1
|
Park J, Mok J, Park S, Kim D, Kang M, Park T, Park J. Celecoxib Enhances Oxidative Muscle Fibre Formation and Improves Muscle Functions Through Prokr1 Activation in Mice. J Cachexia Sarcopenia Muscle 2025; 16:e13704. [PMID: 39887895 PMCID: PMC11780397 DOI: 10.1002/jcsm.13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 11/21/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Muscle diseases are serious challenges to human health. Prokineticin receptor 1 (PROKR1) has emerged as a potential target to improve muscle function through increasing oxidative muscle fibres, but there are no clinically applicable synthetic PROKR1 agonists. METHODS Drugs with biological properties of prokineticin 2 (PK2) were discovered through connectivity map (CMap) analysis. Their effects on PROKR1 were evaluated using molecular docking, PROKR1 signalling and competitive binding assays. Pregnant dams were fed diets containing varying celecoxib concentrations (0, 500, 1000 and 1500 ppm) from gestation day 5 through weaning. Offspring were given high-fat diets (HFD) from weaning until 20 weeks old, and body composition, insulin resistance, energy expenditure, exercise performance and histological analysis of muscle tissues were evaluated. RESULTS Celecoxib, with a connectivity score of 64.19 to PK2 and a docking score of -9.0 to PROKR1, selectively activated Gs signalling at 4 μM of EC50 and increased NR4A2 protein levels by 1.6-fold (p < 0.01) in PROKR1-overexpressing cells. It competitively inhibited PK2 binding to PROKR1 and reduced cAMP accumulation. In murine and human myotubes, celecoxib increased Prokr1 protein levels by 1.8-fold (p < 0.05), pCreb by 1.5-fold (p < 0.05) and Nr4a2 by 1.3-fold (p < 0.05). It also elevated Myh7 index by 2.2-fold (p < 0.0001), mitochondrial content by 1.6-fold (p < 0.001) and fatty acid oxidation (FAO) activity by 4.1-fold (p < 0.05). Offspring exposed to celecoxib during pre- and postnatal muscle development exhibited activated Prokr1 signalling, enhanced oxidative muscle fibre formation and improved muscle phenotype despite HFD. At weaning, both male and female offspring showed dose-dependent increases in lean mass (> 9.35%, p < 0.001) and grip strength (< 18.0%, p < 0.01). At 12 weeks old, mice displayed a dose-dependent decrease in weight loss (> 13.3%, p < 0.05), increased lean mass (> 16.2%, p < 0.05), improved insulin resistance (> 70.4%, p < 0.0001), energy expenditure (> 173%, p < 0.0001) and grip strength (> 23.5%, p < 0.001). Celecoxib also increased Myh7-positive muscle fibre composition (> 10.8%, p < 0.05) and mitochondrial mass (> 32.8%, p < 0.05) in the gastrocnemius and soleus muscles, accompanied by significant Prokr1 signalling activation. These effects persisted in both male and female mice at 20 weeks old. CONCLUSIONS Celecoxib shows promise as a PROKR1 agonist and clinically applicable exercise mimetic for the treatment of muscular disorders.
Collapse
Affiliation(s)
- Jeong Hwan Park
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Jongsoo Mok
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Seoah Park
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Dooho Kim
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Min‐Su Kang
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Tae Sub Park
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
- Institute of Green Bio Science and TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Joonghoon Park
- Department of International Agricultural Technology, Graduate School of International Agricultural TechnologySeoul National UniversitySeoulRepublic of Korea
- Institute of Green Bio Science and TechnologySeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
2
|
Vincenzi M, Kremić A, Jouve A, Lattanzi R, Miele R, Benharouga M, Alfaidy N, Migrenne-Li S, Kanthasamy AG, Porcionatto M, Ferrara N, Tetko IV, Désaubry L, Nebigil CG. Therapeutic Potential of Targeting Prokineticin Receptors in Diseases. Pharmacol Rev 2023; 75:1167-1199. [PMID: 37684054 PMCID: PMC10595023 DOI: 10.1124/pharmrev.122.000801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 09/10/2023] Open
Abstract
The prokineticins (PKs) were discovered approximately 20 years ago as small peptides inducing gut contractility. Today, they are established as angiogenic, anorectic, and proinflammatory cytokines, chemokines, hormones, and neuropeptides involved in variety of physiologic and pathophysiological pathways. Their altered expression or mutations implicated in several diseases make them a potential biomarker. Their G-protein coupled receptors, PKR1 and PKR2, have divergent roles that can be therapeutic target for treatment of cardiovascular, metabolic, and neural diseases as well as pain and cancer. This article reviews and summarizes our current knowledge of PK family functions from development of heart and brain to regulation of homeostasis in health and diseases. Finally, the review summarizes the established roles of the endogenous peptides, synthetic peptides and the selective ligands of PKR1 and PKR2, and nonpeptide orthostatic and allosteric modulator of the receptors in preclinical disease models. The present review emphasizes the ambiguous aspects and gaps in our knowledge of functions of PKR ligands and elucidates future perspectives for PK research. SIGNIFICANCE STATEMENT: This review provides an in-depth view of the prokineticin family and PK receptors that can be active without their endogenous ligand and exhibits "constitutive" activity in diseases. Their non- peptide ligands display promising effects in several preclinical disease models. PKs can be the diagnostic biomarker of several diseases. A thorough understanding of the role of prokineticin family and their receptor types in health and diseases is critical to develop novel therapeutic strategies with safety concerns.
Collapse
Affiliation(s)
- Martina Vincenzi
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Amin Kremić
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Appoline Jouve
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Roberta Lattanzi
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Rossella Miele
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Mohamed Benharouga
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Nadia Alfaidy
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Stephanie Migrenne-Li
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Anumantha G Kanthasamy
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Marimelia Porcionatto
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Napoleone Ferrara
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Igor V Tetko
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Laurent Désaubry
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Canan G Nebigil
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| |
Collapse
|
3
|
Çiftci N, Akıncı A, Akbulut E, Çamtosun E, Dündar İ, Doğan M, Kayaş L. Clinical Characteristics and Genetic Analyses of Patients with Idiopathic Hypogonadotropic Hypogonadism. J Clin Res Pediatr Endocrinol 2023; 15:160-171. [PMID: 36700485 PMCID: PMC10234052 DOI: 10.4274/jcrpe.galenos.2023.2022-10-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Objective Idiopathic hypogonadotropic hypogonadism (IHH) is classified into two groups-Kalman syndrome and normosmic IHH (nIHH). Half of all cases can be explained by mutations in >50 genes. Targeted gene panel testing with nexrt generation sequencing (NGS) is required for patients without typical phenotypic findings. The aim was to determine the genetic etiologies of patients with IHH using NGS, including 54 IHH-associated genes, and to present protein homology modeling and protein stability analyzes of the detected variations. Methods Clinical and demographic data of 16 patients (eight female), aged between 11.6-17.8 years, from different families were assessed. All patients were followed up for a diagnosis of nIHH, had normal cranial imaging, were without anterior pituitary hormone deficiency other than gonadotropins, had no sex chromosome anomaly, had no additional disease, and underwent genetic analysis with NGS between the years 2008-2021. Rare variants were classified according to the variant interpretation framework of the American College of Medical Genetics and Genomics (ACMG)/Association for Molecular Pathology. Changes in protein structure caused by variations were modeled using RoseTTAFold and changes in protein stability resulting from variation were analyzed. Results Half of the 16 had no detectable variation. Three (18.75%) had a homozygous (pathogenic) variant in the GNRHR gene, one (6.25%) had a compound heterozygous [likely pathogenic-variants of uncertain significance (VUS)] variant in PROK2 and four (25%) each had a heterozygous (VUS) variant in HESX1, FGF8, FLRT3 and DMXL2. Protein models showed that variants interpreted as VUS according to ACMG could account for the clinical IHH. Conclusion The frequency of variation detection was similar to the literature. Modelling showed that the variant in five different genes, interpreted as VUS according to ACMG, could explain the clinical IHH.
