the common interaction of these disease states likely complicates their effective treatment. Here we highlight promising new discoveries as well as identify gaps in research that could lead to more effective and even simultaneous treatment of SUDs and co-morbid hyperalgesia symptoms.
Collapse
Affiliation(s)
- Elizabeth C Delery
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Scott Edwards
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
29
|
Yang Y, Harmon CM. Molecular determinants of ACTH receptor for ligand selectivity. Mol Cell Endocrinol 2020; 503:110688. [PMID: 31866318 DOI: 10.1016/j.mce.2019.110688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 01/04/2023]
Abstract
The adrenocorticotropic hormone (ACTH) receptor, known as the melanocortin-2 receptor (MC2R), plays a key role in regulating adrenocortical function. ACTH receptor is a subtype of the melanocortin receptor family which is a member of the G-protein coupled receptor (GPCR) superfamily. ACTH receptor has unique characteristics among MCRs. α-MSH, β-MSH, γ-MSH and ACTH are agonists for MCRs but only ACTH is the agonist for ACTH receptor. In addition, the melanocortin receptor accessory protein (MRAP) is required for ACTH receptor expression at cell surface and function. In this review, we summarized the information available on the relationship between ACTH and ACTH receptor and provide the latest understanding of the molecular basis of the ACTH receptor responsible for ligand selectivity and function.
Collapse
Affiliation(s)
- Yingkui Yang
- Department of Surgery, State University of New York at Buffalo, USA.
| | - Carroll M Harmon
- Department of Surgery, State University of New York at Buffalo, USA
| |
Collapse
|
30
|
Lerma-Cabrera JM, Carvajal F, Garbutt JC, Navarro M, Thiele TE. The melanocortin system as a potential target for treating alcohol use disorders: A review of pre-clinical data. Brain Res 2019; 1730:146628. [PMID: 31891691 DOI: 10.1016/j.brainres.2019.146628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/02/2019] [Accepted: 12/26/2019] [Indexed: 12/17/2022]
Abstract
The melanocortin (MC) system consists of neuropeptides that are cleaved from the polypeptide precursor proopiomelanocortin (POMC). In the brain, MC neuropeptides signal primarily through the MC-3 and MC-4 receptors, which are widely expressed throughout the brain. While the MC system has been largely studied for its role in food intake and body weight regulation, converging evidence has emerged over approximately the last 20-years showing that alcohol (ethanol), and other drugs of abuse influence the central MC system, and that manipulating MC receptor signalling modulates ethanol intake. Although there is divergent evidence, the wealth of data appears to suggest that activating MC signalling, primarily through the MC-4 receptor, is protective against excessive ethanol consumption. In the present review, we first describe the MC system and then detail how ethanol exposure and consumption alters central MC and MC-receptor expression and levels. This is followed by a review of the data, from pharmacological and genetic studies, which show that manipulations of MC receptor activity alter ethanol intake. We then briefly highlight studies implicating a role for the MC system in modulating neurobiological responses and intake of other drugs of abuse, including amphetamine, cocaine and opioids. Finally, we introduce relatively new observations that the drug, bupropion (BUP), a drug that activates central MC activity, significantly reduces ethanol intake in rodent models when administered alone and in combination with the non-selective opioid receptor antagonist, naltrexone. Phase II clinical trials are currently underway to assess the efficacy of BUP as a treatment for alcohol use disorders.
Collapse
Affiliation(s)
| | | | - James C Garbutt
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Montserrat Navarro
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Todd E Thiele
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
31
|
Lainez NM, Coss D. Obesity, Neuroinflammation, and Reproductive Function. Endocrinology 2019; 160:2719-2736. [PMID: 31513269 PMCID: PMC6806266 DOI: 10.1210/en.2019-00487] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
The increasing occurrence of obesity has become a significant public health concern. Individuals with obesity have higher prevalence of heart disease, stroke, osteoarthritis, diabetes, and reproductive disorders. Reproductive problems include menstrual irregularities, pregnancy complications, and infertility due to anovulation, in women, and lower testosterone and diminished sperm count, in men. In particular, women with obesity have reduced levels of both gonadotropin hormones, and, in obese men, lower testosterone is accompanied by diminished LH. Taken together, these findings indicate central dysregulation of the hypothalamic-pituitary-gonadal axis, specifically at the level of the GnRH neuron function, which is the final brain output for the regulation of reproduction. Obesity is a state of hyperinsulinemia, hyperlipidemia, hyperleptinemia, and chronic inflammation. Herein, we review recent advances in our understanding of how these metabolic and immune changes affect hypothalamic function and regulation of GnRH neurons. In the latter part, we focus on neuroinflammation as a major consequence of obesity and discuss findings that reveal that GnRH neurons are uniquely positioned to respond to inflammatory changes.
