1
|
Chen A, Li J, Wang H, Zhao P. Identification and Expression Profile of NCED Genes in Arachis hypogaea L. during Drought Stress. Int J Mol Sci 2024; 25:5564. [PMID: 38791604 PMCID: PMC11122452 DOI: 10.3390/ijms25105564] [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: 03/17/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Peanut (Arachis hypogaea L.) is an important crop that provides essential proteins and oils for human and animal consumption. 9-cis-epoxycarotenoid dioxygenase (NCED) have been found can play a vital role in abscisic acid (ABA) biosynthesis and may be a response to drought stress. Until now, in Arachis hypogaea, no information about the NCED gene family has been reported and the importance of NCED-related drought tolerance is unclear. In this study, eight NCED genes in Arachis hypogaea, referred to as AhNCEDs, are distributed across eight chromosomes, with duplication events in AhNCED1 and AhNCED2, AhNCED3 and AhNCED4, and AhNCED6 and AhNCED7. Comparative analysis revealed that NCED genes are highly conserved among plant species, including Pisum sativum, Phaseolus vulgaris, Glycine max, Arabidopsis thaliana, Gossypium hirsutum, and Oryza sativa. Further promoter analysis showed AhNCEDs have ABA-related and drought-inducible elements. The phenotyping of Arachis hypogaea cultivars NH5 and FH18 demonstrated that NH5 is drought-tolerant and FH18 is drought-sensitive. Transcriptome expression analysis revealed the differential regulation of AhNCEDs expression in both NH5 and FH18 cultivars under drought stress. Furthermore, compared to the Arachis hypogaea cultivar FH18, the NH5 exhibited a significant upregulation of AhNCED1/2 expression under drought. To sum up, this study provides an insight into the drought-related AhNCED genes, screened out the potential candidates to regulate drought tolerance and ABA biosynthesis in Arachis hypogaea.
Collapse
Affiliation(s)
- Ao Chen
- Guangzhou Dublin International College of Life Sciences and Technology, South China Agricultural University, Guangzhou 510642, China;
| | - Jingyan Li
- College of Horticulture, South China Agricultural University, Guangzhou 510642, China; (J.L.); (H.W.)
| | - Heping Wang
- College of Horticulture, South China Agricultural University, Guangzhou 510642, China; (J.L.); (H.W.)
| | - Puyan Zhao
- College of Horticulture, South China Agricultural University, Guangzhou 510642, China; (J.L.); (H.W.)
| |
Collapse
|
2
|
Donizetti A, Calicchio M, Romano MZ, Rosati L, Turco M, Carrese AM, del Gaudio R, Ferrandino I, Aniello F. Expression of Insl3 Protein in Adult Danio rerio. Int J Mol Sci 2024; 25:5419. [PMID: 38791457 PMCID: PMC11122137 DOI: 10.3390/ijms25105419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/04/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Insulin-like peptide 3 (INSL3) is a biomarker for Leydig cells in the testes of vertebrates, and it is principally involved in spermatogenesis through specific binding with the RXFP2 receptor. This study reports the insl3 gene transcript and the Insl3 prepropeptide expression in both non-reproductive and reproductive tissues of Danio rerio. An immunohistochemistry analysis shows that the hormone is present at a low level in the Leydig cells and germ cells at all stages of Danio rerio testis differentiation. Considering that the insl3 gene is transcribed in Leydig cells, our results highlight an autocrine and paracrine function of this hormone in the Danio rerio testis, adding new information on the Insl3 mode of action in reproduction. We also show that Insl3 and Rxfp2 belonging to Danio rerio and other vertebrate species share most of the amino acid residues involved in the ligand-receptor interaction and activation, suggesting a conserved mechanism of action during vertebrate evolution.
Collapse
Affiliation(s)
- Aldo Donizetti
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Mauro Calicchio
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Maria Zelinda Romano
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Luigi Rosati
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Manuela Turco
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Anna Maria Carrese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Rosanna del Gaudio
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Ida Ferrandino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| | - Francesco Aniello
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (M.C.); (L.R.); (M.T.); (A.M.C.); (R.d.G.); (I.F.)
| |
Collapse
|
3
|
Neagu AN, Whitham D, Bruno P, Arshad A, Seymour L, Morrissiey H, Hukovic AI, Darie CC. Onco-Breastomics: An Eco-Evo-Devo Holistic Approach. Int J Mol Sci 2024; 25:1628. [PMID: 38338903 PMCID: PMC10855488 DOI: 10.3390/ijms25031628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Known as a diverse collection of neoplastic diseases, breast cancer (BC) can be hyperbolically characterized as a dynamic pseudo-organ, a living organism able to build a complex, open, hierarchically organized, self-sustainable, and self-renewable tumor system, a population, a species, a local community, a biocenosis, or an evolving dynamical ecosystem (i.e., immune or metabolic ecosystem) that emphasizes both developmental continuity and spatio-temporal change. Moreover, a cancer cell community, also known as an oncobiota, has been described as non-sexually reproducing species, as well as a migratory or invasive species that expresses intelligent behavior, or an endangered or parasite species that fights to survive, to optimize its features inside the host's ecosystem, or that is able to exploit or to disrupt its host circadian cycle for improving the own proliferation and spreading. BC tumorigenesis has also been compared with the early embryo and placenta development that may suggest new strategies for research and therapy. Furthermore, BC has also been characterized as an environmental disease or as an ecological disorder. Many mechanisms of cancer progression have been explained by principles of ecology, developmental biology, and evolutionary paradigms. Many authors have discussed ecological, developmental, and evolutionary strategies for more successful anti-cancer therapies, or for understanding the ecological, developmental, and evolutionary bases of BC exploitable vulnerabilities. Herein, we used the integrated framework of three well known ecological theories: the Bronfenbrenner's theory of human development, the Vannote's River Continuum Concept (RCC), and the Ecological Evolutionary Developmental Biology (Eco-Evo-Devo) theory, to explain and understand several eco-evo-devo-based principles that govern BC progression. Multi-omics fields, taken together as onco-breastomics, offer better opportunities to integrate, analyze, and interpret large amounts of complex heterogeneous data, such as various and big-omics data obtained by multiple investigative modalities, for understanding the eco-evo-devo-based principles that drive BC progression and treatment. These integrative eco-evo-devo theories can help clinicians better diagnose and treat BC, for example, by using non-invasive biomarkers in liquid-biopsies that have emerged from integrated omics-based data that accurately reflect the biomolecular landscape of the primary tumor in order to avoid mutilating preventive surgery, like bilateral mastectomy. From the perspective of preventive, personalized, and participatory medicine, these hypotheses may help patients to think about this disease as a process governed by natural rules, to understand the possible causes of the disease, and to gain control on their own health.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Aneeta Arshad
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Logan Seymour
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Angiolina I. Hukovic
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| |
Collapse
|
4
|
Lv C, Zheng H, Jiang B, Ren Q, Zhang J, Zhang X, Li J, Wang Y. Characterization of relaxin 3 and its receptors in chicken: Evidence for relaxin 3 acting as a novel pituitary hormone. Front Physiol 2022; 13:1010851. [DOI: 10.3389/fphys.2022.1010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
Mammalian relaxin (RLN) family peptides binding their receptors (RXFPs) play a variety of roles in many physiological processes, such as reproduction, stress, appetite regulation, and energy balance. In birds, although two relaxin family peptides (RLN3 and INSL5) and four receptors (RXFP1, RXFP2, RXFP2-like, and RXFP3) were predicated, their sequence features, signal properties, tissue distribution, and physiological functions remain largely unknown. In this study, using chickens as the experimental model, we cloned the cDNA of the cRLN3 gene and two receptor (cRXFP1 and cRXFP3) genes. Using cell-based luciferase reporter assays, we demonstrate that cRLN3 is able to activate both cRXFP1 and cRXFP3 for downstream signaling. cRXFP1, rather than cRXFP3, is a cognate receptor for cRLN3, which is different from the mammals. Tissue distribution analyses reveal that cRLN3 is highly expressed in the pituitary with lower abundance in the hypothalamus and ovary of female chicken, together with the detection that cRLN3 co-localizes with pituitary hormone genes LHB/FSHB/GRP/CART and its expression is tightly regulated by hypothalamic factors (GnRH and CRH) and sex steroid hormone (E2). The present study supports that cRLN3 may function as a novel pituitary hormone involving female reproduction.
Collapse
|
5
|
Blasiak A, Gugula A, Gundlach AL, Olucha-Bordonau FE, Aniello F, Donizetti A. Relaxin ligand/receptor systems in the developing teleost fish brain: Conserved features with mammals and a platform to address neuropeptide system functions. Front Mol Neurosci 2022; 15:984524. [PMID: 36277494 PMCID: PMC9580368 DOI: 10.3389/fnmol.2022.984524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
The relaxins (RLNs) are a group of peptide hormone/neuromodulators that can regulate a wide range of physiological processes ranging from reproduction to brain function. All the family members have originated from a RLN3-like ancestor via different rounds of whole genome and gene specific duplications during vertebrate evolution. In mammals, including human, the divergence of the different family members and the emergence of new members led to the acquisition of specific functions for the various relaxin family peptide and associated receptor genes. In particular, in mammals, it was shown, that the role of RLN3 is correlated to the modulation of arousal, stress responses, emotion, social recognition, and other brain functions, positioning this gene/peptide as a potential therapeutic target for neuropsychiatric disorders. This review highlights the evolutionary conservation of relaxin family peptide and receptor gene expression and their associated brain neural circuits. In the zebrafish, the expression pattern of the different relaxin family members has specific features that are conserved in higher species, including a likely similar functional role for the ancestral RLN3-like gene. The use of different model organisms, particularly the zebrafish, to explore the diversification and conservation of relaxin family ligands and receptor systems, provides a relatively high-throughput platform to identify their specific conserved or differential neuromodulatory roles in higher species including human.