Collapse
Affiliation(s)
- Nurdan Çiftci
- İnönü University Faculty of Medicine, Department of Pediatric Endocrinology, Malatya, Turkey
| | - Ayşehan Akıncı
- İnönü University Faculty of Medicine, Department of Pediatric Endocrinology, Malatya, Turkey
| | - Ekrem Akbulut
- Turgut Özal University Faculty of Biomedical Engineering, Malatya, Turkey
| | - Emine Çamtosun
- İnönü University Faculty of Medicine, Department of Pediatric Endocrinology, Malatya, Turkey
| | - İsmail Dündar
- İnönü University Faculty of Medicine, Department of Pediatric Endocrinology, Malatya, Turkey
| | - Mustafa Doğan
- University of Health Sciences Turkey, Başakşehir Çam and Sakura City Hospital, Clinic of Medical Genetics, İstanbul, Turkey
| | - Leman Kayaş
- İnönü University Faculty of Medicine, Department of Pediatric Endocrinology, Malatya, Turkey
| |
Collapse
|
4
|
Wang X, Chen D, Zhao Y, Men M, Chen Z, Jiang F, Zheng R, Stamou MI, Plummer L, Balasubramanian R, Li JD. A functional spectrum of PROKR2 mutations identified in isolated hypogonadotropic hypogonadism. Hum Mol Genet 2023; 32:1722-1729. [PMID: 36694982 PMCID: PMC10422949 DOI: 10.1093/hmg/ddad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/04/2022] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Isolated hypogonadotropic hypogonadism (IHH) is a rare disease with hypogonadism and infertility caused by the defects in embryonic migration of hypothalamic gonadotropin-releasing hormone (GnRH) neurons, hypothalamic GnRH secretion or GnRH signal transduction. PROKR2 gene, encoding a G-protein coupled receptor PROKR2, is one of the most frequently mutated genes identified in IHH patients. However, the functional consequences of several PROKR2 mutants remain elusive. In this study, we systematically analyzed the Gαq, Gαs and ERK1/2 signaling of 23 IHH-associated PROKR2 mutations which are yet to be functionally characterized. We demonstrate that blockage of Gαq, instead of MAPK/ERK pathway, inhibited PROK2-induced migration of PROKR2-expressing cells, implying that PROKR2-related IHH results primarily due to Gαq signaling pathway disruption. Combined with previous reports, we categorized a total of 63 IHH-associated PROKR2 mutations into four distinct groups according Gαq pathway functionality: (i) neutral (N, >80% activity); (ii) low pathogenicity (L, 50-80% activity); (iii) medium pathogenicity (M, 20-50% activity) and (iv) high pathogenicity (H, <20% activity). We further compared the cell-based functional results with in silico mutational prediction programs. Our results indicated that while Sorting Intolerant from Tolerant predictions were accurate for transmembrane region mutations, mutations localized in the intracellular and extracellular domains were accurately predicted by the Combined Annotation Dependent Depletion prediction tool. Our results thus provide a functional database that can be used to guide diagnosis and appropriate genetic counseling in IHH patients with PROKR2 mutations.
Collapse
Affiliation(s)
- Xinying Wang
- School of Life Sciences, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410078, China
| | - Danna Chen
- Department of Basic Medical Sciences, Changsha Medical University, Changsha, Hunan 410219, China
| | - Yaguang Zhao
- School of Life Sciences, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410078, China
| | - Meichao Men
- Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Zhiheng Chen
- School of Life Sciences, Central South University, Changsha, Hunan 410078, China
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Fang Jiang
- School of Life Sciences, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410078, China
| | - Ruizhi Zheng
- Department of Endocrinology, The People's Hospital of Henan Province, Zhengzhou, Henan 450003, China
| | - Maria I Stamou
- Reproductive Endocrine Unit, Massachusetts General Hospital and the Center for Reproductive Medicine, Boston, MA 02141, USA
| | - Lacey Plummer
- Reproductive Endocrine Unit, Massachusetts General Hospital and the Center for Reproductive Medicine, Boston, MA 02141, USA
| | - Ravikumar Balasubramanian
- Reproductive Endocrine Unit, Massachusetts General Hospital and the Center for Reproductive Medicine, Boston, MA 02141, USA
| | - Jia-Da Li
- School of Life Sciences, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan 410078, China
- Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Disease, Changsha, Hunan 410078, China
| |
Collapse
|
5
|
Lattanzi R, Miele R. Non-Peptide Agonists and Antagonists of the Prokineticin Receptors. Curr Issues Mol Biol 2022; 44:6323-6332. [PMID: 36547092 PMCID: PMC9776816 DOI: 10.3390/cimb44120431] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
The prokineticin family comprises a group of secreted peptides that can be classified as chemokines based on their structural features and chemotactic and immunomodulatory functions. Prokineticins (PKs) bind with high affinity to two G protein-coupled receptors (GPCRs). Prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2) are involved in a variety of physiological functions such as angiogenesis and neurogenesis, hematopoiesis, the control of hypothalamic hormone secretion, the regulation of circadian rhythm and the modulation of complex behaviors such as feeding and drinking. Dysregulation of the system leads to an inflammatory process that is the substrate for many pathological conditions such as cancer, pain, neuroinflammation and neurodegenerative diseases such as Alzheimer's and Parkinson's disease. The use of PKR's antagonists reduces PK2/PKRs upregulation triggered by various inflammatory processes, suggesting that a pharmacological blockade of PKRs may be a successful strategy to treat inflammatory/neuroinflammatory diseases, at least in rodents. Under certain circumstances, the PK system exhibits protective/neuroprotective effects, so PKR agonists have also been developed to modulate the prokineticin system.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|
6
|
Van Baelen AC, Robin P, Kessler P, Maïga A, Gilles N, Servent D. Structural and Functional Diversity of Animal Toxins Interacting With GPCRs. Front Mol Biosci 2022; 9:811365. [PMID: 35198603 PMCID: PMC8859281 DOI: 10.3389/fmolb.2022.811365] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Peptide toxins from venoms have undergone a long evolutionary process allowing host defense or prey capture and making them highly selective and potent for their target. This has resulted in the emergence of a large panel of toxins from a wide diversity of species, with varied structures and multiple associated biological functions. In this way, animal toxins constitute an inexhaustible reservoir of druggable molecules due to their interesting pharmacological properties. One of the most interesting classes of therapeutic targets is the G-protein coupled receptors (GPCRs). GPCRs represent the largest family of membrane receptors in mammals with approximately 800 different members. They are involved in almost all biological functions and are the target of almost 30% of drugs currently on the market. Given the interest of GPCRs in the therapeutic field, the study of toxins that can interact with and modulate their activity with the purpose of drug development is of particular importance. The present review focuses on toxins targeting GPCRs, including peptide-interacting receptors or aminergic receptors, with a particular focus on structural aspects and, when relevant, on potential medical applications. The toxins described here exhibit a great diversity in size, from 10 to 80 amino acids long, in disulfide bridges, from none to five, and belong to a large panel of structural scaffolds. Particular toxin structures developed here include inhibitory cystine knot (ICK), three-finger fold, and Kunitz-type toxins. We summarize current knowledge on the structural and functional diversity of toxins interacting with GPCRs, concerning first the agonist-mimicking toxins that act as endogenous agonists targeting the corresponding receptor, and second the toxins that differ structurally from natural agonists and which display agonist, antagonist, or allosteric properties.
Collapse
Affiliation(s)
- Anne-Cécile Van Baelen
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Philippe Robin
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Pascal Kessler
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Arhamatoulaye Maïga
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
- CHU Sainte Justine, Université de Montréal, Montreal, QC, Canada
| | - Nicolas Gilles
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
- *Correspondence: Denis Servent,
| |
Collapse
|
7
|
Lattanzi R, Miele R. Prokineticin-Receptor Network: Mechanisms of Regulation. Life (Basel) 2022; 12:172. [PMID: 35207461 PMCID: PMC8877203 DOI: 10.3390/life12020172] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Prokineticins are a new class of chemokine-like peptides that bind their G protein-coupled receptors, PKR1 and PKR2, and promote chemotaxis and the production of pro-inflammatory cytokines following tissue injury or infection. This review summarizes the major cellular and biochemical mechanisms of prokineticins pathway regulation that, like other chemokines, include: genetic polymorphisms; mRNA splice modulation; expression regulation at transcriptional and post-transcriptional levels; prokineticins interactions with cell-surface glycosaminoglycans; PKRs degradation, localization, post-translational modifications and oligomerization; alternative signaling responses; binding to pharmacological inhibitors. Understanding these mechanisms, which together exert substantial biochemical control and greatly enhance the complexity of the prokineticin-receptor network, leads to novel opportunities for therapeutic intervention. In this way, besides targeting prokineticins or their receptors directly, it could be possible to indirectly influence their activity by modulating their expression and localization or blocking the downstream signaling pathways.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|
8
|
Transcriptome Profiling of the Pacific Oyster Crassostrea gigas Visceral Ganglia over a Reproduction Cycle Identifies Novel Regulatory Peptides. Mar Drugs 2021; 19:md19080452. [PMID: 34436291 PMCID: PMC8398477 DOI: 10.3390/md19080452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
The neuropeptides involved in the regulation of reproduction in the Pacific oyster (Crassostrea gigas) are quite diverse. To investigate this diversity, a transcriptomic survey of the visceral ganglia (VG) was carried out over an annual reproductive cycle. RNA-seq data from 26 samples corresponding to VG at different stages of reproduction were de novo assembled to generate a specific reference transcriptome of the oyster nervous system and used to identify differentially expressed transcripts. Transcriptome mining led to the identification of novel neuropeptide precursors (NPPs) related to the bilaterian Eclosion Hormone (EH), crustacean female sex hormone/Interleukin 17, Nesfatin, neuroparsin/IGFBP, prokineticins, and urotensin I; to the protostome GNQQN, pleurin, prohormones 3 and 4, prothoracotropic hormones (PTTH), and QSamide/PXXXamide; to the lophotrochozoan CCWamide, CLCCY, HFAamide, and LXRX; and to the mollusk-specific NPPs CCCGS, clionin, FYFY, GNamide, GRWRN, GSWN, GWE, IWMPxxGYxx, LXRYamide, RTLFamide, SLRFamide, and WGAGamide. Among the complete repertoire of NPPs, no sex-biased expression was observed. However, 25 NPPs displayed reproduction stage-specific expression, supporting their involvement in the control of gametogenesis or associated metabolisms.