Collapse
Affiliation(s)
- Nancy M Lainez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California
- Correspondence: Djurdjica Coss, PhD, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, 303 SOM Research Building, 900 University Avenue, Riverside, California 92521. E-mail:
| |
Collapse
|
32
|
Wolverton EA, Wong MKS, Davis PE, Hoglin B, Braasch I, Dores RM. Analyzing the signaling properties of gar (Lepisosteus oculatus) melanocortin receptors: Evaluating interactions with MRAP1 and MRAP2. Gen Comp Endocrinol 2019; 282:113215. [PMID: 31276671 PMCID: PMC7263024 DOI: 10.1016/j.ygcen.2019.113215] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 12/18/2022]
Abstract
RT-PCR analysis of gar pituitary and brain indicated that different combinations of gar melanocortin receptor mRNAs are present in the same tissues with mRNAs for gar mrap1 and gar mrap2. Against this background, an objective of this study was to determine whether the ligand sensitivity for either ACTH or α-MSH was affected when gar (g) melanocortin receptors (Mcrs) were co-expressed with either of the accessory proteins gMrap1 or gMrap2 in Chinese Hamster Ovary cells. The results indicated that gMc2r has an obligatory requirement for co-expression with gMrap1 in order for the receptor to be activated by hACTH(1-24). In addition, activation of gMc2r did not occur when the receptor was expressed alone or co-expressed with gMrap2. Furthermore, co-expression of gMc2r with gMrap1 followed by stimulation with NDP-MSH resulted in a low level of activation (only at 10-7 M and 10-6 M). However, gMc1r, gMc3r, gMc4r, and gMc5r responded to stimulation by NDP-MSH in a more robust manner. Co-expression of gMc1r, gMc3r, gMc4r, and gMc5r with gMRAP1 had no effect on sensitivity to stimulation by NDP-MSH or hACTH(1-24). Co-expression with gMRAP2 had no negative or positive effect on ligand sensitivity for gMc1r, gMc3r, and gMc5r, however this treatment did increase the activation of CHO cells transfected with gMc4r following stimulation with both hACTH(1-24) (p < 0.001), and NDP-MSH (p < 0.001). Co-expression of gMC5R with either gMRAP1 or gMRAP2 increased trafficking of gMC5R to the plasma membrane. These pharmacological observations are compared to the response of melanocortin receptors from other neopterygian fishes, cartilaginous fishes, and tetrapods to stimulation by ACTH(1-24) and forms of α-MSH.
Collapse
Affiliation(s)
| | | | - Perry E Davis
- Department of Biological Sciences, University of Denver, USA
| | - Brianne Hoglin
- Department of Biological Sciences, University of Denver, USA
| | - Ingo Braasch
- Integrative Biology, Michigan State University, USA
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, USA.
| |
Collapse
|
33
|
Davis PE, Wilkinson EC, Dores RM. Identifying Common Features in the Activation of Melanocortin-2 Receptors: Studies on the Xenopus tropicalis Melanocortin-2 Receptor. Int J Mol Sci 2019; 20:ijms20174166. [PMID: 31454910 PMCID: PMC6747542 DOI: 10.3390/ijms20174166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/21/2019] [Accepted: 08/24/2019] [Indexed: 12/03/2022] Open
Abstract
The interaction between the pituitary hormone, adrenocorticotropin (ACTH), and melanocortin-2 receptor (MC2R) orthologs involves the H6 F7 R8 W9 and R/K15 K16 R17 R18 motifs in ACTH making contact with corresponding contact sites on MC2R. Earlier studies have localized the common HFRW binding site of all melanocortin receptors to residues in TM2, TM3, and TM6 that are located close to the extracellular space. The current study has identified residues in Xenopus tropicalis (xt) MC2R in TM4 (I158, F161), in EC2 (M166), and in TM5 (V172) that also are involved in activation of xtMC2R, and may be in the R/KKRR contact site of xtMC2R. These results are compared to earlier studies on the corresponding domains of human MC2R and rainbow trout MC2R in an effort to identify common features in the activation of teleost and tetrapod MC2R orthologs following stimulation with ACTH.
Collapse
Affiliation(s)
- Perry E Davis
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Emily C Wilkinson
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA.
| |
Collapse
|
34
|
Abstract
Proopiomelanocortin (POMC) belongs to the opioid/orphanin gene family whose peptide precursors include either opioid (YGGF) or the orphanin/nociceptin core sequences (FGGF). In addition to POMC the family includes the proenkephalin (PENK), prodynorphin (PDYN), and nociceptin/proorphanin (PNOC) precursors. The opioid core sequence in POMC is incorporated by the β-endorphin that occupies the C-terminal region but this propeptide also exhibits at least two "alien" melanocortin core sequences (HFRW). An ACTH/MSH fragment merged into the opioid fragment not earlier than the two tetraploidizations of the vertebrate genome. Therefore, POMC exhibit a complex "evolutionary life" since the gene has coevolved together with two different receptor systems, i.e., opioid and melanocortin following a horse trading system. In this article, we summarize the different evolutionary hypotheses proposed for POMC evolution.
Collapse
Affiliation(s)
- Ana Rocha
- Department of Fish Physiology and Biotechnology, Institute of Aquaculture from Torre la Sal (IATS-CSIC), Castellon, Spain
| | - Alejandra Godino-Gimeno
- Department of Fish Physiology and Biotechnology, Institute of Aquaculture from Torre la Sal (IATS-CSIC), Castellon, Spain
| | - José Miguel Cerdá-Reverter
- Department of Fish Physiology and Biotechnology, Institute of Aquaculture from Torre la Sal (IATS-CSIC), Castellon, Spain..
| |
Collapse
|
35
|
Baldini G, Phelan KD. The melanocortin pathway and control of appetite-progress and therapeutic implications. J Endocrinol 2019; 241:R1-R33. [PMID: 30812013 PMCID: PMC6500576 DOI: 10.1530/joe-18-0596] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022]
Abstract
The initial discovery that ob/ob mice become obese because of a recessive mutation of the leptin gene has been crucial to discover the melanocortin pathway to control appetite. In the melanocortin pathway, the fed state is signaled by abundance of circulating hormones such as leptin and insulin, which bind to receptors expressed at the surface of pro-opiomelanocortin (POMC) neurons to promote processing of POMC to the mature hormone α-melanocyte-stimulating hormone (α-MSH). The α-MSH released by POMC neurons then signals to decrease energy intake by binding to melanocortin-4 receptor (MC4R) expressed by MC4R neurons to the paraventricular nucleus (PVN). Conversely, in the 'starved state' activity of agouti-related neuropeptide (AgRP) and of neuropeptide Y (NPY)-expressing neurons is increased by decreased levels of circulating leptin and insulin and by the orexigenic hormone ghrelin to promote food intake. This initial understanding of the melanocortin pathway has recently been implemented by the description of the complex neuronal circuit that controls the activity of POMC, AgRP/NPY and MC4R neurons and downstream signaling by these neurons. This review summarizes the progress done on the melanocortin pathway and describes how obesity alters this pathway to disrupt energy homeostasis. We also describe progress on how leptin and insulin receptors signal in POMC neurons, how MC4R signals and how altered expression and traffic of MC4R change the acute signaling and desensitization properties of the receptor. We also describe how the discovery of the melanocortin pathway has led to the use of melanocortin agonists to treat obesity derived from genetic disorders.