Collapse
Affiliation(s)
- Anna Blasiak
- Department of Neurophysiology and Chronobiology, Jagiellonian University, Krakow, Poland
| | - Anna Gugula
- Department of Neurophysiology and Chronobiology, Jagiellonian University, Krakow, Poland
| | - Andrew L. Gundlach
- Florey Department of Neuroscience and Mental Health, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | | | - Francesco Aniello
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Aldo Donizetti
- Department of Biology, University of Naples Federico II, Naples, Italy
- *Correspondence: Aldo Donizetti,
| |
Collapse
|
6
|
Crespo D, Assis LHC, Zhang YT, Safian D, Furmanek T, Skaftnesmo KO, Norberg B, Ge W, Choi YC, den Broeder MJ, Legler J, Bogerd J, Schulz RW. Insulin-like 3 affects zebrafish spermatogenic cells directly and via Sertoli cells. Commun Biol 2021; 4:204. [PMID: 33589679 PMCID: PMC7884674 DOI: 10.1038/s42003-021-01708-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023] Open
Abstract
Pituitary hormones can use local signaling molecules to regulate target tissue functions. In adult zebrafish testes, follicle-stimulating hormone (Fsh) strongly increases the production of insulin-like 3 (Insl3), a Leydig cell-derived growth factor found in all vertebrates. Little information is available regarding Insl3 function in adult spermatogenesis. The Insl3 receptors Rxfp2a and 2b were expressed by type A spermatogonia and Sertoli and myoid cells, respectively, in zebrafish testis tissue. Loss of insl3 increased germ cell apoptosis in males starting at 9 months of age, but spermatogenesis appeared normal in fully fertile, younger adults. Insl3 changed the expression of 409 testicular genes. Among others, retinoic acid (RA) signaling was up- and peroxisome proliferator-activated receptor gamma (Pparg) signaling was down-regulated. Follow-up studies showed that RA and Pparg signaling mediated Insl3 effects, resulting in the increased production of differentiating spermatogonia. This suggests that Insl3 recruits two locally active nuclear receptor pathways to implement pituitary (Fsh) stimulation of spermatogenesis.
Collapse
Affiliation(s)
- Diego Crespo
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Present Address: Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Luiz H. C. Assis
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Yu Ting Zhang
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, PR China ,grid.449133.80000 0004 1764 3555Present Address: Institute of Oceanography, Minjiang University, Fuzhou, PR China
| | - Diego Safian
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Present Address: Experimental Zoology Group and Aquaculture and Fisheries Group, Department of Animal Science, Wageningen University, Wageningen, The Netherlands
| | - Tomasz Furmanek
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Kai Ove Skaftnesmo
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Birgitta Norberg
- grid.10917.3e0000 0004 0427 3161Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Wei Ge
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Yung-Ching Choi
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Marjo J. den Broeder
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Juliette Legler
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan Bogerd
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W. Schulz
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
7
|
Irwin DM. Evolution of the Insulin Gene: Changes in Gene Number, Sequence, and Processing. Front Endocrinol (Lausanne) 2021; 12:649255. [PMID: 33868177 PMCID: PMC8051583 DOI: 10.3389/fendo.2021.649255] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/01/2021] [Indexed: 02/05/2023] Open
Abstract
Insulin has not only made major contributions to the field of clinical medicine but has also played central roles in the advancement of fundamental molecular biology, including evolution. Insulin is essential for the health of vertebrate species, yet its function has been modified in species-specific manners. With the advent of genome sequencing, large numbers of insulin coding sequences have been identified in genomes of diverse vertebrates and have revealed unexpected changes in the numbers of genes within genomes and in their sequence that likely impact biological function. The presence of multiple insulin genes within a genome potentially allows specialization of an insulin gene. Discovery of changes in proteolytic processing suggests that the typical two-chain hormone structure is not necessary for all of inulin's biological activities.
Collapse
Affiliation(s)
- David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
- *Correspondence: David M. Irwin,
| |
Collapse
|
8
|
Mita M, Osugi T, Matsubara S, Kawada T, Satake H, Katayama H. A relaxin-like gonad-stimulating peptide identified from the starfish Astropecten scoparius. Mol Reprod Dev 2020; 88:34-42. [PMID: 33244845 DOI: 10.1002/mrd.23444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 11/12/2022]
Abstract
A relaxin-like gonad-stimulating peptide (RGP) in starfish was the first identified invertebrate gonadotropin responsible for final gamete maturation. An RGP ortholog was newly identified from Astropecten scoparius of the order Paxillosida. The A. scoparius RGP (AscRGP) precursor is encoded by a 354 base pair open reading frame and is a 118 amino acid (aa) protein consisting of a signal peptide (26 aa), B-chain (21 aa), C-peptide (47 aa), and A-chain (24 aa). There are three putative processing sites (Lys-Arg) between the B-chain and C-peptide, between the C-peptide and A-chain, and within the C-peptide. This structural organization revealed that the mature AscRGP is composed of A- and B-chains with two interchain disulfide bonds and one intrachain disulfide bond. The C-terminal residues of the B-chain are Gln-Gly-Arg, which is a potential substrate for formation of an amidated C-terminal Gln residue. Non-amidated (AscRGP-GR) and amidated (AscRGP-NH2 ) peptides were chemically synthesized and their effect on gamete shedding activity was examined using A. scoparius ovaries. Both AscRGP-GR and AscRGP-NH2 induced oocyte maturation and ovulation in similar dose-dependent manners. This is the first report on a C-terminally amidated functional RGP. Collectively, these results suggest that AscRGP-GR and AscRGP-NH2 act as a natural gonadotropic hormone in A. scoparius.
Collapse
Affiliation(s)
- Masatoshi Mita
- Department of Biochemistry, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Tomohiro Osugi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Soraku-gun, Kyoto, Japan
| | - Shin Matsubara
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Soraku-gun, Kyoto, Japan
| | - Tsuyoshi Kawada
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Soraku-gun, Kyoto, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Soraku-gun, Kyoto, Japan
| | - Hidekazu Katayama
- Department of Applied Biochemistry, Tokai University, Hiratsuka, Kanagawa, Japan
| |
Collapse
|
9
|
Mita M, Matsubara S, Osugi T, Shiraishi A, Wada A, Satake H. A novel G protein-coupled receptor for starfish gonadotropic hormone, relaxin-like gonad-stimulating peptide. PLoS One 2020; 15:e0242877. [PMID: 33226996 PMCID: PMC7682835 DOI: 10.1371/journal.pone.0242877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Gonadotropic hormones play important regulatory roles in reproduction. Relaxin-like gonad-stimulating peptide (RGP) is a gonadotropin-like hormone in starfish. However, a receptor for RGP remains to be identified. Here, we describe the identification of an authentic receptor for RGP (RGPR) in the starfish, Patiria pectinifera. A binding assay using radioiodinated P. pectinifera RGP (PpeRGP) revealed that RGPR was expressed in ovarian follicle cells. A RGPR candidate was identified by homology-searching of transcriptome data of P. pectinifera follicle cells. Based on the contig sequences, a putative 947-amino acid PpeRGPR was cloned from follicle cells. Like the vertebrate relaxin family peptide receptors (RXFP 1 and 2), PpeRGPR was a G protein-coupled receptor that harbored a low-density lipoprotein-receptor class A motif and leucine-rich repeat sequences in the extracellular domain of the N-terminal region. Sf9 cells transfected with Gαq16-fused PpeRGPR activated calcium ion mobilization in response to PpeRGP, but not to RGP of another starfish Asterias amurensis, in a dose-dependent fashion. These results confirmed the species-specific reactivity of RGP and the cognate receptor. Thus, the present study provides evidence that PpeRGPR is a specific receptor for PpeRGP. To the best of our knowledge, this is the first report on the identification of a receptor for echinoderm RGP.
Collapse
Affiliation(s)
- Masatoshi Mita
- Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan
| | - Shin Matsubara
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Tomohiro Osugi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Akira Shiraishi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Azumi Wada
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| |
Collapse
|
10
|
Affiliation(s)
- Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA, 93407, USA.
| |
Collapse
|
11
|
Cartography of rhodopsin-like G protein-coupled receptors across vertebrate genomes. Sci Rep 2019; 9:7058. [PMID: 31064998 PMCID: PMC6504862 DOI: 10.1038/s41598-018-33120-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/17/2018] [Indexed: 12/26/2022] Open
Abstract
We conduct a cartography of rhodopsin-like non-olfactory G protein-coupled receptors in the Ensembl database. The most recent genomic data (releases 90–92, 90 vertebrate genomes) are analyzed through the online interface and receptors mapped on phylogenetic guide trees that were constructed based on a set of ~14.000 amino acid sequences. This snapshot of genomic data suggest vertebrate genomes to harbour 142 clades of GPCRs without human orthologues. Among those, 69 have not to our knowledge been mentioned or studied previously in the literature, of which 28 are distant from existing receptors and likely new orphans. These newly identified receptors are candidates for more focused evolutionary studies such as chromosomal mapping as well for in-depth pharmacological characterization. Interestingly, we also show that 37 of the 72 human orphan (or recently deorphanized) receptors included in this study cluster into nineteen closely related groups, which implies that there are less ligands to be identified than previously anticipated. Altogether, this work has significant implications when discussing nomenclature issues for GPCRs.
Collapse
|
12
|
Alnafea H, Vahkal B, Zelmer CK, Yegorov S, Bogerd J, Good SV. Japanese medaka as a model for studying the relaxin family genes involved in neuroendocrine regulation: Insights from the expression of fish-specific rln3 and insl5 and rxfp3/4-type receptor paralogues. Mol Cell Endocrinol 2019; 487:2-11. [PMID: 30703485 DOI: 10.1016/j.mce.2019.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 12/14/2022]
Abstract
The goal of this paper is to establish Japanese medaka (Oryzias latipes) as a model for relaxin family peptide research, particularly for studying the functions of RLN3 and INSL5, hormones playing roles in neuroendocrine regulation. Medaka, like other teleosts, retained duplicate copies of rln3, insl5 and their rxfp3/4-type receptors following fish-specific whole genome duplication (WGD) and paralogous copies of these genes may have sub-functionalised providing an intuitive model for teasing apart the pleiotropic roles of the corresponding genes in mammals. To this end, we provide experimental evidence for the expression of the relaxin family genes in medaka that had previously only been identified in-silico, confirm the gene structure of five of the ligand genes, characterise gene expression across multiple tissues and during embryonic development, perform in situ hybridization with anti-sense insl5a on embryos and in adult brain and intestinal samples, and compare these results to the data available in zebrafish. We find broad similarities but also some differences in the expression of relaxin family genes in zebrafish versus medaka, and find support for the hypothesis that the rln3a/rln3b and insl5a/insl5b paralogues have been subfunctionalized. Given that medaka has a suite of relaxin family genes more similar to other teleosts, and has retained the gene for rxfp4 (which is lost in zebrafish), our results suggest that O. latipes may be a good model for delineating the ancestral function of the relaxin family genes involved in neuroendocrine regulation.