Collapse
|
9
|
Magnan C, Migrenne-Li S. Pleiotropic effects of prokineticin 2 in the control of energy metabolism. Biochimie 2021; 186:73-81. [PMID: 33932486 DOI: 10.1016/j.biochi.2021.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/09/2021] [Accepted: 04/24/2021] [Indexed: 11/19/2022]
Abstract
Prokineticins are family of small proteins involved in many important biological processes including food intake and control of energy balance. The prokineticin 2 (PROK2) is expressed in several peripheral tissues and areas in the central nervous system. PROK2 activates G protein-coupled receptors, namely, prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). Preclinical models exhibiting disturbances of the PROK2 pathway (at the level of PROK2 or its receptors) are characterized by changes in food intake, feeding behavior and insulin sensitivity related to a dysfunction of the energy balance control. In Humans, mutations of PROK2 and PROKR2 genes are associated to the Kallmann syndrome (KS) that affects both the hormonal reproductive axis and the sense of smell and may also lead to obesity. Moreover, plasma PROK2 concentration has been correlated with various cardiometabolic risk factors and type 2 diabetes (T2D). The present review summarizes knowledge on PROK2 structure, signaling and function focusing on its role in control of food intake and energy homeostasis.
Collapse
|
10
|
Fullone MR, Lattanzi R, Maftei D, Bonaccorsi MC, Miele R. Analysis of role of aromatic residues in extracellular loop 2 of Prokineticin receptor 2 in ligand binding probed with genetically encoded photo-crosslinkers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183549. [PMID: 33412090 DOI: 10.1016/j.bbamem.2020.183549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 01/14/2023]
Abstract
Prokineticin 2 (PK2) and Prokineticin 2 beta (PK2β), products of alternative splicing of pk2 gene, are chemokine-like proteins. While PK2 mediates its biological activities by signaling with the same efficiency through two homologous G protein coupled receptors, prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2), PK2β is able to bind specifically PKR1. Extracellular loop 2 (ECL2) of chemokine receptors is a part of a transmembrane (TM) ligand binding site. In the ECL2 of PKR2 is present, as well as in all chemokine receptors, an aromatic residue cluster, involving tryptophan 212 localized four residues after an ECL2 conserved cysteine, and Phenylalanine 198 located in the top of TM 4. In this work, the photoactivatable unnatural amino acid p-benzoyl-L-phenylalanine is incorporated by amber codon suppression technology into PKR2 in position 212. Experiments of photoactivatable cross-linking demonstrated the role of tryptophan in position 212 for binding the ligand contacting Tryptophan in position 24. We also analyzed the role of Phenylalanine 198 in the specificity of PKRs binding. The comparison of TM-bundle binding sites between PKR1 and PKR2 revealed that they are completely conserved except for one residue: valine 207 in human PKR1, which is phenylalanine 198 in human PKR2. The F198V mutation in PKR2 permits to obtain a receptor able to bind more efficiently PK2β, a ligand highly specific for PKR1.
Collapse
Affiliation(s)
- Maria Rosaria Fullone
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Daniela Maftei
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Maria Carmela Bonaccorsi
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy.
| |
Collapse
|
11
|
Bv8-Like Toxin from the Frog Venom of Amolops jingdongensis Promotes Wound Healing via the Interleukin-1 Signaling Pathway. Toxins (Basel) 2019; 12:toxins12010015. [PMID: 31905801 PMCID: PMC7020442 DOI: 10.3390/toxins12010015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/25/2019] [Accepted: 12/27/2019] [Indexed: 11/28/2022] Open
Abstract
Prokineticins are highly conserved small peptides family expressed in all vertebrates, which contain a wide spectrum of functions. In this study, a prokineticin homolog (Bv8-AJ) isolated from the venom of frog Amolops jingdongensis was fully characterized. Bv8-AJ accelerated full-thickness wounds healing of mice model by promoting the initiation and the termination of inflammatory phase. Moreover, Bv8-AJ exerted strong proliferative effect on fibroblasts and keratinocytes isolated from newborn mice by activating interleukin (IL)-1 production. Our findings indicate that Bv8 is a potent wound healing regulator and may reveal the mechanism of rapid wound-healing in amphibian skins.
Collapse
|
12
|
Negri L, Ferrara N. The Prokineticins: Neuromodulators and Mediators of Inflammation and Myeloid Cell-Dependent Angiogenesis. Physiol Rev 2018. [PMID: 29537336 DOI: 10.1152/physrev.00012.2017] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mammalian prokineticins family comprises two conserved proteins, EG-VEGF/PROK1 and Bv8/PROK2, and their two highly related G protein-coupled receptors, PKR1 and PKR2. This signaling system has been linked to several important biological functions, including gastrointestinal tract motility, regulation of circadian rhythms, neurogenesis, angiogenesis and cancer progression, hematopoiesis, and nociception. Mutations in PKR2 or Bv8/PROK2 have been associated with Kallmann syndrome, a developmental disorder characterized by defective olfactory bulb neurogenesis, impaired development of gonadotropin-releasing hormone neurons, and infertility. Also, Bv8/PROK2 is strongly upregulated in neutrophils and other inflammatory cells in response to granulocyte-colony stimulating factor or other myeloid growth factors and functions as a pronociceptive mediator in inflamed tissues as well as a regulator of myeloid cell-dependent tumor angiogenesis. Bv8/PROK2 has been also implicated in neuropathic pain. Anti-Bv8/PROK2 antibodies or small molecule PKR inhibitors ameliorate pain arising from tissue injury and inhibit angiogenesis and inflammation associated with tumors or some autoimmune disorders.
Collapse
Affiliation(s)
- Lucia Negri
- Sapienza University of Rome, Rome, Italy ; and University of California, San Diego, La Jolla, California
| | - Napoleone Ferrara
- Sapienza University of Rome, Rome, Italy ; and University of California, San Diego, La Jolla, California
| |
Collapse
|
13
|
Zhao Y, Wu J, Wang X, Jia H, Chen DN, Li JD. Prokineticins and their G protein-coupled receptors in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 161:149-179. [PMID: 30711026 DOI: 10.1016/bs.pmbts.2018.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prokineticins are two conserved small proteins (~8kDa), prokineticin 1 (PROK1; also called EG-VEGF) and prokineticin 2 (PROK2; also called Bv8), with an N-terminal AVITGA sequence and 10 cysteines forming 5 disulfide bridges. PROK1 and PROK2 bind to two highly related G protein-coupled receptors (GPCRs), prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). Prokineticins and their receptors are widely expressed. PROK1 is predominantly expressed in peripheral tissues, especially steroidogenic organs, whereas PROK2 is mainly expressed in the central nervous system and nonsteroidogenic cells of the testes. Prokineticins signaling has been implicated in several important physiological functions, including gastrointestinal smooth muscle contraction, circadian rhythm regulation, neurogenesis, angiogenesis, pain perception, mood regulation, and reproduction. Dysregulation of prokineticins signaling has been observed in a variety of diseases, such as cancer, ischemia, and neurodegeneration, in which prokineticins signaling seems to be a promising therapeutic target. Based on the phenotypes of knockout mice, PROKR2 and PROK2 have recently been identified as causative genes for idiopathic hypogonadotropic hypogonadism, a developmental disorder characterized by impaired development of gonadotropin-releasing hormone neurons and infertility. In vitro functional studies with these disease-associated PROKR2 mutations uncovered some novel features for this receptor, such as biased signaling, which may be used to understand GPCR signaling regulation in general.