Collapse
Affiliation(s)
- Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kevin D. Phelan
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
36
|
Bertolesi GE, Zhang JZ, McFarlane S. Plasticity for colour adaptation in vertebrates explained by the evolution of the genes pomc, pmch and pmchl. Pigment Cell Melanoma Res 2019; 32:510-527. [PMID: 30791235 PMCID: PMC7167667 DOI: 10.1111/pcmr.12776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/27/2019] [Accepted: 02/16/2019] [Indexed: 02/06/2023]
Abstract
Different camouflages work best with some background matching colour. Our understanding of the evolution of skin colour is based mainly on the genetics of pigmentation ("background matching"), with little known about the evolution of the neuroendocrine systems that facilitate "background adaptation" through colour phenotypic plasticity. To address the latter, we studied the evolution in vertebrates of three genes, pomc, pmch and pmchl, that code for α-MSH and two melanin-concentrating hormones (MCH and MCHL). These hormones induce either dispersion/aggregation or the synthesis of pigments. We find that α-MSH is highly conserved during evolution, as is its role in dispersing/synthesizing pigments. Also conserved is the three-exon pmch gene that encodes MCH, which participates in feeding behaviours. In contrast, pmchl (known previously as pmch), is a teleost-specific intron-less gene. Our data indicate that in zebrafish, pmchl-expressing neurons extend axons to the pituitary, supportive of an MCHL hormonal role, whereas zebrafish and Xenopus pmch+ neurons send axons dorsally in the brain. The evolution of these genes and acquisition of hormonal status for MCHL explain different mechanisms used by vertebrates to background-adapt.
Collapse
Affiliation(s)
- Gabriel E Bertolesi
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - John Zhijia Zhang
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sarah McFarlane
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
37
|
Barney E, Dores MR, McAvoy D, Davis P, Racareanu RC, Iki A, Hyodo S, Dores RM. Elephant shark melanocortin receptors: Novel interactions with MRAP1 and implication for the HPI axis. Gen Comp Endocrinol 2019; 272:42-51. [PMID: 30468718 DOI: 10.1016/j.ygcen.2018.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
Abstract
The presence of Mrap1 and Mrap2 orthologs in the genome of the elephant shark (es), a cartilaginous fish, presented an opportunity to evaluate the potential interactions between these accessory proteins and melanocortin receptors of a cartilaginous fish. RT-PCR analysis indicated that Mrap1 mRNA was present in interrenal, brain, and pituitary tissue with mRNA for Mc2R, Mc3R, Mc4R, and Mc5r. Co-expression of esMrap1 cDNA with esMc2r cDNA or esMc5r cDNA in CHO cells increased sensitivity to stimulation with ACTH(1-24) 10 fold and 100 fold, respectfully, but had no effect on sensitivity to stimulation with DesAc-αMSH [i.e., ACTH(1-13)NH2] for either receptor, and had no effect on the ligand sensitivity of either esMc3r or esMc4r. Fluorescence image analysis indicated co-localization of esMrap1/esMc2r, and esMrap1/esMc5r on the plasma membrane; however, cell surface ELISA analysis indicated that co-expression with esMrap1 had no effect, positive or negative, on the trafficking of either esMc2r or esMc5r to the plasma membrane. RT-PCR analysis also indicated that Mrap2 mRNA, as well as, mRNAs for Mc2r, Mc3r, Mc4r, and Mc5r could be detected in brain tissue, however no Mrap2 mRNA was detected in interrenal tissue. Co-expression of esMrap2 in CHO cells with, respectively, esMc2r, esMc4r, or esMc5r had no effect on ligand sensitivity. However, co-expression of esMrap2 with esMc3r did lower sensitivity to stimulation by DesAc-αMSH 10 fold. These observations are discussed in the context of the parallel evolution of melanocortin receptors and their accessory proteins, and the hypothalamus/pituitary/adrenal axis and the hypothalamus/pituitary/interrenal axis in bony vertebrates and cartilaginous fishes.
Collapse
Affiliation(s)
- Emily Barney
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Michael R Dores
- Department of Biology, Hofstra University, Hampstead, NY, USA
| | - Danielle McAvoy
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Perry Davis
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | | | - Ayuko Iki
- Atmosphere and Ocean Research Institute, University of Tokyo, Chiba, Japan
| | - Susumu Hyodo
- Atmosphere and Ocean Research Institute, University of Tokyo, Chiba, Japan
| | - Robert M Dores
- Department of Biological Sciences, University of Denver, Denver, CO, USA.