Collapse
Affiliation(s)
- Hend Alnafea
- Department of Biology, The University of Winnipeg, Winnipeg, MB, Canada
| | - Brett Vahkal
- Department of Biology, The University of Winnipeg, Winnipeg, MB, Canada
| | - C Kellie Zelmer
- Department of Biology, The University of Winnipeg, Winnipeg, MB, Canada
| | - Sergey Yegorov
- Department of Immunology, The University of Toronto, Toronto, ON, Canada
| | - Jan Bogerd
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Sara V Good
- Department of Biology, The University of Winnipeg, Winnipeg, MB, Canada; Department of Biology, The University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
13
|
Novel Zebrafish Mono-α2,8-sialyltransferase (ST8Sia VIII): An Evolutionary Perspective of α2,8-Sialylation. Int J Mol Sci 2019; 20:ijms20030622. [PMID: 30709055 PMCID: PMC6387029 DOI: 10.3390/ijms20030622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/28/2022] Open
Abstract
The mammalian mono-α2,8-sialyltransferase ST8Sia VI has been shown to catalyze the transfer of a unique sialic acid residues onto core 1 O-glycans leading to the formation of di-sialylated O-glycosylproteins and to a lesser extent to diSia motifs onto glycolipids like GD1a. Previous studies also reported the identification of an orthologue of the ST8SIA6 gene in the zebrafish genome. Trying to get insights into the biosynthesis and function of the oligo-sialylated glycoproteins during zebrafish development, we cloned and studied this fish α2,8-sialyltransferase homologue. In situ hybridization experiments demonstrate that expression of this gene is always detectable during zebrafish development both in the central nervous system and in non-neuronal tissues. Intriguingly, using biochemical approaches and the newly developed in vitro MicroPlate Sialyltransferase Assay (MPSA), we found that the zebrafish recombinant enzyme does not synthetize diSia motifs on glycoproteins or glycolipids as the human homologue does. Using comparative genomics and molecular phylogeny approaches, we show in this work that the human ST8Sia VI orthologue has disappeared in the ray-finned fish and that the homologue described in fish correspond to a new subfamily of α2,8-sialyltransferase named ST8Sia VIII that was not maintained in Chondrichtyes and Sarcopterygii.
Collapse
|
14
|
Bathgate RA, Kocan M, Scott DJ, Hossain MA, Good SV, Yegorov S, Bogerd J, Gooley PR. The relaxin receptor as a therapeutic target – perspectives from evolution and drug targeting. Pharmacol Ther 2018; 187:114-132. [DOI: 10.1016/j.pharmthera.2018.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Ocampo Daza D, Larhammar D. Evolution of the receptors for growth hormone, prolactin, erythropoietin and thrombopoietin in relation to the vertebrate tetraploidizations. Gen Comp Endocrinol 2018; 257:143-160. [PMID: 28652136 DOI: 10.1016/j.ygcen.2017.06.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/16/2017] [Accepted: 06/22/2017] [Indexed: 12/19/2022]
Abstract
The receptors for the pituitary hormones growth hormone (GH), prolactin (PRL) and somatolactin (SL), and the hematopoietic hormones erythropoietin (EPO) and thrombopoietin (TPO), comprise a structurally related family in the superfamily of cytokine class-I receptors. GH, PRL and SL receptors have a wide variety of effects in development, osmoregulation, metabolism and stimulation of growth, while EPO and TPO receptors guide the production and differentiation of erythrocytes and thrombocytes, respectively. The evolution of the receptors for GH, PRL and SL has been partially investigated by previous reports suggesting different time points for the hormone and receptor gene duplications. This raises questions about how hormone-receptor partnerships have emerged and evolved. Therefore, we have investigated in detail the expansion of this receptor family, especially in relation to the basal vertebrate (1R, 2R) and teleost (3R) tetraploidizations. Receptor family genes were identified in a broad range of vertebrate genomes and investigated using a combination of sequence-based phylogenetic analyses and comparative genomic analyses of synteny. We found that 1R most likely generated EPOR/TPOR and GHR/PRLR ancestors; following this, 2R resulted in EPOR and TPOR genes. No GHR/PRLR duplicate seems to have survived after 2R. Instead the single GHR/PRLR underwent a local duplication sometime after 2R, generating separate syntenic genes for GHR and PRLR. Subsequently, 3R duplicated the gene pair in teleosts, resulting in two GHR and two PRLR genes, but no EPOR or TPOR duplicates. These analyses help illuminate the evolution of the regulatory mechanisms for somatic growth, metabolism, osmoregulation and hematopoiesis in vertebrates.
Collapse
Affiliation(s)
- Daniel Ocampo Daza
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden.
| | - Dan Larhammar
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden
| |
Collapse
|
16
|
Venditti M, Donizetti A, Fiengo M, Fasano C, Santillo A, Aniello F, Minucci S. Temporal and spatial expression of insulin-like peptide (insl5a and insl5b) paralog genes during the embryogenesis of Danio rerio. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2018; 330:33-40. [PMID: 29319231 DOI: 10.1002/jez.b.22787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/31/2017] [Accepted: 12/10/2017] [Indexed: 12/29/2022]
Abstract
Relaxin (RLN) and insulin (INSL)-like peptides are member of the INSL/RLN superfamily, which are encoded by seven genes in humans and can activate the G-protein coupled receptor RXFP 1-4. These peptides evolved from a common ancestor, RLN3-like gene. Two rounds of whole genome duplication (WGD) in early vertebrate evolution, together with an additional WGD in the teleost lineage, caused an expansion of RLN genes set in the genome of Danio rerio. In particular, six RLN genes are present: a single copy of rln and insl3 genes, and two paralogs for the rln3 gene (rln3a and rln3b), and the insl5 gene (insl5a and insl5b). We have already reported the presence of rln3a and rln3b genes in the developing zebrafish brain, as well as the expression of rln gene in the developing zebrafish brain and extraneural territories, such as thyroid gland and pancreas. Here, we report for the first time the expression of the two parologs genes for insl5, insl5a, and insl5b in D. rerio embryonic development. The corresponding transcripts of both the paralogs are present in all embryonic stages analyzed by RT-qPCR. In situ hybridization analyses showed a restricted signal in intestinal cells and the pancreatic region at 72 hpf for insl5a, while at 96 hpf both genes are expressed in specific intestinal cells. Furthermore, in adult zebrafish intestine tissue, in situ hybridation experiments showed that insl5a transcript is specifically localized in the goblet cells, while insl5b transcript is in enteroendocrine cells. These data revealed a high degree of gene expression pattern conservation for such genes in vertebrate evolution.
Collapse
Affiliation(s)
- Massimo Venditti
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate, Università degli Studi della Campania "Luigi Vanvitelli, Napoli, Italy
| | - Aldo Donizetti
- Dipartimento di Biologia, Università di Napoli "Federico II, Napoli, Italy
| | - Marcella Fiengo
- Dipartimento di Biologia, Università di Napoli "Federico II, Napoli, Italy
| | - Chiara Fasano
- Dipartimento di Biologia, Università di Napoli "Federico II, Napoli, Italy
| | - Alessandra Santillo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania "Luigi Vanvitelli, Caserta, Italy
| | - Francesco Aniello
- Dipartimento di Biologia, Università di Napoli "Federico II, Napoli, Italy
| | - Sergio Minucci
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate, Università degli Studi della Campania "Luigi Vanvitelli, Napoli, Italy
| |
Collapse
|
17
|
Malone L, Opazo JC, Ryan PL, Hoffmann FG. Progressive erosion of the Relaxin1 gene in bovids. Gen Comp Endocrinol 2017; 252:12-17. [PMID: 28733228 DOI: 10.1016/j.ygcen.2017.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/20/2017] [Accepted: 07/12/2017] [Indexed: 02/02/2023]
Abstract
The relaxin/insulin-like (RLN/INSL) gene family is a group of genes that encode peptide hormones involved in a variety of physiological functions related to reproduction. Previous studies have shown that relaxin plays a key role in widening of the pubic bone during labor and in gamete maturation. Because of these functions, studying the evolution of RLN1, the gene encoding for relaxin, is relevant in livestock species, most of which belong in the group Laurasiatheria, which includes cow, pig, horse, goat, and sheep in addition to bats, cetaceans and carnivores. Experimental evidence suggests that cows do not synthesize relaxin, but respond to it, and sheep apparently have a truncated RLN1 gene. Thus, we made use of genome sequence data to characterize the genomic locus of the RLN1 gene in Laurasiatherian mammals to better understand how cows lost the ability to synthesize this peptide. We found that all ruminants in our study (cow, giraffe, goat, sheep and Tibetan antelope) lack a functional RLN1 gene, and document the progressive loss of RLN1 in the lineage leading to cows. Our analyses indicate that 1 - all ruminants have lost all key regulatory elements upstream of the first exon, 2 - giraffe, goat, sheep and Tibetan antelope have multiple inactivating mutations in the RLN1 pseudogene, and 3 - the cow genome has lost all traces of RLN1. The 5' regulatory sequence plays a key role in activating expression, and the loss of this sequence would impair synthesis of mRNA. Our results suggest that changes in regulatory sequence preceded mutations in coding sequence and highlight the importance of these regions in maintaining proper gene function. In addition, we found that all bovids examined posses copies of the relaxin receptors, which explains why they are able to respond to relaxin despite their inability to produce it.
Collapse
Affiliation(s)
- Loggan Malone
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, MS 39762, USA
| | - Juan C Opazo
- Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Peter L Ryan
- Department of Animal and Dairy Sciences, Facility for Organismal and Cellular Imaging (FOCI), Mississippi State University, MS 39762, USA; Department of Pathobiology & Population Medicine, Mississippi State University, MS 39762, USA
| | - Federico G Hoffmann
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, MS 39762, USA; Institute for Genomics, Biocomputing, and Biotechnology, Mississippi State University, MS 39762, USA.