Collapse
Affiliation(s)
- Yaguang Zhao
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Jiayu Wu
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Xinying Wang
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Hong Jia
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Dan-Na Chen
- Department of Basic Medical Sciences, Changsha Medical University, Changsha, China.
| | - Jia-Da Li
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China.
| |
Collapse
|
14
|
Lattanzi R, Maftei D, Negri L, Fusco I, Miele R. PK2β ligand, a splice variant of prokineticin 2, is able to modulate and drive signaling through PKR1 receptor. Neuropeptides 2018; 71:32-42. [PMID: 30253862 DOI: 10.1016/j.npep.2018.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 11/15/2022]
Abstract
Prokineticin-2 (PK2) is a secreted bioactive peptide that signals through two GPCRs, the prokineticin receptors (PKRs), and regulates a variety of biological processes including angiogenesis, immunity and nociception. The PK2 primary transcript has two alternative splice variants, PK2 and PK2L (a Long form) which is cleaved in an active peptide, named PK2β that preferentially binds to PKR1 receptor. The aim of this study was to characterize the PK2β. Using different Saccharomyces cerevisiae strains, we examined the specificity of PKR1 and PKR2 G-protein coupling following PK2β binding. Data obtained in yeast confirmed that PK2 binds both receptors, inducing a comparable response throughout a promiscuous coupling of G protein subtypes. Conversely, we demonstrated, for the first time, that PK2β preferentially binding to PKR1, activates a signaling cascade that not depends on Gαi/o coupling. The binding specificity of PK2β for PKR1 was evaluated by the analysis of PKR mutant in yeast and GST pull-down experiments, suggesting an important role of PKR1 amino-terminal region. We also evaluated the ability of PK2β to differentially activate PKR1 and/or PKR2 by in vivo nociceptive experiments and we showed that PK2β induces intense sensitization of peripheral nociceptors to painful stimuli through the activation of PKR1. To analyze PK2β-induced signal transduction, we demonstrated the inability of PK2β to induce STAT3 protein phosphorylation in organotypic primary explants from mice Dorsal Root Ganglion (DRG), an important pain station. The control of the concentration ratio between PK2β and PK2 could be one of the keys to allow the specificity of the cell response of prokineticin signaling pathway.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Dipartimento di Fisiologia e Farmacologia "Vittorio Erspamer", Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Daniela Maftei
- Dipartimento di Fisiologia e Farmacologia "Vittorio Erspamer", Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Lucia Negri
- Dipartimento di Fisiologia e Farmacologia "Vittorio Erspamer", Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Ilaria Fusco
- Dipartimento di Fisiologia e Farmacologia "Vittorio Erspamer", Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Dipartimento di Scienze Biochimiche A. Rossi Fanelli, CNR Istituto di Biologia e Patologia Molecolare, Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Rome, Italy.
| |
Collapse
|
15
|
Accorsi A, Benatti S, Ross E, Nasi M, Malagoli D. A prokineticin-like protein responds to immune challenges in the gastropod pest Pomacea canaliculata. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 72:37-43. [PMID: 28163091 DOI: 10.1016/j.dci.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 06/06/2023]
Abstract
The golden apple snail Pomacea canaliculata is an invasive pest originating from South America. It has already been found in Asia, the southern United States and more recently in the EU. Aiming to target the immune system of the snail as a way to control its spreading, we have developed organ-specific transcriptomes and looked for molecules controlling replication and differentiation of snail hemocytes. The prokineticin domain-containing protein Astakine 1 is the only cytokine known thus far capable of regulating invertebrate hematopoiesis, and we analyzed the transcriptomes looking for molecules containing a prokineticin domain. We have identified a prokineticin-like protein (PlP), that we called Pc-plp and we analyzed by real-time PCR (qPCR) its expression. In control snails, highest levels of Pc-plp were detected in the digestive gland, the ampulla (i.e., a hemocyte reservoir) and the pericardial fluid (i.e., the hematopoietic district). We tested Pc-plp expression after triggering hematopoiesis via multiple hemolymph withdrawals, or during bacterial challenge through LPS injection. In both cases a reduction of Pc-plp mRNA was observed. The multiple hemolymph withdrawals caused a significant decrease of Pc-plp mRNA in pericardial fluid and circulating hemocytes, while the LPS injection promoted the Pc-plp mRNA drop in anterior kidney, mantle and gills, organs that may act as immune barrier in molluscs. Our data indicate an important role for prokineticin domain-containing proteins as immunomodulators also in gastropods and their dynamic expression may serve as a biosensor to gauge the effectiveness of immunological interventions aimed at curtailing the spreading of the gastropod pest P. canaliculata.
Collapse
Affiliation(s)
- Alice Accorsi
- Stowers Institute for Medical Research, Kansas City, MO, USA; Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Stefania Benatti
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Eric Ross
- Stowers Institute for Medical Research, Kansas City, MO, USA; Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Davide Malagoli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
16
|
Chen S, Reichert S, Singh C, Oikonomou G, Rihel J, Prober DA. Light-Dependent Regulation of Sleep and Wake States by Prokineticin 2 in Zebrafish. Neuron 2017. [PMID: 28648499 DOI: 10.1016/j.neuron.2017.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Light affects sleep and wake behaviors by providing an indirect cue that entrains circadian rhythms and also by inducing a direct and rapid regulation of behavior. While circadian entrainment by light is well characterized at the molecular level, mechanisms that underlie the direct effect of light on behavior are largely unknown. In zebrafish, a diurnal vertebrate, we found that both overexpression and mutation of the neuropeptide prokineticin 2 (Prok2) affect sleep and wake behaviors in a light-dependent but circadian-independent manner. In light, Prok2 overexpression increases sleep and induces expression of galanin (galn), a hypothalamic sleep-inducing peptide. We also found that light-dependent, Prok2-induced sedation requires prokineticin receptor 2 (prokr2) and is strongly suppressed in galn mutants. These results suggest that Prok2 antagonizes the direct wake-promoting effect of light in zebrafish, in part through the induction of galn expression in the hypothalamus.
Collapse
Affiliation(s)
- Shijia Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sabine Reichert
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Chanpreet Singh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Grigorios Oikonomou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| | - David A Prober
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
17
|
Chen B, Yu L, Wang J, Li C, Zhao K, Zhang H. Involvement of Prokineticin 2 and Prokineticin Receptor 1 in Lipopolysaccharide-Induced Testitis in Rats. Inflammation 2017; 39:534-42. [PMID: 26490969 DOI: 10.1007/s10753-015-0277-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prokineticin 2, a newly discovered proinflammatory peptide, has been amply evidenced to be involved in the occurrence and progress of local and systematical inflammation. Although the presence of Prokineticn 2 in mammal testis has been documented clearly, research targeting the involvement of prokineticin 2 in testicular pathology, especially testitis, is rather scarce. Employing a lipopolysaccharide-induced testitis rat model, we for the first time demonstrated the expression and upregulation of prokineticin 2 in orchitis at several levels. Our effort also addressed the differential expression patterns of prokineticin 2 and interleukin-1β, a key inflammation indicator, during testitis suggesting Prokineticn 2 serves more than a proinflammatory factor in the context of testitis. Given one of the cognate receptors of prokineticin 2, prokineticin receptor 1 (PKR1) was also significantly upregulated in orchitis as discussed in the current study, it is very likely that PK2/PKR1 signaling contribute to the development of inflammation-related testicular diseases.
Collapse
Affiliation(s)
- Biao Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Yu
- Department of obstetrics, The Maternal & Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jiaojiao Wang
- Institute of Family Planning Research, Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cuiling Li
- Institute of Family Planning Research, Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Zhao
- Institute of Family Planning Research, Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huiping Zhang
- Institute of Family Planning Research, Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China. .,Institute of Family Planning Research, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, China, , 430030.
| |
Collapse
|
18
|
PK2/PKR1 Signaling Regulates Bladder Function and Sensation in Rats with Cyclophosphamide-Induced Cystitis. Mediators Inflamm 2015; 2015:289519. [PMID: 26798205 PMCID: PMC4700194 DOI: 10.1155/2015/289519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 12/15/2022] Open
Abstract
Prokineticin 2 (PK2) is a novel chemokine-like peptide with multiple proinflammatory and nociception-related activities. This study aimed to explore the potential role of PK2 in modulating bladder activity and sensation in rats with cyclophosphamide- (CYP-) induced cystitis. Changes of PK2 and prokineticin receptors (PKRs) in normal and inflamed urinary bladders were determined at several time points (4 h, 48 h, and 8 d) after CYP treatment. Combining a nonselective antagonist of prokineticin receptors (PKRA), we further evaluated the regulatory role of PK2 in modulating bladder function and visceral pain sensation via conscious cystometry and pain behavioral scoring. PK2 and prokineticin receptor 1 (PKR1), but not prokineticin receptor 2, were detected in normal and upregulated in CYP-treated rat bladders at several levels. Immunohistochemistry staining localized PKR1 primarily in the urothelium. Blocking PKRs with PKRA showed no effect on micturition reflex activity and bladder sensation in control rats while it increased the voiding volume, prolonged voiding interval, and ameliorated visceral hyperalgesia in rats suffering from CYP-induced cystitis. In conclusion, PK2/PKR1 signaling pathway contributes to the modulation of inflammation-mediated voiding dysfunction and spontaneous visceral pain. Local blockade of PKRs may represent a novel and promising therapeutic strategy for the clinical management of inflammation-related bladder diseases.