| |
Collapse
|
38
|
Samaras A, Pavlidis M. Regulation of divergent cortisol responsiveness in European sea bass, Dicentrarchus labrax L. PLoS One 2018; 13:e0202195. [PMID: 30096195 PMCID: PMC6086447 DOI: 10.1371/journal.pone.0202195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 07/30/2018] [Indexed: 11/18/2022] Open
Abstract
Mechanisms regulating differences in cortisol responsiveness between low (LR) and high response (HR) individuals have been poorly studied. In this context, we aimed to study key regulatory processes in cortisol dynamics at the head kidneys of LR and HR European sea bass. To do so, resting plasma cortisol and ACTH concentrations were quantified in these fish. Additionally, the head kidneys of these individuals were superfused through an in vitro superfusion system and stimulated with the same amount of ACTH to assess their cortisol biosynthetic capacity. Moreover, the expression of important genes in cortisol regulation was assessed. Results showed that LR fish had lower resting cortisol concentrations than HR, although no differences existed in the circulating levels of ACTH. Additionally, the biosynthetic capacity of HR was higher than that of LR fish when in vitro stimulated with ACTH. At the molecular level, a statistically significant 3.4-fold higher expression of the ACTH receptor, mc2r, and a 2.3-fold, though not significant, higher expression of 11β-hydroxylase (cyp11b1), an enzyme involved in cortisol biosynthesis, was observed in the HR fish. Finally, a statistically significant 1.3-fold lower expression of 11β-hydroxysteroid dehydrogenase 2 (hsd11b2), an enzyme involved in cortisol inactivation, was observed in HR when compared to LR fish. Therefore, it was for the first time indicated that cortisol dynamics can also be regulated at the post-production level in the head kidney. Collectively, our results highlight the crucial role of the interrenal tissue in the regulation of differences in cortisol response between LR and HR sea bass individuals.
Collapse
Affiliation(s)
| | - Michail Pavlidis
- Department of Biology, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
39
|
Integrating Thyroid Hormone Signaling in Hypothalamic Control of Metabolism: Crosstalk Between Nuclear Receptors. Int J Mol Sci 2018; 19:ijms19072017. [PMID: 29997323 PMCID: PMC6073315 DOI: 10.3390/ijms19072017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/06/2018] [Accepted: 07/06/2018] [Indexed: 12/18/2022] Open
Abstract
The obesity epidemic is well recognized as a significant global health issue. A better understanding of the energy homeostasis mechanisms could help to identify promising anti-obesity therapeutic strategies. It is well established that the hypothalamus plays a pivotal role governing energy balance. The hypothalamus consists of tightly interconnected and specialized neurons that permit the sensing and integration of several peripheral inputs, including metabolic and hormonal signals for an appropriate physiological response. Current evidence shows that thyroid hormones (THs) constitute one of the key endocrine factors governing the regulation and the integration of metabolic homeostasis at the hypothalamic level. THs modulate numerous genes involved in the central control of metabolism, as TRH (Thyrotropin-Releasing Hormone) and MC4R (Melanocortin 4 Receptor). THs act through their interaction with thyroid hormone receptors (TRs). Interestingly, TH signaling, especially regarding metabolic regulations, involves TRs crosstalk with other metabolically linked nuclear receptors (NRs) including PPAR (Peroxisome proliferator-activated receptor) and LXR (Liver X receptor). In this review, we will summarize current knowledge on the important role of THs integration of metabolic pathways in the central regulation of metabolism. Particularly, we will shed light on the crosstalk between TRs and other NRs in controlling energy homeostasis. This could be an important track for the development of attractive therapeutic compounds.
Collapse
|
40
|
Ishizu H, Sekiguchi T, Ikari T, Kitamura KI, Kitani Y, Endo M, Urata M, Kinoshita Y, Hattori A, Srivastav AK, Mishima H, Mizusawa K, Takahashi A, Suzuki N. α-Melanocyte-stimulating hormone promotes bone resorption resulting from increased osteoblastic and osteoclastic activities in goldfish. Gen Comp Endocrinol 2018; 262:99-105. [PMID: 29574148 DOI: 10.1016/j.ygcen.2018.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 01/29/2023]
Abstract
We examined the effects of α-melanocyte-stimulating hormone (α-MSH) on bone metabolism using regenerating goldfish scales. Normally developed scales on the bodies of goldfish were removed to allow the regeneration of scales under anesthesia. Thereafter, the influence of α-MSH on the regeneration of goldfish scales was investigated in vivo. In brief, α-MSH was injected at a low dose (0.1 μg/g body weight) or a high dose (1 μg/g body weight) into goldfish every other day. Ten days after removing the scales, we collected regenerating scales and analyzed osteoblastic and osteoclastic activities as respective marker enzyme (alkaline phosphatase for osteoblasts, tartrate-resistant acid phosphatase for osteoclasts) activity in the regenerating scales as well as plasma calcium levels. At both doses, osteoblastic and osteoclastic activities in the regenerating scales increased significantly. Plasma calcium concentrations in the α-MSH-treated group (high doses) were significantly higher than those in the control group. Next, in vitro experiments were performed to confirm the results of in vivo experiments. In the cultured regenerating scales, osteoblastic and osteoclastic activities significantly increased with α-MSH (10-7 and 10-6 M) treatment. In addition, real-time PCR analysis indicated that osteoclastogenesis in α-MSH-treated scales was induced by the receptor activator of the NF-κB/receptor activator of the NF-κB ligand/osteoprotegerin pathway. Furthermore, we found that α-MSH receptors (melanocortin receptors 4 and 5) were detected in the regenerating scales. Thus, in teleosts, we are the first to demonstrate that α-MSH functions in bone metabolism and promotes bone resorption via melatonin receptors 4 and/or 5.