| |
Collapse
|
18
|
Ivell R, Agoulnik AI, Anand‐Ivell R. Relaxin-like peptides in male reproduction - a human perspective. Br J Pharmacol 2017; 174:990-1001. [PMID: 27933606 PMCID: PMC5406299 DOI: 10.1111/bph.13689] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/10/2016] [Accepted: 12/05/2016] [Indexed: 12/26/2022] Open
Abstract
The relaxin family of peptide hormones and their cognate GPCRs are becoming physiologically well-characterized in the cardiovascular system and particularly in female reproductive processes. Much less is known about the physiology and pharmacology of these peptides in male reproduction, particularly as regards humans. H2-relaxin is involved in prostate function and growth, while insulin-like peptide 3 (INSL3) is a major product of the testicular Leydig cells and, in the adult, appears to modulate steroidogenesis and germ cell survival. In the fetus, INSL3 is a key hormone expressed shortly after sex determination and is responsible for the first transabdominal phase of testicular descent. Importantly, INSL3 is becoming a very useful constitutive biomarker reflecting both fetal and post-natal development. Nothing is known about roles for INSL4 in male reproduction and only very little about relaxin-3, which is mostly considered as a brain peptide, or INSL5. The former is expressed at very low levels in the testes, but has no known physiology there, whereas the INSL5 knockout mouse does exhibit a testicular phenotype with mild effects on spermatogenesis, probably due to a disruption of glucose homeostasis. INSL6 is a major product of male germ cells, although it is relatively unexplored with regard to its physiology or pharmacology, except that in mice disruption of the INSL6 gene leads to a disruption of spermatogenesis. Clinically, relaxin analogues may be useful in the control of prostate cancer, and both relaxin and INSL3 have been considered as sperm adjuvants for in vitro fertilization. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
Affiliation(s)
- Richard Ivell
- School of BiosciencesUniversity of NottinghamNottinghamLE12 5RDUK
- School of Veterinary and Medical SciencesUniversity of NottinghamNottinghamLE12 5RDUK
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | | |
Collapse
|
19
|
Furlong M, Seong JY. Evolutionary and Comparative Genomics to Drive Rational Drug Design, with Particular Focus on Neuropeptide Seven-Transmembrane Receptors. Biomol Ther (Seoul) 2017; 25:57-68. [PMID: 28035082 PMCID: PMC5207463 DOI: 10.4062/biomolther.2016.199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/03/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022] Open
Abstract
Seven transmembrane receptors (7TMRs), also known as G protein-coupled receptors, are popular targets of drug development, particularly 7TMR systems that are activated by peptide ligands. Although many pharmaceutical drugs have been discovered via conventional bulk analysis techniques the increasing availability of structural and evolutionary data are facilitating change to rational, targeted drug design. This article discusses the appeal of neuropeptide-7TMR systems as drug targets and provides an overview of concepts in the evolution of vertebrate genomes and gene families. Subsequently, methods that use evolutionary concepts and comparative analysis techniques to aid in gene discovery, gene function identification, and novel drug design are provided along with case study examples.
Collapse
Affiliation(s)
- Michael Furlong
- Graduate School of Biomedical Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Jae Young Seong
- Graduate School of Biomedical Sciences, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
20
|
Semmens DC, Mirabeau O, Moghul I, Pancholi MR, Wurm Y, Elphick MR. Transcriptomic identification of starfish neuropeptide precursors yields new insights into neuropeptide evolution. Open Biol 2016; 6:150224. [PMID: 26865025 PMCID: PMC4772807 DOI: 10.1098/rsob.150224] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neuropeptides are evolutionarily ancient mediators of neuronal signalling in nervous systems. With recent advances in genomics/transcriptomics, an increasingly wide range of species has become accessible for molecular analysis. The deuterostomian invertebrates are of particular interest in this regard because they occupy an ‘intermediate' position in animal phylogeny, bridging the gap between the well-studied model protostomian invertebrates (e.g. Drosophila melanogaster, Caenorhabditis elegans) and the vertebrates. Here we have identified 40 neuropeptide precursors in the starfish Asterias rubens, a deuterostomian invertebrate from the phylum Echinodermata. Importantly, these include kisspeptin-type and melanin-concentrating hormone-type precursors, which are the first to be discovered in a non-chordate species. Starfish tachykinin-type, somatostatin-type, pigment-dispersing factor-type and corticotropin-releasing hormone-type precursors are the first to be discovered in the echinoderm/ambulacrarian clade of the animal kingdom. Other precursors identified include vasopressin/oxytocin-type, gonadotropin-releasing hormone-type, thyrotropin-releasing hormone-type, calcitonin-type, cholecystokinin/gastrin-type, orexin-type, luqin-type, pedal peptide/orcokinin-type, glycoprotein hormone-type, bursicon-type, relaxin-type and insulin-like growth factor-type precursors. This is the most comprehensive identification of neuropeptide precursor proteins in an echinoderm to date, yielding new insights into the evolution of neuropeptide signalling systems. Furthermore, these data provide a basis for experimental analysis of neuropeptide function in the unique context of the decentralized, pentaradial echinoderm bauplan.
Collapse
Affiliation(s)
- Dean C Semmens
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Olivier Mirabeau
- Institut Curie, Genetics and Biology of Cancers Unit, INSERM U830, PSL Research University, Paris 75005, France
| | - Ismail Moghul
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Mahesh R Pancholi
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Yannick Wurm
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Maurice R Elphick
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
21
|
Hu CK, Southey BR, Romanova EV, Maruska KP, Sweedler JV, Fernald RD. Identification of prohormones and pituitary neuropeptides in the African cichlid, Astatotilapia burtoni. BMC Genomics 2016; 17:660. [PMID: 27543050 PMCID: PMC4992253 DOI: 10.1186/s12864-016-2914-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/06/2016] [Indexed: 12/14/2022] Open
Abstract
Background Cichlid fishes have evolved remarkably diverse reproductive, social, and feeding behaviors. Cell-to-cell signaling molecules, notably neuropeptides and peptide hormones, are known to regulate these behaviors across vertebrates. This class of signaling molecules derives from prohormone genes that have undergone multiple duplications and losses in fishes. Whether and how subfunctionalization, neofunctionalization, or losses of neuropeptides and peptide hormones have contributed to fish behavioral diversity is largely unknown. Information on fish prohormones has been limited and is complicated by the whole genome duplication of the teleost ancestor. We combined bioinformatics, mass spectrometry-enabled peptidomics, and molecular techniques to identify the suite of neuropeptide prohormones and pituitary peptide products in Astatotilapia burtoni, a well-studied member of the diverse African cichlid clade. Results Utilizing the A. burtoni genome, we identified 148 prohormone genes, with 21 identified as a single copy and 39 with at least 2 duplicated copies. Retention of prohormone duplicates was therefore 41 %, which is markedly above previous reports for the genome-wide average in teleosts. Beyond the expected whole genome duplication, differences between cichlids and mammals can be attributed to gene loss in tetrapods and additional duplication after divergence. Mass spectrometric analysis of the pituitary identified 620 unique peptide sequences that were matched to 120 unique proteins. Finally, we used in situ hybridization to localize the expression of galanin, a prohormone with exceptional sequence divergence in cichlids, as well as the expression of a proopiomelanocortin, prohormone that has undergone an additional duplication in some bony fish lineages. Conclusion We characterized the A. burtoni prohormone complement. Two thirds of prohormone families contain duplications either from the teleost whole genome duplication or a more recent duplication. Our bioinformatic and mass spectrometric findings provide information on a major vertebrate clade that will further our understanding of the functional ramifications of these prohormone losses, duplications, and sequence changes across vertebrate evolution. In the context of the cichlid radiation, these findings will also facilitate the exploration of neuropeptide and peptide hormone function in behavioral diversity both within A. burtoni and across cichlid and other fish species. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2914-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caroline K Hu
- Department of Biology, Stanford University, Stanford, CA, 94305, USA.,Present address: Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Karen P Maruska
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Russell D Fernald
- Department of Biology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
22
|
Phylogenetic-Derived Insights into the Evolution of Sialylation in Eukaryotes: Comprehensive Analysis of Vertebrate β-Galactoside α2,3/6-Sialyltransferases (ST3Gal and ST6Gal). Int J Mol Sci 2016; 17:ijms17081286. [PMID: 27517905 PMCID: PMC5000683 DOI: 10.3390/ijms17081286] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 12/21/2022] Open
Abstract
Cell surface of eukaryotic cells is covered with a wide variety of sialylated molecules involved in diverse biological processes and taking part in cell–cell interactions. Although the physiological relevance of these sialylated glycoconjugates in vertebrates begins to be deciphered, the origin and evolution of the genetic machinery implicated in their biosynthetic pathway are poorly understood. Among the variety of actors involved in the sialylation machinery, sialyltransferases are key enzymes for the biosynthesis of sialylated molecules. This review focus on β-galactoside α2,3/6-sialyltransferases belonging to the ST3Gal and ST6Gal families. We propose here an outline of the evolutionary history of these two major ST families. Comparative genomics, molecular phylogeny and structural bioinformatics provided insights into the functional innovations in sialic acid metabolism and enabled to explore how ST-gene function evolved in vertebrates.
Collapse
|
23
|
Hojo K, Hossain MA, Tailhades J, Shabanpoor F, Wong LLL, Ong-Pålsson EEK, Kastman HE, Ma S, Gundlach AL, Rosengren KJ, Wade JD, Bathgate RAD. Development of a Single-Chain Peptide Agonist of the Relaxin-3 Receptor Using Hydrocarbon Stapling. J Med Chem 2016; 59:7445-56. [PMID: 27464307 DOI: 10.1021/acs.jmedchem.6b00265] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Structure-activity studies of the insulin superfamily member, relaxin-3, have shown that its G protein-coupled receptor (RXFP3) binding site is contained within its central B-chain α-helix and this helical structure is essential for receptor activation. We sought to develop a single B-chain mimetic that retained agonist activity. This was achieved by use of solid phase peptide synthesis together with on-resin ruthenium-catalyzed ring closure metathesis of a pair of judiciously placed i,i+4 α-methyl, α-alkenyl amino acids. The resulting hydrocarbon stapled peptide was shown by solution NMR spectroscopy to mimic the native helical conformation of relaxin-3 and to possess potent RXFP3 receptor binding and activation. Alternative stapling procedures were unsuccessful, highlighting the critical need to carefully consider both the peptide sequence and stapling methodology for optimal outcomes. Our result is the first successful minimization of an insulin-like peptide to a single-chain α-helical peptide agonist which will facilitate study of the function of relaxin-3.