Collapse
|
19
|
Gasser A, Brogi S, Urayama K, Nishi T, Kurose H, Tafi A, Ribeiro N, Désaubry L, Nebigil CG. Discovery and cardioprotective effects of the first non-Peptide agonists of the G protein-coupled prokineticin receptor-1. PLoS One 2015; 10:e0121027. [PMID: 25831128 PMCID: PMC4382091 DOI: 10.1371/journal.pone.0121027] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/27/2015] [Indexed: 11/19/2022] Open
Abstract
Prokineticins are angiogenic hormones that activate two G protein-coupled receptors: PKR1 and PKR2. PKR1 has emerged as a critical mediator of cardiovascular homeostasis and cardioprotection. Identification of non-peptide PKR1 agonists that contribute to myocardial repair and collateral vessel growth hold promises for treatment of heart diseases. Through a combination of in silico studies, medicinal chemistry, and pharmacological profiling approaches, we designed, synthesized, and characterized the first PKR1 agonists, demonstrating their cardioprotective activity against myocardial infarction (MI) in mice. Based on high throughput docking protocol, 250,000 compounds were computationally screened for putative PKR1 agonistic activity, using a homology model, and 10 virtual hits were pharmacologically evaluated. One hit internalizes PKR1, increases calcium release and activates ERK and Akt kinases. Among the 30 derivatives of the hit compound, the most potent derivative, IS20, was confirmed for its selectivity and specificity through genetic gain- and loss-of-function of PKR1. Importantly, IS20 prevented cardiac lesion formation and improved cardiac function after MI in mice, promoting proliferation of cardiac progenitor cells and neovasculogenesis. The preclinical investigation of the first PKR1 agonists provides a novel approach to promote cardiac neovasculogenesis after MI.
Collapse
Affiliation(s)
- Adeline Gasser
- Biotechnology and Cell Signaling Laboratory (UMR 7242), CNRS-University of Strasbourg, Illkirch, France
| | - Simone Brogi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Siena, Italy
- Departments of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Kyoji Urayama
- Biotechnology and Cell Signaling Laboratory (UMR 7242), CNRS-University of Strasbourg, Illkirch, France
| | - Toshishide Nishi
- Biotechnology and Cell Signaling Laboratory (UMR 7242), CNRS-University of Strasbourg, Illkirch, France
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812–8582, Japan
| | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812–8582, Japan
| | - Andrea Tafi
- Departments of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Nigel Ribeiro
- Therapeutic Innovation Laboratory (UMR7200), CNRS-University of Strasbourg, Illkirch, France
| | - Laurent Désaubry
- Therapeutic Innovation Laboratory (UMR7200), CNRS-University of Strasbourg, Illkirch, France
| | - Canan G. Nebigil
- Biotechnology and Cell Signaling Laboratory (UMR 7242), CNRS-University of Strasbourg, Illkirch, France
| |
Collapse
|
20
|
Evans J, D'Sylva R, Volpert M, Jamsai D, Merriner DJ, Nie G, Salamonsen LA, O'Bryan MK. Endometrial CRISP3 is regulated throughout the mouse estrous and human menstrual cycle and facilitates adhesion and proliferation of endometrial epithelial cells. Biol Reprod 2015; 92:99. [PMID: 25715794 DOI: 10.1095/biolreprod.114.127480] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023] Open
Abstract
The endometrium (the mucosal lining of the uterus) is a dynamic tissue that undergoes extensive remodeling, secretory transformation in preparation for implantation of an embryo, inflammatory and proteolytic activity during menstruation, and rapid postmenstrual repair. A plethora of local factors influence these processes. Recently, a cysteine-rich protein, CRISP3, a clade of the CRISP, antigen 5, pathogenesis-related (CAP) protein superfamily, has been implicated in uterine function. The localization, regulation, and potential function of CRISP3 in both the human and mouse endometrium is described. CRISP3 localizes to the luminal and glandular epithelium of the endometrium within both species, with increased immunoreactivity during the proliferative phase of the human cycle. CRISP3 also localizes to neutrophils, particularly within the premenstrual human endometrium and during the postbreakdown repair phase of a mouse model of endometrial breakdown and repair. Endometrial CRISP3 is produced by primary human endometrial epithelial cells and secreted in vivo to accumulate in the uterine cavity. Secreted CRISP3 is more abundant in uterine lavage fluid during the proliferative phase of the menstrual cycle. Human endometrial epithelial CRISP3 is present in both a glycosylated and a nonglycosylated form in vitro and in vivo. Treatment of endometrial epithelial cells in vitro with recombinant CRISP3 enhances both adhesion and proliferation. These data suggest roles for epithelial and neutrophil-derived CRISP3 in postmenstrual endometrial repair and regeneration.
Collapse
Affiliation(s)
- Jemma Evans
- MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Rebecca D'Sylva
- MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Marianna Volpert
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Duangporn Jamsai
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Donna Jo Merriner
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Guiying Nie
- MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia
| | - Lois A Salamonsen
- MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia
| | - Moira K O'Bryan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
21
|
Brouillet S, Hoffmann P, Alfaidy N, Feige JJ. [Prokineticins: new regulatory peptides in human reproduction]. Med Sci (Paris) 2014; 30:274-9. [PMID: 24685218 DOI: 10.1051/medsci/20143003015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
During the last decade, there has been growing evidence for the involvement of prokineticins and their receptors (PROK/PROKR) in human reproduction, with multiple roles in the female and male reproductive systems. The PROK/PROKR signalling complex has been reported as a new actor in ovary, uterus, placenta, and testis physiology, with marked dysfunction in various pathological conditions such as polycystic ovary syndrome, recurrent pregnancy loss, preeclampsia, and ectopic pregnancy. Altogether, the results strongly suggest the involvement of prokineticins in spermatogenesis, oocyte competence, embryo implantation, pregnancy, and delivery, and argue for the clinical relevance of these cytokines and their receptors as diagnostic markers for several reproductive diseases.
Collapse
Affiliation(s)
- Sophie Brouillet
- Centre hospitalier universitaire de Grenoble, hôpital couple-enfant, centre d'aide médicale à la procréation, CS 10217, 38043 Grenoble Cedex 9, France - Inserm U1036, biologie du cancer et de l'infection, iRTSV, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France - Commissariat à l'énergie atomique, institut de recherche en technologie et sciences pour le vivant, 38054 Grenoble Cedex 9, France - Université Grenoble-Alpes, 38041 Grenoble, France
| | - Pascale Hoffmann
- Centre hospitalier universitaire de Grenoble, hôpital couple-enfant, centre d'aide médicale à la procréation, CS 10217, 38043 Grenoble Cedex 9, France - Inserm U1036, biologie du cancer et de l'infection, iRTSV, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France - Commissariat à l'énergie atomique, institut de recherche en technologie et sciences pour le vivant, 38054 Grenoble Cedex 9, France - Université Grenoble-Alpes, 38041 Grenoble, France
| | - Nadia Alfaidy
- Inserm U1036, biologie du cancer et de l'infection, iRTSV, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France - Commissariat à l'énergie atomique, institut de recherche en technologie et sciences pour le vivant, 38054 Grenoble Cedex 9, France - Université Grenoble-Alpes, 38041 Grenoble, France
| | - Jean-Jacques Feige
- Inserm U1036, biologie du cancer et de l'infection, iRTSV, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France - Commissariat à l'énergie atomique, institut de recherche en technologie et sciences pour le vivant, 38054 Grenoble Cedex 9, France - Université Grenoble-Alpes, 38041 Grenoble, France
| |
Collapse
|
22
|
Xiao L, Zhang C, Li X, Gong S, Hu R, Balasubramanian R, Crowley W. Jr. WF, Hastings MH, Zhou QY. Signaling role of prokineticin 2 on the estrous cycle of female mice. PLoS One 2014; 9:e90860. [PMID: 24633064 PMCID: PMC3954593 DOI: 10.1371/journal.pone.0090860] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 02/06/2014] [Indexed: 01/06/2023] Open
Abstract
The possible signaling role of prokineticin 2 (PK2) and its receptor, prokineticin receptor 2 (PKR2), on female reproduction was investigated. First, the expression of PKR2 and its co-localization with estrogen receptor (ERα) in the hypothalamus was examined. Sexually dimorphic expression of PKR2 in the preoptic area of the hypothalamus was observed. Compared to the male mice, there was more widespread PKR2 expression in the preoptic area of the hypothalamus in the female mice. The likely co-expression of PKR2 and ERα in the preoptic area of the hypothalamus was observed. The estrous cycles in female PK2-null, and PKR2-null heterozygous mice, as well as in PK2-null and PKR2-null compound heterozygous mice were examined. Loss of one copy of PK2 or PKR2 gene caused elongated and irregular estrous cycle in the female mice. The alterations in the estrous cycle were more pronounced in PK2-null and PKR2-null compound heterozygous mice. Consistent with these observations, administration of a small molecule PK2 receptor antagonist led to temporary blocking of estrous cycle at the proestrous phase in female mice. The administration of PKR2 antagonist was found to blunt the circulating LH levels. Taken together, these studies indicate PK2 signaling is required for the maintenance of normal female estrous cycles.