Collapse
Affiliation(s)
- Hidenori Ishizu
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan
| | - Toshio Sekiguchi
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan
| | - Takahiro Ikari
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan
| | - Kei-Ichiro Kitamura
- Department of Clinical Laboratory Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, Kodatsuno, Ishikawa 920-0942, Japan
| | - Yoichiro Kitani
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan
| | - Masato Endo
- Department of Marine Biosciences, Division of Marine Science, Tokyo University of Marine Science and Technology, Minato-ku, Tokyo 108-8477, Japan
| | - Makoto Urata
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan; Institute of Noto SATOUMI Education Research, Noto-cho, Ishikawa 927-0553, Japan
| | - Yasuko Kinoshita
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan; Institute of Noto SATOUMI Education Research, Noto-cho, Ishikawa 927-0553, Japan
| | - Atsuhiko Hattori
- Department of Biology, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, Ichikawa, Chiba 272-0827, Japan
| | - Ajai K Srivastav
- Department of Zoology, D.D.U. Gorakhpur University, Gorakhpur 273-009, India
| | - Hiroyuki Mishima
- Department of Dental Engineering, Tsurumi University School of Dental Medicine, Yokohama, Kanagawa 230-8501, Japan
| | - Kanta Mizusawa
- School of Marine Biosciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Akiyoshi Takahashi
- School of Marine Biosciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Nobuo Suzuki
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Division of Marine Environmental Studies, Kanazawa University, Noto-cho, Ishikawa 927-0553, Japan.
| |
Collapse
|
41
|
Liang L, Davis PV, Dores MR, Dores RM. The melanocortin-2 receptor of the rainbow trout: Identifying a role for critical positions in transmembrane domain 4, extracellular loop 2, and transmembrane domain 5 in the activation of rainbow trout MC2R. Gen Comp Endocrinol 2018; 257:161-167. [PMID: 28495271 DOI: 10.1016/j.ygcen.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 11/25/2022]
Abstract
The activation of either teleost or tetrapod melanocortin-2 receptor (MC2R) orthologs requires interaction between the HFRW motif and R/KKRRP motif in the primary sequence of ACTH, and two corresponding sites on the melanocortin 2 receptor. While the HFRW contact site on MC2R appears to involve residues in TM2, TM3, and TM6, several studies on human MC2R point to the EC2/TM5 region of MC2R as a possible location for the R/KKRRP contact site. In this study nineteen single-alanine mutants of rainbow trout (rt) MC2R were made beginning at V153 in TM4, at all positions in EC2 (extracellular loop 2), to F175 in TM5. For twelve of these alanine mutants (i.e., V153, G155, C162, D163, T165, V166, I167, H169, F170, H172, V173, L174), alanine substitution did not have a statistically significant effect on activation of the receptor. For four of these alanine mutations (i.e., V157, M158, F161, K168), while the negative shift in ligand sensitivity was statistically significant, the magnitude of the negative shift in activation was fivefold or less. However, for substitution at V159 in TM4 (negative shift in activation: 110 fold), F171 in TM5 (negative shift in activation: 48-fold), and F175 in TM5 (negative shift in activation: 100 fold), the effect on activation was both statistically significant and may be physiologically relevant. To support this conclusion, a triple alanine mutant of rtMC2R (V159/A, F171/A, F175/A), and this mutant receptor could not be activated by ACTH at concentrations as high as 10-6M. A Cell Surface ELISA analysis indicated that the trafficking of the triple alanine mutant rtMC2R to the plasma membrane was not impaired by the alanine substitutions. Collectively, these observations point to a critical role for TM4 and TM5 in the activation of the rainbow trout melanocortin-2 receptor.
Collapse
Affiliation(s)
- Liang Liang
- University of Denver, Department of Biological Sciences, Denver, CO, USA
| | - Perry V Davis
- University of Denver, Department of Biological Sciences, Denver, CO, USA
| | - Michael R Dores
- Hofstra University, Biology Department, Hempstead, NY 11549, USA
| | - Robert M Dores
- University of Denver, Department of Biological Sciences, Denver, CO, USA.
| |
Collapse
|
42
|
Thomas AL, Maekawa F, Kawashima T, Sakamoto H, Sakamoto T, Davis P, Dores RM. Analyzing the effects of co-expression of chick (Gallus gallus) melanocortin receptors with either chick MRAP1 or MRAP2 in CHO cells on sensitivity to ACTH(1-24) or ACTH(1-13)NH 2: Implications for the avian HPA axis and avian melanocortin circuits in the hypothalamus. Gen Comp Endocrinol 2018; 256:50-56. [PMID: 28888694 DOI: 10.1016/j.ygcen.2017.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/29/2017] [Accepted: 09/03/2017] [Indexed: 10/18/2022]
Abstract
In order to better understand the roles that melanocortin receptors (cMCRs) and melanocortin-2 receptor accessory proteins (cMRAP1 and cMRAP2) play in the HPA axis and hypothalamus, adrenal gland and hypothalamus mRNA from 1day-old white leghorn chicks (Gallus gallus), were analyzed by real-time PCR. mRNA was also made for kidney, ovary, and liver. Mrap1 mRNA could be detected in adrenal tissue, but not in any of the other tissues, and mrap2 mRNA was also detected in the adrenal gland. Finally, all five melanocortin receptors mRNAs could be detected in the adrenal gland; mc2r and mc5r mRNAs were the most abundant. To evaluate any potential interactions between MRAP1 and the MCRs that may occur in adrenal cells, individual chick mcr cDNA constructs were transiently expressed in CHO cells either in the presence or absence of a chick mrap1 cDNA, and the transfected cells were stimulated with hACTH(1-24) at concentrations ranging from 10-13M to 10-6M. As expected, MC2R required co-expression with MRAP1 for functional expression; whereas, co-expression of cMC3R with cMRAP1 had no statistically significant effect on sensitivity to hACTH(1-24). However, co-expression of MC4R and MC5R with MRAP1, increased sensitivity for ACTH(1-24) by approximately 35 fold and 365 fold, respectively. However, co-expressing of cMRAP2 with these melanocortin receptors had no effect on sensitivity to hACTH(1-24). Since the real-time PCR analysis detected mrap2 mRNA and mc4r mRNA in the hypothalamus, the interaction between cMC4R and cMRAP2 with respect to sensitivity to ACTH(1-13)NH2 stimulation was also evaluated. However, no effect, either positive or negative, was observed. Finally, the highest levels of mc5r mRNA were detected in liver cells. This observation raises the possibility that in one-day old chicks, activation of the HPA axis may also involve a physiological response from liver cells.