Collapse
Affiliation(s)
- Keiko Hojo
- Faculty of Pharmaceutical Sciences and Cooperative Research Center of Life Sciences, Kobe Gakuin University , Chuo-ku, Kobe 650-8586, Japan
| | - Mohammed Akhter Hossain
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia.,School of Chemistry, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Julien Tailhades
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Fazel Shabanpoor
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia.,School of Chemistry, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Lilian L L Wong
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Emma E K Ong-Pålsson
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Hanna E Kastman
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Sherie Ma
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Andrew L Gundlach
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia.,Department of Anatomy and Neuroscience, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - K Johan Rosengren
- School of Biomedical Sciences, The University of Queensland , Brisbane, Queensland 4072, Australia
| | - John D Wade
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia.,School of Chemistry, University of Melbourne , Melbourne, Victoria 3052, Australia
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne , Melbourne, Victoria 3052, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne , Melbourne, Victoria 3052, Australia
| |
Collapse
|
24
|
Mita M, Katayama H. A relaxin-like gonad-stimulating peptide from the starfish Aphelasterias japonica. Gen Comp Endocrinol 2016; 229:56-61. [PMID: 26944483 DOI: 10.1016/j.ygcen.2016.02.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/19/2016] [Accepted: 02/29/2016] [Indexed: 11/21/2022]
Abstract
Relaxin-like gonad-stimulating peptide (RGP) in starfish is the first identified invertebrate gonadotropin responsible for final gamete maturation. In this study, a new ortholog RGP was identified from Aphelasterias japonica. The DNA sequence encoding A. japonica RGP (AjaRGP) consists of 342 base pairs with an open reading frame encoding a peptide of 113 amino acids (aa), including a signal peptide (26aa), B-chain (20aa), C-peptide (42aa), and A-chain (25aa). AjaRGP is a heterodimeric peptide with disulfide cross-linkages. Comparing with Asterias amurensis RGP (AamRGP) and Patiria (=Asterina) pectinifera RGP (PpeRGP), the amino acid identity levels of AjaRGP with respect to AamRGP and PpeRGP are 84% and 58% for the A-chain and 90% and 68% for the B-chain, respectively. This suggests that AjaRGP is closer to AmaRGP rather than PpeRGP. Although chemical synthetic AjaRGP can induce gamete spawning and oocyte maturation in ovarian fragments of A. japonica, the ovary of P. pectinifera fails to respond to AjaRGP. This suggests that AjaRGP acts species-specifically.
Collapse
Affiliation(s)
- Masatoshi Mita
- Department of Biology, Faculty of Education, Tokyo Gakugei University, Nukuikita-machi 4-1-1, Koganei-shi, Tokyo 184-8501, Japan.
| | - Hidekazu Katayama
- Department of Applied Biochemistry, School of Engineering, Tokai University, 4-1-1, Kitakaname, Hiratuska, Kanagawa 259-1292, Japan
| |
Collapse
|
25
|
Garelli A, Heredia F, Casimiro AP, Macedo A, Nunes C, Garcez M, Dias ARM, Volonte YA, Uhlmann T, Caparros E, Koyama T, Gontijo AM. Dilp8 requires the neuronal relaxin receptor Lgr3 to couple growth to developmental timing. Nat Commun 2015; 6:8732. [PMID: 26510564 PMCID: PMC4640092 DOI: 10.1038/ncomms9732] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/25/2015] [Indexed: 11/18/2022] Open
Abstract
How different organs in the body sense growth perturbations in distant tissues to coordinate their size during development is poorly understood. Here we mutate an invertebrate orphan relaxin receptor gene, the Drosophila Leucine-rich repeat-containing G protein-coupled receptor 3 (Lgr3), and find body asymmetries similar to those found in insulin-like peptide 8 (dilp8) mutants, which fail to coordinate growth with developmental timing. Indeed, mutation or RNA intereference (RNAi) against Lgr3 suppresses the delay in pupariation induced by imaginal disc growth perturbation or ectopic Dilp8 expression. By tagging endogenous Lgr3 and performing cell type-specific RNAi, we map this Lgr3 activity to a new subset of CNS neurons, four of which are a pair of bilateral pars intercerebralis Lgr3-positive (PIL) neurons that respond specifically to ectopic Dilp8 by increasing cAMP-dependent signalling. Our work sheds new light on the function and evolution of relaxin receptors and reveals a novel neuroendocrine circuit responsive to growth aberrations. The orphan ligand Dilp8 has been shown to coordinate growth and developmental timing in Drosophila. Here, using Gal4 drivers and CRISPR/Cas9 approaches, Garelli et al. identify a role for relaxin-like receptor Lgr3 in regulating the Dilp8 developmental delay pathway.
Collapse
Affiliation(s)
- Andres Garelli
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal.,Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET and Universidad Nacional del Sur, Camino La Carrindanga km7, Bahía Blanca B8000 FWB, Argentina
| | - Fabiana Heredia
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Andreia P Casimiro
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Andre Macedo
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Catarina Nunes
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Marcia Garcez
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Angela R Mantas Dias
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Yanel A Volonte
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET and Universidad Nacional del Sur, Camino La Carrindanga km7, Bahía Blanca B8000 FWB, Argentina
| | - Thomas Uhlmann
- Dualsystems Biotech AG, Grabenstrasse 11a, Schlieren CH-8952, Switzerland
| | - Esther Caparros
- Departamento de Medicina Clínica, Facultad de Medicina, Universidad Miguel Hernández, Ctra. Alicante-Valencia, km 87, San Juan, Alicante 03550, Spain
| | - Takashi Koyama
- Development, Evolution and the Environment Laboratory, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Alisson M Gontijo
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| |
Collapse
|
26
|
Mita M, Daiya M, Haraguchi S, Tsutsui K, Nagahama Y. A new relaxin-like gonad-stimulating peptide identified in the starfish Asterias amurensis. Gen Comp Endocrinol 2015; 222:144-9. [PMID: 26163025 DOI: 10.1016/j.ygcen.2015.07.002] [Citation(s) in RCA: 20] [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: 04/08/2015] [Revised: 06/12/2015] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
Abstract
Relaxin-like gonad-stimulating peptide (RGP) of starfish Asterina pectinifera was the first invertebrate gonadotropin to have its chemical structure identified. However, it is unclear whether gonadotropic hormones in other species starfish are relaxin-like peptides. Thus, this study tried to identify the molecular structure of gonadotropic hormone in Asterias amurensis. As a result, we identified A. amurensis gonadotropic hormone as the RGP (AamRGP). The DNA sequence encoding AamRGP consisted of 330 base pairs with an open reading frame encoding a peptide of 109 amino acids (aa), including a signal peptide (26 aa), B-chain (20 aa), C-peptide (38 aa) and A-chain (25 aa). Comparing with A. pectinifera RGP (ApeRGP), the amino acid identity levels between AmaRGP and ApeRGP were 58% for the A-chain and 73% for the B-chain. Furthermore, chemical synthetic AamRGP induced gamete spawning and oocyte maturation in ovarian fragments of A. amurensis. In contrast, the ovary of A. pectinifera failed to respond to the AamRGP. This suggested that AamRGP is a new relaxin-like peptide.
Collapse
Affiliation(s)
- Masatoshi Mita
- Department of Biology, Faculty of Education, Tokyo Gakugei University, Nukuikita-machi 4-1-1, Koganei-shi, Tokyo 184-8501, Japan.
| | - Misaki Daiya
- Department of Biology, Faculty of Education, Tokyo Gakugei University, Nukuikita-machi 4-1-1, Koganei-shi, Tokyo 184-8501, Japan
| | - Shogo Haraguchi
- Department of Biology, Faculty of Education, Tokyo Gakugei University, Nukuikita-machi 4-1-1, Koganei-shi, Tokyo 184-8501, Japan; Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Wakamatsucho 2-2, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Wakamatsucho 2-2, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Yoshitaka Nagahama
- Institute for Collaborative Relations, Ehime University, Matsuyama, Ehime 790-8577, Japan
| |
Collapse
|
27
|
Yun S, Furlong M, Sim M, Cho M, Park S, Cho EB, Reyes-Alcaraz A, Hwang JI, Kim J, Seong JY. Prevertebrate Local Gene Duplication Facilitated Expansion of the Neuropeptide GPCR Superfamily. Mol Biol Evol 2015; 32:2803-17. [DOI: 10.1093/molbev/msv179] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
28
|
Donizetti A, Fiengo M, Del Gaudio R, Iazzetti G, Pariante P, Minucci S, Aniello F. Expression pattern of zebrafish rxfp2 homologue genes during embryonic development. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2015; 324:605-13. [PMID: 26173401 DOI: 10.1002/jez.b.22637] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 06/05/2015] [Indexed: 12/19/2022]
Abstract
RXFP2 is one of the 4 receptors for relaxin insulin-like peptides, in particular it binds with high affinity the INSL3 peptide. INSL3/RXFP2 pair is essential for testicular descent during placental mammalian development. The evolutionary history of this ligand/receptor pair has received much attention, since its function in vertebrate species lacking testicular descent, such as the fishes, remains elusive. Herein, we analyzed the expression pattern of three rxfp2 homologue genes in zebrafish embryonic development. For all the three rxfp2 genes (rxfp2a, rxfp2b, and rxfp2-like) we showed the presence of maternally derived transcripts. Later in the development, rxfp2a is only expressed at larval stage, whereas rxfp2b is expressed in all the analyzed stage with highest level in the larvae. The rxfp2-like gene is expressed in all the analyzed stage with a transcript level that increased starting at early pharyngula stage. The spatial localization analysis of rxfp2-like gene showed that it is expressed in many cell clusters in the developing brain. In addition, other rxfp2-like-expressing cells were identified in the retina and oral epithelium. This analysis provides new insights to elucidate the evolution of rxfp2 genes in vertebrate lineage and lays the foundations to study their role in vertebrate embryonic development.
Collapse
Affiliation(s)
- Aldo Donizetti
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marcella Fiengo
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | - Giovanni Iazzetti
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paolo Pariante
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Sergio Minucci
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Francesco Aniello
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
29
|
Spielman SJ, Kumar K, Wilke CO. Comprehensive, structurally-informed alignment and phylogeny of vertebrate biogenic amine receptors. PeerJ 2015; 3:e773. [PMID: 25737813 PMCID: PMC4338800 DOI: 10.7717/peerj.773] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/26/2015] [Indexed: 01/29/2023] Open
Abstract
Biogenic amine receptors play critical roles in regulating behavior and physiology in both vertebrates and invertebrates, particularly within the central nervous system. Members of the G-protein coupled receptor (GPCR) family, these receptors interact with endogenous bioamine ligands such as dopamine, serotonin, and epinephrine, and are targeted by a wide array of pharmaceuticals. Despite the clear clinical and biological importance of these receptors, their evolutionary history remains poorly characterized. In particular, the relationships among biogenic amine receptors and any specific evolutionary constraints acting within distinct receptor subtypes are largely unknown. To advance and facilitate studies in this receptor family, we have constructed a comprehensive, high-quality sequence alignment of vertebrate biogenic amine receptors. In particular, we have integrated a traditional multiple sequence approach with robust structural domain predictions to ensure that alignment columns accurately capture the highly-conserved GPCR structural domains, and we demonstrate how ignoring structural information produces spurious inferences of homology. Using this alignment, we have constructed a structurally-partitioned maximum-likelihood phylogeny from which we deduce novel biogenic amine receptor relationships and uncover previously unrecognized lineage-specific receptor clades. Moreover, we find that roughly 1% of the 3039 sequences in our final alignment are either misannotated or unclassified, and we propose updated classifications for these receptors. We release our comprehensive alignment and its corresponding phylogeny as a resource for future research into the evolution and diversification of biogenic amine receptors.