Collapse
Affiliation(s)
- Ling Xiao
- Department of Pharmacology, University of California, Irvine, California, United States of America
- Department of Endocrinology, Jinshan Hospital affiliated to Fudan University, Shanghai, China
| | - Chengkang Zhang
- Department of Pharmacology, University of California, Irvine, California, United States of America
| | - Xiaohan Li
- Department of Pharmacology, University of California, Irvine, California, United States of America
| | - Shiaoching Gong
- GENSAT Project, The Rockefeller University, New York, New York, United States of America
| | - Renming Hu
- Institute of Endocrinology and Diabetology, Huashan Hospital affiliated to Fudan University, Shanghai, China
| | - Ravikumar Balasubramanian
- Harvard Reproductive Endocrine Sciences Center & The Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - William F. Crowley W. Jr.
- Harvard Reproductive Endocrine Sciences Center & The Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Michael H. Hastings
- Division of Neurobiology, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Qun-Yong Zhou
- Department of Pharmacology, University of California, Irvine, California, United States of America
| |
Collapse
|
23
|
Mittal K, Ebos J, Rini B. Angiogenesis and the tumor microenvironment: vascular endothelial growth factor and beyond. Semin Oncol 2014; 41:235-51. [PMID: 24787295 DOI: 10.1053/j.seminoncol.2014.02.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our understanding of the dynamic tumor microenvironment (TME) has improved exponentially over the last few decades. In addition to traditional cytotoxic agents, anti-cancer strategies now include numerous molecular-targeted drugs that modulate distinct elements of the TME. Angiogenesis is an underlying promoter of tumor growth, invasion, and metastases. From traditional and emerging angiogenic cytokines and their receptors to novel immune checkpoint inhibitors, regulation of the tumor microenvironment is potentially key in countering tumor progression. In this article, an overview of the architecture of the TME and the orchestration of angiogenesis within the TME is provided. Additionally, traditional and novel angiogenic targets of current interest within the TME are reviewed.
Collapse
Affiliation(s)
- Kriti Mittal
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center Cleveland, OH 44195.
| | - John Ebos
- Roswell Park Cancer Institute, Buffalo, NY
| | - Brian Rini
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center Cleveland, OH 44195
| |
Collapse
|
24
|
Meczekalski B, Podfigurna-Stopa A, Smolarczyk R, Katulski K, Genazzani AR. Kallmann syndrome in women: from genes to diagnosis and treatment. Gynecol Endocrinol 2013; 29:296-300. [PMID: 23368665 DOI: 10.3109/09513590.2012.752459] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Kallmann syndrome (KS) can be characterized as genetic disorder marked by hypogonadotropic hypogonadism and anosmia. Franz Jozef Kallmann was the first who described this disease in 1944. He suggested, that this disease has hereditary background. At present, six genes are regarded as causal genes of KS. These genes can be listed in chronological order: KAL1, FGFR1, FGF8, CHD7, PROKR2 and PROK2. The sensitivity of molecular testing of KS is only about 30%. Diagnosis based on clinical findings is therefore such important. Cardinal features of patients with KS include hypogonadotropic hypogonadism and anosmia or hyposmia. Some non-reproductive, non-olfactory symptoms can also be present, depending on the genetic form of disease. Some patients with KS present midline cranial anomalies (cleft lip, cleft palate and imperfect fusion). Sometimes patients can also suffer from missing teeth (dental agenesis). Optic problems, such as colour blindness or optic atrophy also can occur in KS patients. Very characteristic symptom in KS patients is mirror movements of the upper limbs (imitation synkinesis for contralateral limbs). The type of treatment in women with KS depends on the goal of therapy. After the diagnosis of syndrome, the main goal of the treatment is to induce and maintain secondary sex characteristic (estrogen-progestin therapy). The further goal in some patients can be related to enable fertility (gonadotropin, gonadotropin-releasing hormone therapy).
Collapse
Affiliation(s)
- Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland.
| | | | | | | | | |
Collapse
|
25
|
Brouillet S, Murthi P, Hoffmann P, Salomon A, Sergent F, De Mazancourt P, Dakouane-Giudicelli M, Dieudonné MN, Rozenberg P, Vaiman D, Barbaux S, Benharouga M, Feige J, Alfaidy N. EG-VEGF controls placental growth and survival in normal and pathological pregnancies: case of fetal growth restriction (FGR). Cell Mol Life Sci 2013; 70:511-25. [PMID: 22941044 PMCID: PMC11113665 DOI: 10.1007/s00018-012-1141-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 08/08/2012] [Accepted: 08/14/2012] [Indexed: 02/05/2023]
Abstract
Identifiable causes of fetal growth restriction (FGR) account for 30 % of cases, but the remainders are idiopathic and are frequently associated with placental dysfunction. We have shown that the angiogenic factor endocrine gland-derived VEGF (EG-VEGF) and its receptors, prokineticin receptor 1 (PROKR1) and 2, (1) are abundantly expressed in human placenta, (2) are up-regulated by hypoxia, (3) control trophoblast invasion, and that EG-VEGF circulating levels are the highest during the first trimester of pregnancy, the period of important placental growth. These findings suggest that EG-VEGF/PROKR1 and 2 might be involved in normal and FGR placental development. To test this hypothesis, we used placental explants, primary trophoblast cultures, and placental and serum samples collected from FGR and age-matched control women. Our results show that (1) EG-VEGF increases trophoblast proliferation ([(3)H]-thymidine incorporation and Ki67-staining) via the homeobox-gene, HLX (2) the proliferative effect involves PROKR1 but not PROKR2, (3) EG-VEGF does not affect syncytium formation (measurement of syncytin 1 and 2 and β hCG production) (4) EG-VEGF increases the vascularization of the placental villi and insures their survival, (5) EG-VEGF, PROKR1, and PROKR2 mRNA and protein levels are significantly elevated in FGR placentas, and (6) EG-VEGF circulating levels are significantly higher in FGR patients. Altogether, our results identify EG-VEGF as a new placental growth factor acting during the first trimester of pregnancy, established its mechanism of action, and provide evidence for its deregulation in FGR. We propose that EG-VEGF/PROKR1 and 2 increases occur in FGR as a compensatory mechanism to insure proper pregnancy progress.