Collapse
|
43
|
Stein MB, Ware EB, Mitchell C, Chen C, Borja S, Cai T, Dempsey CL, Fullerton CS, Gelernter J, Heeringa SG, Jain S, Kessler RC, Naifeh JA, Nock MK, Ripke S, Sun X, Beckham JC, Kimbrel NA, Ursano RJ, Smoller JW. Genomewide association studies of suicide attempts in US soldiers. Am J Med Genet B Neuropsychiatr Genet 2017; 174:786-797. [PMID: 28902444 PMCID: PMC5685938 DOI: 10.1002/ajmg.b.32594] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/11/2017] [Indexed: 12/19/2022]
Abstract
Suicide is a global public health problem with particular resonance for the US military. Genetic risk factors for suicidality are of interest as indicators of susceptibility and potential targets for intervention. We utilized population-based nonclinical cohorts of US military personnel (discovery: N = 473 cases and N = 9778 control subjects; replication: N = 135 cases and N = 6879 control subjects) and a clinical case-control sample of recent suicide attempters (N = 51 cases and N = 112 control subjects) to conduct GWAS of suicide attempts (SA). Genomewide association was evaluated within each ancestral group (European-, African-, Latino-American) and study using logistic regression models. Meta-analysis of the European ancestry discovery samples revealed a genomewide significant locus in association with SA near MRAP2 (melanocortin 2 receptor accessory protein 2) and CEP162 (centrosomal protein 162); 12 genomewide significant SNPs in the region; peak SNP rs12524136-T, OR = 2.88, p = 5.24E-10. These findings were not replicated in the European ancestry subsamples of the replication or suicide attempters samples. However, the association of the peak SNP remained significant in a meta-analysis of all studies and ancestral subgroups (OR = 2.18, 95%CI 1.70, 2.80). Polygenic risk score (PRS) analyses showed some association of SA with bipolar disorder. The association with SNPs encompassing MRAP2, a gene expressed in brain and adrenal cortex and involved in neural control of energy homeostasis, points to this locus as a plausible susceptibility gene for suicidality that should be further studied. Larger sample sizes will be needed to confirm and extend these findings.
Collapse
Affiliation(s)
- Murray B. Stein
- Department of PsychiatryUniversity of California San Diego and VA San Diego Healthcare SystemLa JollaCalifornia
- Department of Family Medicine and Public HealthUniversity of California San DiegoLa JollaCalifornia
| | - Erin B. Ware
- Institute for Social ResearchUniversity of MichiganAnn ArborMichigan
| | - Colter Mitchell
- Institute for Social ResearchUniversity of MichiganAnn ArborMichigan
| | - Chia‐Yen Chen
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic MedicineMassachusetts General HospitalBostonMassachusetts
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
| | - Susan Borja
- National Institute of Mental HealthBethesdaMaryland
| | - Tianxi Cai
- Harvard T.H. Chan School of Public HealthBostonMassachusetts
| | | | | | - Joel Gelernter
- Departments of Psychiatry, Genetics, and NeurobiologyYale UniversityNew HavenConnecticut
| | | | - Sonia Jain
- Department of Family Medicine and Public HealthUniversity of California San DiegoLa JollaCalifornia
| | - Ronald C. Kessler
- Department of Health Care PolicyHarvard Medical SchoolBostonMassachusetts
| | - James A. Naifeh
- Uniformed Services University of the Health SciencesBethesdaMaryland
| | - Matthew K. Nock
- Department of PsychologyHarvard UniversityCambridgeMassachusetts
| | - Stephan Ripke
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
| | - Xiaoying Sun
- Department of Family Medicine and Public HealthUniversity of California San DiegoLa JollaCalifornia
| | - Jean C. Beckham
- Durham Veterans Affairs Health Care System and Duke University Health SystemDurhamNorth Carolina
- VA MIRECCDurhamNorth Carolina
| | - Nathan A. Kimbrel
- Durham Veterans Affairs Health Care System and Duke University Health SystemDurhamNorth Carolina
- VA MIRECCDurhamNorth Carolina
| | - Robert J Ursano
- Uniformed Services University of the Health SciencesBethesdaMaryland
| | - Jordan W. Smoller
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic MedicineMassachusetts General HospitalBostonMassachusetts
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
| | | |
Collapse
|
44
|
Palmer D, Gonçalves JPL, V Hansen L, Wu B, Hald H, Schoffelen S, Diness F, Le Quement ST, Nielsen TE, Meldal M. Click-Chemistry-Mediated Synthesis of Selective Melanocortin Receptor 4 Agonists. J Med Chem 2017; 60:8716-8730. [PMID: 28972753 DOI: 10.1021/acs.jmedchem.7b00353] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The melanocortin receptor 4 (MC4R) subtype of the melanocortin receptor family is a target for therapeutics to ameliorate metabolic dysfunction. Endogenous MC4R agonists possess a critical pharmacophore (HFRW), and cyclization of peptide agonists often enhances potency. Thus, 17 cyclized peptides were synthesized by solid phase click chemistry to develop novel, potent, selective MC4R agonists. Using cAMP measurements and a transcriptional reporter assay, we observed that several constrained agonists generated by a cycloaddition reaction displayed high selectivity (223- to 467-fold) toward MC4R over MC3R and MC5R receptor subtypes without compromising agonist potency. Significant variation was also observed between the EC50 values for the two assays, with robust levels of reporter expression measured at lower concentrations than those effecting appreciable increases in cAMP levels for the majority of the compounds tested. Collectively, we characterized significant elements that modulate the activity of the core pharmacophore for MC4R and provide a rationale for careful assay selection for agonist screening.