Collapse
Affiliation(s)
| | - Keerthana Kumar
- Department of Integrative Biology, The University of Texas at Austin, Austin, USA
| | - Claus O. Wilke
- Department of Integrative Biology, The University of Texas at Austin, Austin, USA
| |
Collapse
|
30
|
Petit D, Teppa E, Mir AM, Vicogne D, Thisse C, Thisse B, Filloux C, Harduin-Lepers A. Integrative view of α2,3-sialyltransferases (ST3Gal) molecular and functional evolution in deuterostomes: significance of lineage-specific losses. Mol Biol Evol 2014; 32:906-27. [PMID: 25534026 PMCID: PMC4379398 DOI: 10.1093/molbev/msu395] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Sialyltransferases are responsible for the synthesis of a diverse range of sialoglycoconjugates predicted to be pivotal to deuterostomes’ evolution. In this work, we reconstructed the evolutionary history of the metazoan α2,3-sialyltransferases family (ST3Gal), a subset of sialyltransferases encompassing six subfamilies (ST3Gal I–ST3Gal VI) functionally characterized in mammals. Exploration of genomic and expressed sequence tag databases and search of conserved sialylmotifs led to the identification of a large data set of st3gal-related gene sequences. Molecular phylogeny and large scale sequence similarity network analysis identified four new vertebrate subfamilies called ST3Gal III-r, ST3Gal VII, ST3Gal VIII, and ST3Gal IX. To address the issue of the origin and evolutionary relationships of the st3gal-related genes, we performed comparative syntenic mapping of st3gal gene loci combined to ancestral genome reconstruction. The ten vertebrate ST3Gal subfamilies originated from genome duplication events at the base of vertebrates and are organized in three distinct and ancient groups of genes predating the early deuterostomes. Inferring st3gal gene family history identified also several lineage-specific gene losses, the significance of which was explored in a functional context. Toward this aim, spatiotemporal distribution of st3gal genes was analyzed in zebrafish and bovine tissues. In addition, molecular evolutionary analyses using specificity determining position and coevolved amino acid predictions led to the identification of amino acid residues with potential implication in functional divergence of vertebrate ST3Gal. We propose a detailed scenario of the evolutionary relationships of st3gal genes coupled to a conceptual framework of the evolution of ST3Gal functions.
Collapse
Affiliation(s)
- Daniel Petit
- INRA, UMR 1061, Unité Génétique Moléculaire Animale, F-87060 Limoges Cedex, France Université de Limoges, UMR 1061, Unité Génétique Moléculaire Animale, 123 avenue Albert Thomas, F-87060 Limoges Cedex, France
| | - Elin Teppa
- Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Anne-Marie Mir
- Laboratoire de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS, Université Lille Nord de France, Lille1, Villeneuve d'Ascq, France
| | - Dorothée Vicogne
- Laboratoire de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS, Université Lille Nord de France, Lille1, Villeneuve d'Ascq, France
| | - Christine Thisse
- Department of Cell Biology, School of Medicine, University of Virginia
| | - Bernard Thisse
- Department of Cell Biology, School of Medicine, University of Virginia
| | - Cyril Filloux
- INRA, UMR 1061, Unité Génétique Moléculaire Animale, F-87060 Limoges Cedex, France Université de Limoges, UMR 1061, Unité Génétique Moléculaire Animale, 123 avenue Albert Thomas, F-87060 Limoges Cedex, France
| | - Anne Harduin-Lepers
- Laboratoire de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS, Université Lille Nord de France, Lille1, Villeneuve d'Ascq, France
| |
Collapse
|
31
|
Yegorov S, Bogerd J, Good SV. The relaxin family peptide receptors and their ligands: new developments and paradigms in the evolution from jawless fish to mammals. Gen Comp Endocrinol 2014; 209:93-105. [PMID: 25079565 DOI: 10.1016/j.ygcen.2014.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 07/01/2014] [Accepted: 07/16/2014] [Indexed: 12/13/2022]
Abstract
Relaxin family peptide receptors (Rxfps) and their ligands, relaxin (Rln) and insulin-like (Insl) peptides, are broadly implicated in the regulation of reproductive and neuroendocrine processes in mammals. Most placental mammals harbour genes for four receptors, namely rxfp1, rxfp2, rxfp3 and rxfp4. The number and identity of rxfps in other vertebrates are immensely variable, which is probably attributable to intraspecific variation in reproductive and neuroendocrine regulation. Here, we highlight several interesting, but greatly overlooked, aspects of the rln/insl-rxfp evolutionary history: the ancient origin, recruitment of novel receptors, diverse roles of selection, differential retention and lineage-specific loss of genes over evolutionary time. The tremendous diversity of rln/insl and rxfp genes appears to have arisen from two divergent receptors and one ligand that were duplicated by whole genome duplications (WGD) in early vertebrate evolution, although several genes, notably relaxin in mammals, were also duplicated via small scale duplications. Duplication and loss of genes have varied across lineages: teleosts retained more WGD-derived genes, dominated by those thought to be involved in neuroendocrine regulation (rln3, insl5 and rxfp 3/4 genes), while eutherian mammals witnessed the diversification and rapid evolution of genes involved in reproduction (rln/insl3). Several genes that arose early in evolutionary history were lost in most mammals, but retained in teleosts and, to a lesser extent, in early diverging tetrapods. To elaborate on their evolutionary history, we provide updated phylogenies of the Rxfp1/2 and Rxfp3/4 receptors and their ligands, including new sequences from early diverging vertebrate taxa such as coelacanth, skate, spotted gar, and lamprey. We also summarize the recent progress made towards understanding the functional biology of Rxfps in non-mammalian taxa, providing a new conceptual framework for research on Rxfp signaling across vertebrates.
Collapse
Affiliation(s)
- Sergey Yegorov
- Department of Biology, University of Winnipeg, 599 Portage Ave., Winnipeg, MB, Canada
| | - Jan Bogerd
- Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Sara V Good
- Department of Biology, University of Winnipeg, 599 Portage Ave., Winnipeg, MB, Canada.
| |
Collapse
|
32
|
Donizetti A, Fiengo M, Iazzetti G, del Gaudio R, Di Giaimo R, Pariante P, Minucci S, Aniello F. Expression analysis of five zebrafish RXFP3 homologues reveals evolutionary conservation of gene expression pattern. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2014; 324:22-9. [PMID: 25384467 DOI: 10.1002/jez.b.22591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/19/2014] [Indexed: 12/19/2022]
Abstract
Relaxin peptides exert different functions in reproduction and neuroendocrine processes via interaction with two evolutionarily unrelated groups of receptors: RXFP1 and RXFP2 on one hand, RXFP3 and RXFP4 on the other hand. Evolution of receptor genes after splitting of tetrapods and teleost lineage led to a different retention rate between mammals and fish, with the latter having more gene copies compared to the former. In order to improve our knowledge on the evolution of the relaxin ligands/receptors system and have insights on their function in early stages of life, in the present paper we analyzed the expression pattern of five zebrafish RXFP3 homologue genes during embryonic development. In our analysis, we show that only two of the five genes are expressed during embryogenesis and that their transcripts are present in all the developmental stages. Spatial localization analysis of these transcripts revealed that the gene expression is restricted in specific territories starting from early pharyngula stage. Both genes are expressed in the brain but in different cell clusters and in extra-neural territories, one gene in the interrenal gland and the other in the pancreas. These two genes share expression territories with the homologue mammalian counterpart, highlighting a general conservation of gene expression regulatory processes and their putative function during evolution that are established early in vertebrate embryogenesis.
Collapse
Affiliation(s)
- Aldo Donizetti
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Arroyo JI, Hoffmann FG, Opazo JC. Evolution of the relaxin/insulin-like gene family in anthropoid primates. Genome Biol Evol 2014; 6:491-9. [PMID: 24493383 PMCID: PMC3971578 DOI: 10.1093/gbe/evu023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The relaxin/insulin-like gene family includes signaling molecules that perform a variety of physiological roles mostly related to reproduction and neuroendocrine regulation. Several previous studies have focused on the evolutionary history of relaxin genes in anthropoid primates, with particular attention on resolving the duplication history of RLN1 and RLN2 genes, which are found as duplicates only in apes. These studies have revealed that the RLN1 and RLN2 paralogs in apes have a more complex history than their phyletic distribution would suggest. In this regard, alternative scenarios have been proposed to explain the timing of duplication, and the history of gene gain and loss along the organismal tree. In this article, we revisit the question and specifically reconstruct phylogenies based on coding and noncoding sequence in anthropoid primates to readdress the timing of the duplication event giving rise to RLN1 and RLN2 in apes. Results from our phylogenetic analyses based on noncoding sequence revealed that the duplication event that gave rise to the RLN1 and RLN2 occurred in the last common ancestor of catarrhine primates, between ∼44.2 and 29.6 Ma, and not in the last common ancestor of apes or anthropoids, as previously suggested. Comparative analyses based on coding and noncoding sequence suggests an event of convergent evolution at the sequence level between co-ortholog genes, the single-copy RLN gene found in New World monkeys and the RLN1 gene of apes, where changes in a fraction of the convergent sites appear to be driven by positive selection.
Collapse
Affiliation(s)
- José Ignacio Arroyo
- Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | | | | |
Collapse
|
34
|
Abstract
Glucagon-like peptide 1 (GLP1) is an intestinal incretin that regulates glucose homeostasis through stimulation of insulin secretion from pancreatic β-cells and inhibits appetite by acting on the brain. Thus, it is a promising therapeutic agent for the treatment of type 2 diabetes mellitus and obesity. Studies using synteny and reconstructed ancestral chromosomes suggest that families for GLP1 and its receptor (GLP1R) have emerged through two rounds (2R) of whole genome duplication and local gene duplications before and after 2R. Exon duplications have also contributed to the expansion of the peptide family members. Specific changes in the amino acid sequence following exon/gene/genome duplications have established distinct yet related peptide and receptor families. These specific changes also confer selective interactions between GLP1 and GLP1R. In this review, we present a possible macro (genome level)- and micro (gene/exon level)-evolution mechanisms of GLP1 and GLP1R, which allows them to acquire selective interactions between this ligand-receptor pair. This information may provide critical insight for the development of potent therapeutic agents targeting GLP1R.