Collapse
Affiliation(s)
- S. Brouillet
- Laboratoire BCI -iRTSV, Institut National de la Santé et de la Recherche Médicale U1036, Biologie du Cancer et de l’Infection, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - P. Murthi
- Department of Perinatal Medicine Pregnancy Research Centre, Royal Women’s Hospital, Parkville, VIC 3052 Australia
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, University of Melbourne, Parkville, VIC 3052 Australia
| | - P. Hoffmann
- Laboratoire BCI -iRTSV, Institut National de la Santé et de la Recherche Médicale U1036, Biologie du Cancer et de l’Infection, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
- Département de Gynécologie, Obstétrique et Médecine de la Reproduction, Centre Hospitalier Régional Universitaire de Grenoble, Grenoble, France
| | - A. Salomon
- Laboratoire BCI -iRTSV, Institut National de la Santé et de la Recherche Médicale U1036, Biologie du Cancer et de l’Infection, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - F. Sergent
- Laboratoire BCI -iRTSV, Institut National de la Santé et de la Recherche Médicale U1036, Biologie du Cancer et de l’Infection, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - P. De Mazancourt
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire EA2493, Poissy, France
| | - M. Dakouane-Giudicelli
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire EA2493, Poissy, France
| | - M. N. Dieudonné
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire EA2493, Poissy, France
| | - P. Rozenberg
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire EA2493, Poissy, France
| | - D. Vaiman
- Département Génétique et Développement, Institut Cochin, Paris, France
| | - S. Barbaux
- Département Génétique et Développement, Institut Cochin, Paris, France
| | - M. Benharouga
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
- Centre National de la Recherche Scientifique, UMR 5249, Grenoble, France
| | - J.–J. Feige
- Laboratoire BCI -iRTSV, Institut National de la Santé et de la Recherche Médicale U1036, Biologie du Cancer et de l’Infection, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - N. Alfaidy
- Laboratoire BCI -iRTSV, Institut National de la Santé et de la Recherche Médicale U1036, Biologie du Cancer et de l’Infection, CEA Grenoble, 17, rue des Martyrs, 38054 Grenoble Cedex 9, France
- Commissariat à l’Energie Atomique, Institut de Recherche en Technologie et Sciences pour le Vivant, Grenoble, France
- Université Joseph Fourier, Grenoble, France
| |
Collapse
|
26
|
Curtis VF, Wang H, Yang P, McLendon RE, Li X, Zhou QY, Wang XF. A PK2/Bv8/PROK2 antagonist suppresses tumorigenic processes by inhibiting angiogenesis in glioma and blocking myeloid cell infiltration in pancreatic cancer. PLoS One 2013; 8:e54916. [PMID: 23372791 PMCID: PMC3553000 DOI: 10.1371/journal.pone.0054916] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Accepted: 12/17/2012] [Indexed: 11/18/2022] Open
Abstract
Infiltration of myeloid cells in the tumor microenvironment is often associated with enhanced angiogenesis and tumor progression, resulting in poor prognosis in many types of cancer. The polypeptide chemokine PK2 (Bv8, PROK2) has been shown to regulate myeloid cell mobilization from the bone marrow, leading to activation of the angiogenic process, as well as accumulation of macrophages and neutrophils in the tumor site. Neutralizing antibodies against PK2 were shown to display potent anti-tumor efficacy, illustrating the potential of PK2-antagonists as therapeutic agents for the treatment of cancer. In this study we demonstrate the anti-tumor activity of a small molecule PK2 antagonist, PKRA7, in the context of glioblastoma and pancreatic cancer xenograft tumor models. For the highly vascularized glioblastoma, PKRA7 was associated with decreased blood vessel density and increased necrotic areas in the tumor mass. Consistent with the anti-angiogenic activity of PKRA7 in vivo, this compound effectively reduced PK2-induced microvascular endothelial cell branching in vitro. For the poorly vascularized pancreatic cancer, the primary anti-tumor effect of PKRA7 appears to be mediated by the blockage of myeloid cell migration/infiltration. At the molecular level, PKRA7 inhibits PK2-induced expression of certain pro-migratory chemokines and chemokine receptors in macrophages. Combining PKRA7 treatment with standard chemotherapeutic agents resulted in enhanced effects in xenograft models for both types of tumor. Taken together, our results indicate that the anti-tumor activity of PKRA7 can be mediated by two distinct mechanisms that are relevant to the pathological features of the specific type of cancer. This small molecule PK2 antagonist holds the promise to be further developed as an effective agent for combinational cancer therapy.
Collapse
Affiliation(s)
- Valerie F. Curtis
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
| | - Hui Wang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
| | - Pengyuan Yang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
| | - Roger E. McLendon
- Department of Pathology, Duke University, Durham, North Carolina, United States of America
| | - Xiaohan Li
- Department of Pharmacology, University of California Irvine, Irvine, California, United States of America
| | - Qun-Yong Zhou
- Department of Pharmacology, University of California Irvine, Irvine, California, United States of America
| | - Xiao-Fan Wang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
27
|
Dodé C, Rondard P. PROK2/PROKR2 Signaling and Kallmann Syndrome. Front Endocrinol (Lausanne) 2013; 4:19. [PMID: 23596439 PMCID: PMC3624607 DOI: 10.3389/fendo.2013.00019] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/14/2013] [Indexed: 01/04/2023] Open
Abstract
Kallmann syndrome (KS) is a developmental disease that associates hypogonadism and a deficiency of the sense of smell. The reproductive phenotype of KS results from the primary interruption of the olfactory, vomeronasal, and terminal nerve fibers in the frontonasal region, which in turn disrupts the embryonic migration of neuroendocrine gonadotropin-releasing hormone (GnRH) synthesizing cells from the nose to the brain. This is a highly heterogeneous genetic disease, and mutations in any of the nine genes identified so far have been found in approximately 30% of the KS patients. PROKR2 and PROK2, which encode the G protein-coupled prokineticin receptor-2 and its ligand prokineticin-2, respectively, are two of these genes. Homozygous knockout mice for the orthologous genes exhibit a phenotype reminiscent of the KS features, but biallelic mutations in PROKR2 or PROK2 (autosomal recessive mode of disease transmission) have been found only in a minority of the patients, whereas most patients carrying mutations in these genes are heterozygotes. The mutations, mainly missense mutations, have deleterious effects on PROKR2 signaling in transfected cells, ranging from defective cell surface-targeting of the receptor to defective coupling to G proteins or impaired receptor-ligand interaction, but the same mutations have also been found in apparently unaffected individuals, which suggests a digenic/oligogenic mode of inheritance of the disease in heterozygous patients. This non-Mendelian mode of inheritance has so far been confirmed only in a few patients. However, it may account for the unusually high proportion of KS sporadic cases compared to familial cases.
Collapse
Affiliation(s)
- Catherine Dodé
- INSERM U1016, Institut Cochin, Université Paris-DescartesParis, France
- *Correspondence: Catherine Dodé, INSERM U1016, Institut Cochin, Département de génétique et développement, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France. e-mail:
| | - Philippe Rondard
- CNRS UMR5203, INSERM U661, Institut de Génomique Fonctionnelle, Université Montpellier 1, 2Montpellier, France
| |
Collapse
|
28
|
Prokineticin 2 is an endangering mediator of cerebral ischemic injury. Proc Natl Acad Sci U S A 2012; 109:5475-80. [PMID: 22431614 DOI: 10.1073/pnas.1113363109] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Stroke causes brain dysfunction and neuron death, and the lack of effective therapies heightens the need for new therapeutic targets. Here we identify prokineticin 2 (PK2) as a mediator for cerebral ischemic injury. PK2 is a bioactive peptide initially discovered as a regulator of gastrointestinal motility. Multiple biological roles for PK2 have been discovered, including circadian rhythms, angiogenesis, and neurogenesis. However, the role of PK2 in neuropathology is unknown. Using primary cortical cultures, we found that PK2 mRNA is up-regulated by several pathological stressors, including hypoxia, reactive oxygen species, and excitotoxic glutamate. Glutamate-induced PK2 expression is dependent on NMDA receptor activation and extracellular calcium. Enriched neuronal culture studies revealed that neurons are the principal source of glutamate-induced PK2. Using in vivo models of stroke, we found that PK2 mRNA is induced in the ischemic cortex and striatum. Central delivery of PK2 worsens infarct volume, whereas PK2 receptor antagonist decreases infarct volume and central inflammation while improving functional outcome. Direct central inhibition of PK2 using RNAi also reduces infarct volume. These findings indicate that PK2 can be activated by pathological stimuli such as hypoxia-ischemia and excitotoxic glutamate and identify PK2 as a deleterious mediator for cerebral ischemia.
Collapse
|
29
|
Näreoja K, Näsman J. Selective targeting of G-protein-coupled receptor subtypes with venom peptides. Acta Physiol (Oxf) 2012; 204:186-201. [PMID: 21481193 DOI: 10.1111/j.1748-1716.2011.02305.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The G-protein-coupled receptor (GPCR) family is one of the largest gene superfamilies with approx. 370 members responding to endogenous ligands in humans and a roughly equal amount of receptors sensitive to external stimuli from the surrounding. A number of receptors from this superfamily are well recognized targets for medical treatment of various disease conditions, whereas for many others the potential medical benefit of interference is still obscure. A general problem associated with GPCR research and therapeutics is the insufficient specificity of available ligands to differentiate between closely homologous receptor subtypes. In this context, venom peptides could make a significant contribution to the development of more specific drugs. Venoms from certain animals specialized in biochemical hunting contain a mixture of molecules that are directed towards a variety of membrane proteins. Peptide toxins isolated from these mixtures usually exhibit high specificity for their targets. Muscarinic toxins found from mamba snakes attracted much attention during the 1990s. These are 65-66 amino acid long peptides with a structural three-finger folding similar to the α-neurotoxins and they target the muscarinic acetylcholine receptors in a subtype-selective manner. Recently, several members of the three-finger toxins from mamba snakes as well as conotoxins from marine cone snails have been shown to selectively interact with subtypes of adrenergic receptors. In this review, we will discuss the GPCR-directed peptide toxins found from different venoms and how some of these can be useful in exploring specific roles of receptor subtypes.