Collapse
Affiliation(s)
- Daniel Palmer
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Juliana P L Gonçalves
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Louise V Hansen
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Boqian Wu
- Aquaporin A/S , Ole Maaløes Vej 3, 2200 Copenhagen, Denmark
| | - Helle Hald
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Sanne Schoffelen
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Frederik Diness
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| | | | - Thomas E Nielsen
- Protein & Peptide Chemistry, Novo Nordisk A/S , Novo Nordisk Park, 2760 Måløv, Denmark.,Department of Immunology and Microbiology, University of Copenhagen , Blegdamsvej 3B, 2200 Copenhagen, Denmark.,Singapore Centre on Environmental Life Sciences Engineering, Nanyang Technological University , 60 Nanyang Drive, SG 637551, Singapore
| | - Morten Meldal
- CECB, Department of Chemistry, University of Copenhagen , Universitetsparken 5, 2100 Copenhagen, Denmark
| |
Collapse
|
45
|
da Fonseca ACP, Mastronardi C, Johar A, Arcos-Burgos M, Paz-Filho G. Genetics of non-syndromic childhood obesity and the use of high-throughput DNA sequencing technologies. J Diabetes Complications 2017; 31:1549-1561. [PMID: 28735903 DOI: 10.1016/j.jdiacomp.2017.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Childhood obesity is a serious public health problem associated with the development of several chronic diseases, such as type 2 diabetes mellitus, dyslipidemia, and hypertension. The elevated prevalence of obesity is mostly due to inadequate diet and lifestyle, but it is also influenced by genetic factors. OBJECTIVES To review recent advances in the field of the genetics of obesity. We summarize the list of genes associated with the rare non-syndromic forms of obesity, and explain their function. Furthermore, we discuss the technologies that are available for the genetic diagnosis of obesity. RESULTS Several studies reported that single gene variants cause Mendelian forms of obesity, determined by mutations of major effect in single genes. Rare, non-syndromic forms of obesity are a result of loss-of-function mutations in genes that act on the development and function of the hypothalamus or the leptin-melanocortin pathway. These variants disrupt enzymes and receptors that play a role in energy homeostasis, resulting in severe early-onset obesity and endocrine dysfunctions. Different approaches and technologies have been used to understand the genetic background of obesity. Currently, whole genome and whole exome sequencing are important diagnostic tools to identify new genes and variants associated with severe obesity, but other approaches are also useful at individual or population levels, such as linkage analysis, candidate gene sequencing, chromosomal microarray analysis, and genome-wide association studies. CONCLUSIONS The understanding of the genetic causes of obesity and the usefulness and limitations of the genetic diagnostic approaches can contribute to the development of new personalized therapeutic targets against obesity.
Collapse
Affiliation(s)
| | - Claudio Mastronardi
- Institute of Translational Medicine, Universidad del Rosario, Bogota, Colombia
| | - Angad Johar
- Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Australia.
| | | | - Gilberto Paz-Filho
- Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Australia.
| |
Collapse
|
46
|
Zhang J, Li X, Zhou Y, Cui L, Li J, Wu C, Wan Y, Li J, Wang Y. The interaction of MC3R and MC4R with MRAP2, ACTH, α-MSH and AgRP in chickens. J Endocrinol 2017; 234:155-174. [PMID: 28512117 DOI: 10.1530/joe-17-0131] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 02/05/2023]
Abstract
The interaction of melanocortin-4 (MC4R) and melanocortin-3 (MC3R) receptors with proopiomelanocortin (POMC)-derived peptides (e.g. α-MSH), agouti-related protein (AgRP) and melanocortin-2 receptor accessory protein 2 (MRAP2) is suggested to play critical roles in energy balance of vertebrates. However, evidence on their interaction in birds remains scarce. Our study aims to reveal their interaction in chickens and the results showed that (1) chicken (c-)MC3R and cMC4R expressed in Chinese hamster ovary (CHO) cells can be activated by α-MSH and ACTH1-39 equipotently, monitored by a pGL3-CRE-luciferase reporter system; (2) cMC3R and cMC4R, when co-expressed with cMRAP2 (or cMRAP, a cMRAP2 homolog), show increased sensitivity to ACTH treatment and thus likely act as ACTH-preferring receptors, and the interaction between cMC3R/cMC4R and cMRAP2 was demonstrated by co-immunoprecipitation assay; (3) both cMC3R and cMC4R display constitutive activity when expressed in CHO cells, as monitored by dual-luciferase reporter assay, and cMRAP2 (and cMRAP) can modulate their constitutive activity; (4) AgRP inhibits the constitutive activity of cMC3R/cMC4R, and it also antagonizes ACTH/α-MSH action on cMC4R/cMC3R, indicating that AgRP functions as the inverse agonist and antagonist for both receptors. These findings, together with the co-expression of cMC4R, cMC3R, cMRAP2, cAgRP and cPOMC in chicken hypothalamus detected by quantitative real-time PCR, suggest that within the hypothalamus, α-MSH/ACTH, AgRP and MRAP2 may interact at the MC4R(/MC3R) interface to control energy balance. Furthermore, our data provide novel proof for the involvement of MRAP2 (and MRAP) in fine-tuning the constitutive activity and ligand sensitivity and selectivity of both MC3R and MC4R in vertebrates.