Collapse
Affiliation(s)
- Jong-Ik Hwang
- Graduate School of MedicineKorea University, Seoul 136-705, Republic of Korea
| | - Seongsik Yun
- Graduate School of MedicineKorea University, Seoul 136-705, Republic of Korea
| | - Mi Jin Moon
- Graduate School of MedicineKorea University, Seoul 136-705, Republic of Korea
| | - Cho Rong Park
- Graduate School of MedicineKorea University, Seoul 136-705, Republic of Korea
| | - Jae Young Seong
- Graduate School of MedicineKorea University, Seoul 136-705, Republic of Korea
| |
Collapse
|
35
|
Wendland J, Walther A. Chromosome number reduction in Eremothecium coryli by two telomere-to-telomere fusions. Genome Biol Evol 2014; 6:1186-98. [PMID: 24803574 PMCID: PMC4040997 DOI: 10.1093/gbe/evu089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2014] [Indexed: 11/16/2022] Open
Abstract
The genus Eremothecium belongs to the Saccharomyces complex of pre-whole-genome duplication (WGD) yeasts and contains both dimorphic and filamentous species. We established the 9.1-Mb draft genome of Eremothecium coryli, which encodes 4,682 genes, 186 tRNA genes, and harbors several Ty3 transposons as well as more than 60 remnants of transposition events (LTRs). The initial de novo assembly resulted in 19 scaffolds, which were assembled based on synteny to other Eremothecium genomes into six chromosomes. Interestingly, we identified eight E. coryli loci that bear centromeres in the closely related species E. cymbalariae. Two of these E. coryli loci, CEN1 and CEN8, however, lack conserved DNA elements and did not convey centromere function in a plasmid stability assay. Correspondingly, using a comparative genomics approach we identified two telomere-to-telomere fusion events in E. coryli as the cause of chromosome number reduction from eight to six chromosomes. Finally, with the genome sequences of E. coryli, E. cymbalariae, and Ashbya gossypii a reconstruction of three complete chromosomes of an Eremothecium ancestor revealed that E. coryli is more syntenic to this ancestor than the other Eremothecium species.
Collapse
Affiliation(s)
| | - Andrea Walther
- Carlsberg Laboratory, Yeast Biology, Copenhagen, Denmark
| |
Collapse
|
36
|
Kim DK, Yun S, Son GH, Hwang JI, Park CR, Kim JI, Kim K, Vaudry H, Seong JY. Coevolution of the spexin/galanin/kisspeptin family: Spexin activates galanin receptor type II and III. Endocrinology 2014; 155:1864-73. [PMID: 24517231 DOI: 10.1210/en.2013-2106] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The novel neuropeptide spexin (SPX) was discovered using bioinformatics. The function of this peptide is currently under investigation. Here, we identified SPX along with a second SPX gene (SPX2) in vertebrate genomes. Syntenic analysis and relocating SPXs and their neighbor genes on reconstructed vertebrate ancestral chromosomes revealed that SPXs reside in the near vicinity of the kisspeptin (KISS) and galanin (GAL) family genes on the chromosomes. Alignment of mature peptide sequences showed some extent of sequence similarity among the 3 peptide groups. Gene structure analysis indicated that SPX is more closely related to GAL than KISS. These results suggest that the SPX, GAL, and KISS genes arose through local duplications before 2 rounds (2R) of whole-genome duplication. Receptors of KISS and GAL (GAL receptor [GALR]) are phylogenetically closest among rhodopsin-like G protein-coupled receptors, and synteny revealed the presence of 3 distinct receptor families KISS receptor, GALR1, and GALR2/3 before 2R. A ligand-receptor interaction study showed that SPXs activate human, Xenopus, and zebrafish GALR2/3 family receptors but not GALR1, suggesting that SPXs are natural ligands for GALR2/3. Particularly, SPXs exhibited much higher potency toward GALR3 than GAL. Together, these results identify the coevolution of SPX/GAL/KISS ligand genes with their receptor genes. This study demonstrates the advantage of evolutionary genomics to explore the evolutionary relationship of a peptide gene family that arose before 2R by local duplications.
Collapse
MESH Headings
- Animals
- Chromosome Mapping
- Databases, Nucleic Acid
- Databases, Protein
- Evolution, Molecular
- Galanin/chemistry
- Galanin/genetics
- Galanin/metabolism
- Gene Duplication
- HEK293 Cells
- Humans
- Kisspeptins/chemistry
- Kisspeptins/genetics
- Kisspeptins/metabolism
- Ligands
- Neuropeptides/chemistry
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Peptide Hormones/chemistry
- Peptide Hormones/genetics
- Peptide Hormones/metabolism
- Phylogeny
- Protein Isoforms/chemistry
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Receptor, Galanin, Type 1/agonists
- Receptor, Galanin, Type 1/chemistry
- Receptor, Galanin, Type 1/genetics
- Receptor, Galanin, Type 1/metabolism
- Receptor, Galanin, Type 2/agonists
- Receptor, Galanin, Type 2/chemistry
- Receptor, Galanin, Type 2/genetics
- Receptor, Galanin, Type 2/metabolism
- Receptor, Galanin, Type 3/agonists
- Receptor, Galanin, Type 3/chemistry
- Receptor, Galanin, Type 3/genetics
- Receptor, Galanin, Type 3/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Kisspeptin-1
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Sequence Alignment
- Synteny
- Zebrafish Proteins/chemistry
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Dong-Kyu Kim
- Graduate School of Medicine (D.-K.K., S.Y., G.H.S., J.-I.H., C.R.P., J.Y.S.), Korea University, Seoul 136-705, Republic of Korea; Department of Life Science (J.I.K.), Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea; School of Biological Sciences (K.K.), Seoul National University, Seoul 151-742, Republic of Korea; and Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (H.V.), Inserm U982, University of Rouen, 76821 Mont-St-Aignan, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sefideh FA, Moon MJ, Yun S, Hong SI, Hwang JI, Seong JY. Local duplication of gonadotropin-releasing hormone (GnRH) receptor before two rounds of whole genome duplication and origin of the mammalian GnRH receptor. PLoS One 2014; 9:e87901. [PMID: 24498396 PMCID: PMC3912137 DOI: 10.1371/journal.pone.0087901] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/30/2013] [Indexed: 12/02/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) and the GnRH receptor (GnRHR) play an important role in vertebrate reproduction. Although many GnRHR genes have been identified in a large variety of vertebrate species, the evolutionary history of GnRHR in vertebrates is unclear. To trace the evolutionary origin of GnRHR we examined the conserved synteny of chromosomes harboring GnRHR genes and matched the genes to linkage groups of reconstructed vertebrate ancestor chromosomes. Consistent with the phylogenetic tree, three pairs of GnRHR subtypes were identified in three paralogous linkage groups, indicating that an ancestral pair emerged through local duplication before two rounds of whole genome duplication (2R). The 2R then led to the generation of six subtypes of GnRHR. Some subtypes were lost during vertebrate evolution after the divergence of teleosts and tetrapods. One subtype includes mammalian GnRHR and a coelacanth GnRHR that showed the greatest response to GnRH1 among the three types of GnRH. This study provides new insight into the evolutionary relationship of vertebrate GnRHRs.
Collapse
Affiliation(s)
| | - Mi Jin Moon
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | - Seongsik Yun
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | - Sung In Hong
- Department of East-West Integrated Medicine, College of Oriental Medicine, Gachon University, Incheon, Republic of Korea
| | - Jong-Ik Hwang
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
38
|
Yun S, Kim DK, Furlong M, Hwang JI, Vaudry H, Seong JY. Does Kisspeptin Belong to the Proposed RF-Amide Peptide Family? Front Endocrinol (Lausanne) 2014; 5:134. [PMID: 25165463 PMCID: PMC4131245 DOI: 10.3389/fendo.2014.00134] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/30/2014] [Indexed: 11/13/2022] Open
Abstract
Kisspeptin (KISS) plays a key role in regulating reproduction by binding to its receptor, GPR54. Because of the Arg-Phe (RF) sequence at its carboxyl terminus, KISS has been proposed to be a member of the RF-amide peptide family consisting of neuropeptide FF (NPFF), neuropeptide VF (NPVF), pyroglutamylated RF-amide peptide (QRFP), and prolactin-releasing hormone (PRLH). Evolutionary relationships of protein families can be determined through phylogenetic analysis. However, phylogenetic analysis among related peptide families often fails to provide sufficient information because only short mature peptide sequences from full preprohormone sequences are conserved. Considering the concept of the coevolution of peptide ligands and their cognate receptors, evolutionary relationships among related receptor families provide clues to explore relationships between their peptides. Although receptors for NPFF, NPVF, and QRFP are phylogenetically clustered together, receptors for PRLH and KISS are on different branches of the phylogenetic tree. In particular, KISS has been proposed to be a member of the KISS/galanin/spexin family based on synteny analysis and the phylogenetic relationship between their receptors. This article discusses the evolutionary history of the receptors for the proposed RF-amide peptide family and proposes that, from an evolutionary aspect, KISS has emerged from an ancestor, which is distinct from those of the other RF-amide peptides, and so should be classed separately.
Collapse
Affiliation(s)
- Seongsik Yun
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Dong-Kyu Kim
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Michael Furlong
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Jong-Ik Hwang
- Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Hubert Vaudry
- INSERM U982, University of Rouen, Mont-Saint-Aignan, France
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, Seoul, South Korea
- *Correspondence: Jae Young Seong, Graduate School of Medicine, Korea University, Seoul 136-705, South Korea e-mail:
| |
Collapse
|
39
|
Dupré D, Tostivint H. Evolution of the gastrin-cholecystokinin gene family revealed by synteny analysis. Gen Comp Endocrinol 2014; 195:164-73. [PMID: 24231682 DOI: 10.1016/j.ygcen.2013.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/29/2013] [Accepted: 10/29/2013] [Indexed: 12/17/2022]
Abstract
Gastrin (GAST) and cholecystokinin (CCK) are two structurally and functionally related peptide hormones that exert many functions, including regulation of gastric and pancreatic secretion, feeding behaviour and energy homeostasis. GAST and CCK genes are assumed to have diverged from a common ancestral gene, over 500 million years ago in the vertebrate lineage. However, although a large number of GAST and CCK-related sequences have been identified both in vertebrate and non-vertebrate species, the evolutionary history of the GAST/CCK family remains little understood. To address this issue, we used extensive genome synteny comparisons of vertebrate chromosomes, in particular to evaluate the impact of whole-genome duplications. In the present study, we confirm that the GAST/CCK family in vertebrates is composed of two paralogous genes, namely GAST and CCK, and even three in teleosts, namely GAST, CCK1 and CCK2. We also show that the GAST and CCK genes arose by duplications of a single ancestral gene through the 2R and that the two copies of the CCK gene found in teleosts have probably been generated through the 3R. Finally, our results suggest that the vertebrate ancestor possessed four members of the GAST/CCK family, of which two have likely been lost during evolution.