Collapse
MESH Headings
- Adrenocorticotropic Hormone/genetics
- Adrenocorticotropic Hormone/metabolism
- Agouti-Related Protein/genetics
- Agouti-Related Protein/metabolism
- Amino Acid Sequence
- Animals
- CHO Cells
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Chickens/genetics
- Chickens/metabolism
- Cloning, Molecular
- Cricetinae
- Cricetulus
- DNA, Complementary
- Gene Expression Regulation/physiology
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 3/metabolism
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- alpha-MSH/genetics
- alpha-MSH/metabolism
Collapse
Affiliation(s)
- Jiannan Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Xin Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Yawei Zhou
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Lin Cui
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Jing Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Chenlei Wu
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Yiping Wan
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Juan Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Yajun Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of EducationCollege of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
47
|
Fridmanis D, Roga A, Klovins J. ACTH Receptor (MC2R) Specificity: What Do We Know About Underlying Molecular Mechanisms? Front Endocrinol (Lausanne) 2017; 8:13. [PMID: 28220105 PMCID: PMC5292628 DOI: 10.3389/fendo.2017.00013] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/16/2017] [Indexed: 11/13/2022] Open
Abstract
Coincidentally, the release of this Research Topic in Frontiers in Endocrinology takes place 25 years after the discovery of the adrenocorticotropic hormone receptor (ACTHR) by Mountjoy and colleagues. In subsequent years, following the discovery of other types of mammalian melanocortin receptors (MCRs), ACTHR also became known as melanocortin type 2 receptor (MC2R). At present, five types of MCRs have been reported, all of which share significant sequence similarity at the amino acid level, and all of which specifically bind melanocortins (MCs)-a group of biologically active peptides generated by proteolysis of the proopiomelanocortin precursor. All MCs share an identical -H-F-R-W- pharmacophore sequence. α-Melanocyte-stimulating hormone (α-MSH) and adrenocorticotropic hormone (ACTH) are the most extensively studied MCs and are derived from the same region. Essentially, α-MSH is formed from the first 13 amino acid residues of ACTH. ACTHR is unique among MCRs because it binds one sole ligand-ACTH, which makes it a very attractive research object for molecular pharmacologists. However, much research has failed, and functional studies of this receptor are lagging behind other MCRs. The reason for these difficulties has already been outlined by Mountjoy and colleagues in their publication on ACTHR coding sequence discovery where the Cloudman S91 melanoma cell line was used for receptor expression because it was a "more sensitive assay system." Subsequent work showed that ACTHR could be successfully expressed only in endogenous MCR-expressing cell lines, since in other cell lines it is retained within the endoplasmic reticulum. The resolution of this methodological problem came in 2005 with the discovery of melanocortin receptor accessory protein, which is required for the formation of functionally active ACTHR. The decade that followed this discovery was filled with exciting research that provided insight into the molecular mechanisms underlying the action of ACTHR. The purpose of this review is to summarize the advances in this fascinating research field.
Collapse
Affiliation(s)
| | - Ance Roga
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| |
Collapse
|
48
|
Navarro M. The Role of the Melanocortin System in Drug and Alcohol Abuse. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:121-150. [DOI: 10.1016/bs.irn.2017.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
49
|
Clark AJL, Lowry P. 60 YEARS OF POMC: POMC: the consummate peptide hormone precursor. J Mol Endocrinol 2016; 56:E1-2. [PMID: 27273100 DOI: 10.1530/jme-16-0016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 01/28/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Adrian J L Clark
- Centre for EndocrinologyWilliam Harvey Research Institute, Queen Mary University of London, London, UK
| | - Philip Lowry
- Emeritus Professor School of Biological SciencesThe University of Reading, Reading, UK
| |
Collapse
|
50
|
Gallo-Payet N. 60 YEARS OF POMC: Adrenal and extra-adrenal functions of ACTH. J Mol Endocrinol 2016; 56:T135-56. [PMID: 26793988 DOI: 10.1530/jme-15-0257] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 01/21/2016] [Indexed: 01/27/2023]
Abstract
The pituitary adrenocorticotropic hormone (ACTH) plays a pivotal role in homeostasis and stress response and is thus the major component of the hypothalamo-pituitary-adrenal axis. After a brief summary of ACTH production from proopiomelanocortin (POMC) and on ACTH receptor properties, the first part of the review covers the role of ACTH in steroidogenesis and steroid secretion. We highlight the mechanisms explaining the differential acute vs chronic effects of ACTH on aldosterone and glucocorticoid secretion. The second part summarizes the effects of ACTH on adrenal growth, addressing its role as either a mitogenic or a differentiating factor. We then review the mechanisms involved in steroid secretion, from the classical Cyclic adenosine monophosphate second messenger system to various signaling cascades. We also consider how the interaction between the extracellular matrix and the cytoskeleton may trigger activation of signaling platforms potentially stimulating or repressing the steroidogenic potency of ACTH. Finally, we consider the extra-adrenal actions of ACTH, in particular its role in differentiation in a variety of cell types, in addition to its known lipolytic effects on adipocytes. In each section, we endeavor to correlate basic mechanisms of ACTH function with the pathological consequences of ACTH signaling deficiency and of overproduction of ACTH.
Collapse
Affiliation(s)
- Nicole Gallo-Payet
- Division of EndocrinologyDepartment of Medicine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada Division of EndocrinologyDepartment of Medicine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|