Collapse
Affiliation(s)
- Délia Dupré
- UMR 7221 CNRS/MNHN Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, 75231 Paris, France
| | - Hervé Tostivint
- UMR 7221 CNRS/MNHN Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, 75231 Paris, France.
| |
Collapse
|
40
|
Fiengo M, del Gaudio R, Iazzetti G, Di Giaimo R, Minucci S, Aniello F, Donizetti A. Developmental expression pattern of two zebrafish rxfp3 paralogue genes. Dev Growth Differ 2013; 55:766-75. [PMID: 24147554 DOI: 10.1111/dgd.12093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 09/09/2013] [Accepted: 09/09/2013] [Indexed: 12/15/2022]
Abstract
In mammals, the RXFP3 is the cognate receptor of the relaxin-3 peptide (RLN3). In teleosts, many different orthologue genes for RXFP3 are present. In particular, two paralogue genes, rxfp3-2a and rxfp3-2b, likely encode the receptors for the Rln3a peptide. The transcription of these two rxfp3 genes is differentially regulated early during zebrafish embryogenesis. Indeed, reverse transcription-polymerase chain reaction analyses show that the rxfp3-2b transcript is always present during embryo development, while the rxfp3-2a transcript is detectable only at larval stage. By in situ hybridization experiments on embryos and larvae, the rxfp3-2b transcript was revealed in the brain and in the retinal ganglion cell layer and thymus. Particularly in the brain, many territories are involved in the rxfp3-2b expression, among them the optic tectum, thalamus, preoptic area, different nerve nuclei, habenula and pineal gland. The RXFP3 spatiotemporal expression pattern appears to be conserved between Danio rerio and mammals, as also previously showed for the corresponding ligand, the RLN3. Interestingly, the brain areas expressing the rxfp3-2b receptor gene are involved in the visual system, emotional behaviors and circadian rhythm and could be functionally related to the neurotransmitter Rln3a-expressing territories.
Collapse
Affiliation(s)
- Marcella Fiengo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Italy
| | - Rosanna del Gaudio
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Italy
| | - Giovanni Iazzetti
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Italy
| | - Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Italy
| | - Sergio Minucci
- Department of Experimental Medicine, Second University of Naples, Via Costantinopoli 16, Naples, 80138, Italy
| | - Francesco Aniello
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Italy
| | - Aldo Donizetti
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Italy
| |
Collapse
|
41
|
Park CR, Moon MJ, Park S, Kim DK, Cho EB, Millar RP, Hwang JI, Seong JY. A novel glucagon-related peptide (GCRP) and its receptor GCRPR account for coevolution of their family members in vertebrates. PLoS One 2013; 8:e65420. [PMID: 23776481 PMCID: PMC3679108 DOI: 10.1371/journal.pone.0065420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 04/24/2013] [Indexed: 12/25/2022] Open
Abstract
The glucagon (GCG) peptide family consists of GCG, glucagon-like peptide 1 (GLP1), and GLP2, which are derived from a common GCG precursor, and the glucose-dependent insulinotropic polypeptide (GIP). These peptides interact with cognate receptors, GCGR, GLP1R, GLP2R, and GIPR, which belong to the secretin-like G protein-coupled receptor (GPCR) family. We used bioinformatics to identify genes encoding a novel GCG-related peptide (GCRP) and its cognate receptor, GCRPR. The GCRP and GCRPR genes were found in representative tetrapod taxa such as anole lizard, chicken, and Xenopus, and in teleosts including medaka, fugu, tetraodon, and stickleback. However, they were not present in mammals and zebrafish. Phylogenetic and genome synteny analyses showed that GCRP emerged through two rounds of whole genome duplication (2R) during early vertebrate evolution. GCRPR appears to have arisen by local tandem gene duplications from a common ancestor of GCRPR, GCGR, and GLP2R after 2R. Biochemical ligand-receptor interaction analyses revealed that GCRP had the highest affinity for GCRPR in comparison to other GCGR family members. Stimulation of chicken, Xenopus, and medaka GCRPRs activated Gαs-mediated signaling. In contrast to chicken and Xenopus GCRPRs, medaka GCRPR also induced Gαq/11-mediated signaling. Chimeric peptides and receptors showed that the K16M17K18 and G16Q17A18 motifs in GCRP and GLP1, respectively, may at least in part contribute to specific recognition of their cognate receptors through interaction with the receptor core domain. In conclusion, we present novel data demonstrating that GCRP and GCRPR evolved through gene/genome duplications followed by specific modifications that conferred selective recognition to this ligand-receptor pair.
Collapse
Affiliation(s)
- Cho Rong Park
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
| | - Mi Jin Moon
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
| | - Sumi Park
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
| | - Dong-Kyu Kim
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
| | - Eun Bee Cho
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
| | - Robert Peter Millar
- Mammal Research Institute, Department of Zoology & Entomology, University of Pretoria, Hatfield, South Africa
- Medical Research Council Receptor Biology Unit, University of Cape Town, Observatory 7925, South Africa
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland
| | - Jong-Ik Hwang
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
- * E-mail: (JIH); (JYS)
| | - Jae Young Seong
- Laboratory of G-protein Coupled Receptors, Graduate School of Medicine Korea University, Seoul, Republic of Korea
- * E-mail: (JIH); (JYS)
| |
Collapse
|
42
|
Hwang JI, Moon MJ, Park S, Kim DK, Cho EB, Ha N, Son GH, Kim K, Vaudry H, Seong JY. Expansion of secretin-like G protein-coupled receptors and their peptide ligands via local duplications before and after two rounds of whole-genome duplication. Mol Biol Evol 2013; 30:1119-30. [PMID: 23427277 DOI: 10.1093/molbev/mst031] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In humans, the secretin-like G protein-coupled receptor (GPCR) family comprises 15 members with 18 corresponding peptide ligand genes. Although members have been identified in a large variety of vertebrate and nonvertebrate species, the origin and relationship of these proteins remain unresolved. To address this issue, we employed large-scale genome comparisons to identify genome fragments with conserved synteny and matched these fragments to linkage groups in reconstructed early gnathostome ancestral chromosomes (GAC). This genome comparison revealed that most receptor and peptide genes were clustered in three GAC linkage groups and suggested that the ancestral forms of five peptide subfamilies (corticotropin-releasing hormone-like, calcitonin-like, parathyroid hormone-like, glucagon-like, and growth hormone-releasing hormone-like) and their cognate receptor families emerged through tandem local gene duplications before two rounds (2R) of whole-genome duplication. These subfamily genes have, then, been amplified by 2R whole-genome duplication, followed by additional local duplications and gene loss prior to the divergence of land vertebrates and teleosts. This study delineates a possible evolutionary scenario for whole secretin-like peptide and receptor family members and may shed light on evolutionary mechanisms for expansion of a gene family with a large number of paralogs.
Collapse
Affiliation(s)
- Jong-Ik Hwang
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Mita M. Relaxin-like gonad-stimulating substance in an echinoderm, the starfish: a novel relaxin system in reproduction of invertebrates. Gen Comp Endocrinol 2013; 181:241-5. [PMID: 22841765 DOI: 10.1016/j.ygcen.2012.07.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 07/10/2012] [Accepted: 07/13/2012] [Indexed: 11/15/2022]
Abstract
Gonad-stimulating substance (GSS) in starfish is the only known invertebrate peptide hormone responsible for final gamete maturation, rendering it functionally analogous to gonadotropins in vertebrates. Recently, GSS was purified from the radial nerves of the starfish Asterina pectinifera and its chemical structure determined. This review summarizes the chemical structure of relaxin-like peptide, GSS, from a starfish as the first identified gonadotropin in invertebrates and its hormonal action on reproduction. The starfish GSS is a relaxin-like heterodimeric peptide composed of two peptides (A- and B-chains) with disulfide cross-linkages. Chemically synthesized GSS induced oocyte maturation and ovulation in vitro and an unique spawning behavior followed by release of gametes in vivo. GSS is a first trigger for oocyte maturation in starfish, but its effect is indirect because GSS acts on the ovary to produce a second mediator, 1-methyladenine (1-MeAde), as a maturation-inducing hormone of starfish. The action of GSS on ovarian follicle cells to produce 1-MeAde is mediated through the activation of its receptor, G-protein, and adenylyl cyclase. In contrast to follicle cells in a fully grown state, GSS fails to induce 1-MeAde production in growing follicle cells because of a lack of Gs-proteins. Thus, relaxin-like GSS is a major factor in the neuroendocrine cascade controlling reproduction in starfish.
Collapse
Affiliation(s)
- Masatoshi Mita
- Department of Biology, Faculty of Education, Tokyo Gakugei University, Nukuikita-machi 4-1-1, Koganei-shi, Tokyo 184-8501, Japan.
| |
Collapse
|
44
|
New insights into ligand-receptor pairing and coevolution of relaxin family peptides and their receptors in teleosts. INTERNATIONAL JOURNAL OF EVOLUTIONARY BIOLOGY 2012; 2012:310278. [PMID: 23008798 PMCID: PMC3449138 DOI: 10.1155/2012/310278] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/07/2012] [Accepted: 06/15/2012] [Indexed: 11/18/2022]
Abstract
Relaxin-like peptides (RLN/INSL) play diverse roles in reproductive and neuroendocrine processes in placental mammals and are functionally associated with two distinct types of receptors (RXFP) for each respective function. The diversification of RLN/INSL and RXFP gene families in vertebrates was predominantly driven by whole genome duplications (2R and 3R). Teleosts preferentially retained duplicates of genes putatively involved in neuroendocrine regulation, harboring a total of 10-11 receptors and 6 ligand genes, while most mammals have equal numbers of ligands and receptors. To date, the ligand-receptor relationships of teleost Rln/Insl peptides and their receptors have largely remained unexplored. Here, we use selection analyses based on sequence data from 5 teleosts and qPCR expression data from zebrafish to explore possible ligand-receptor pairings in teleosts. We find support for the hypothesis that, with the exception of RLN, which has undergone strong positive selection in mammalian lineages, the ligand and receptor genes shared between mammals and teleosts appear to have similar pairings. On the other hand, the teleost-specific receptors show evidence of subfunctionalization. Overall, this study underscores the complexity of RLN/INSL and RXFP ligand-receptor interactions in teleosts and establishes theoretical background for further experimental work in nonmammals.
Collapse
|
45
|
INSL4 Pseudogenes Help Define the Relaxin Family Repertoire in the Common Ancestor of Placental Mammals. J Mol Evol 2012; 75:73-8. [DOI: 10.1007/s00239-012-9517-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 08/03/2012] [Indexed: 10/27/2022]
|