Collapse
Affiliation(s)
- K Näreoja
- Department of Biosciences, Biochemistry, Åbo Akademi University, Turku, Finland
| | | |
Collapse
|
30
|
Levit A, Yarnitzky T, Wiener A, Meidan R, Niv MY. Modeling of human prokineticin receptors: interactions with novel small-molecule binders and potential off-target drugs. PLoS One 2011; 6:e27990. [PMID: 22132188 PMCID: PMC3221691 DOI: 10.1371/journal.pone.0027990] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 10/29/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND MOTIVATION The Prokineticin receptor (PKR) 1 and 2 subtypes are novel members of family A GPCRs, which exhibit an unusually high degree of sequence similarity. Prokineticins (PKs), their cognate ligands, are small secreted proteins of ∼80 amino acids; however, non-peptidic low-molecular weight antagonists have also been identified. PKs and their receptors play important roles under various physiological conditions such as maintaining circadian rhythm and pain perception, as well as regulating angiogenesis and modulating immunity. Identifying binding sites for known antagonists and for additional potential binders will facilitate studying and regulating these novel receptors. Blocking PKRs may serve as a therapeutic tool for various diseases, including acute pain, inflammation and cancer. METHODS AND RESULTS Ligand-based pharmacophore models were derived from known antagonists, and virtual screening performed on the DrugBank dataset identified potential human PKR (hPKR) ligands with novel scaffolds. Interestingly, these included several HIV protease inhibitors for which endothelial cell dysfunction is a documented side effect. Our results suggest that the side effects might be due to inhibition of the PKR signaling pathway. Docking of known binders to a 3D homology model of hPKR1 is in agreement with the well-established canonical TM-bundle binding site of family A GPCRs. Furthermore, the docking results highlight residues that may form specific contacts with the ligands. These contacts provide structural explanation for the importance of several chemical features that were obtained from the structure-activity analysis of known binders. With the exception of a single loop residue that might be perused in the future for obtaining subtype-specific regulation, the results suggest an identical TM-bundle binding site for hPKR1 and hPKR2. In addition, analysis of the intracellular regions highlights variable regions that may provide subtype specificity.
Collapse
Affiliation(s)
- Anat Levit
- Institute of Biochemistry, Food Science and Nutrition, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Talia Yarnitzky
- Institute of Biochemistry, Food Science and Nutrition, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ayana Wiener
- Institute of Biochemistry, Food Science and Nutrition, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Rina Meidan
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Masha Y. Niv
- Institute of Biochemistry, Food Science and Nutrition, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Fritz Haber Center for Molecular Dynamics, The Hebrew University of Jerusalem, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
31
|
Boulberdaa M, Urayama K, Nebigil CG. Prokineticin receptor 1 (PKR1) signalling in cardiovascular and kidney functions. Cardiovasc Res 2011; 92:191-8. [DOI: 10.1093/cvr/cvr228] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
32
|
Abstract
Abstract
Major contributions to research in hematopoiesis in invertebrate animals have come from studies in the fruit fly, Drosophila melanogaster, and the freshwater crayfish, Pacifastacus leniusculus. These animals lack oxygen-carrying erythrocytes and blood cells of the lymphoid lineage, which participate in adaptive immune defense, thus making them suitable model animals to study the regulation of blood cells of the innate immune system. This review presents an overview of crustacean blood cell formation, the role of these cells in innate immunity, and how their synthesis is regulated by the astakine cytokines. Astakines are among the first invertebrate cytokines shown to be involved in hematopoiesis, and they can stimulate the proliferation, differentiation, and survival of hematopoietic tissue cells. The astakines and their vertebrate homologues, prokineticins, share similar functions in hematopoiesis; thus, studies of astakine-induced hematopoiesis in crustaceans may not only advance our understanding of the regulation of invertebrate hematopoiesis but may also provide new evolutionary perspectives about this process.
Collapse
|
33
|
Jacobson O, Weiss ID, Niu G, Balboni G, Congiu C, Onnis V, Kiesewetter DO, Lattanzi R, Salvadori S, Chen X. Prokineticin receptor 1 antagonist PC-10 as a biomarker for imaging inflammatory pain. J Nucl Med 2011; 52:600-7. [PMID: 21421710 PMCID: PMC3629974 DOI: 10.2967/jnumed.110.084772] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Prokineticin receptor 1 (PKR1) and its ligand Bv8 were shown to be expressed in inflammation-induced pain and by tumor-supporting fibroblasts. Blocking this receptor might prove useful for reducing pain and for cancer therapy. However, there is no method to quantify the levels of these receptors in vivo. METHODS A nonpeptidic PKR1 antagonist, N-{2-[5-(4-fluoro-benzyl)-1-(4-methoxy-benzyl)-4,6-dioxo-1,4,5,6-tetrahydro-[1,3,5]triazin-2-ylamino]-ethyl}-guanidine, which contains a free guanidine group, was labeled with (18)F by reacting the guanidine function with N-succinimidyl-4-(18)F-fluorobenzoate to give the guanidinyl amide N-(4-(18)F-fluoro-benzoyl)-N'-{2-[5-(4-fluoro-benzyl)-1-(4-methoxy-benzyl)-4,6-dioxo-1,4,5,6-tetrahydro-[1,3,5]triazin-2-ylamino]-ethyl}-guanidine ((18)F-PC-10). Inflammation was induced in C57BL/6 mice by subcutaneous injection of complete Freund adjuvant in the paw. The mice were imaged with (18)F-PC-10, (18)F-FDG, and (64)Cu-pyruvaldehyde bis(4-methyl-3-thiosemicarbazone) ((64)Cu-PTSM) at 24 h after complete Freund adjuvant injection using a small-animal PET device. RESULTS (18)F-PC-10 was synthesized with a radiochemical yield of 16% ± 3% (decay-corrected). (18)F-PC-10 accumulated specifically in the inflamed paw 4- to 5-fold more than in the control paw. Compared with (18)F-PC-10, (18)F-FDG and (64)Cu-PTSM displayed higher accumulation in the inflamed paw but also had higher accumulation in the control paw, demonstrating a reduced signal-to-background ratio. (18)F-PC-10 also accumulated in PKR1-expressing organs, such as the salivary gland and gastrointestinal tract. CONCLUSION (18)F-PC-10 can be used to image PKR1, a biomarker of the inflammation process. However, the high uptake of (18)F-PC-10 in the gastrointestinal tract, due to specific uptake and the metabolic processing of this highly lipophilic molecule, would restrict its utility.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| | - Ido D. Weiss
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| | | | - Cenzo Congiu
- Department of Toxicology, University of Cagliari, Cagliari, Italy
| | - Valentina Onnis
- Department of Toxicology, University of Cagliari, Cagliari, Italy
| | - Dale O. Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology Vittorio Erspamer, Sapienza University Rome, Rome, Italy
| | - Severo Salvadori
- Department of Pharmaceutical Sciences and Biotechnology Center, University of Ferrara, Ferrara, Italy
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
34
|
Martin C, Balasubramanian R, Dwyer AA, Au MG, Sidis Y, Kaiser UB, Seminara SB, Pitteloud N, Zhou QY, Crowley WF. The role of the prokineticin 2 pathway in human reproduction: evidence from the study of human and murine gene mutations. Endocr Rev 2011; 32:225-46. [PMID: 21037178 PMCID: PMC3365793 DOI: 10.1210/er.2010-0007] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A widely dispersed network of hypothalamic GnRH neurons controls the reproductive axis in mammals. Genetic investigation of the human disease model of isolated GnRH deficiency has revealed several key genes crucial for GnRH neuronal ontogeny and GnRH secretion. Among these genes, prokineticin 2 (PROK2), and PROK2 receptor (PROKR2) have recently emerged as critical regulators of reproduction in both mice and humans. Both prok2- and prokr2-deficient mice recapitulate the human Kallmann syndrome phenotype. Additionally, PROK2 and PROKR2 mutations are seen in humans with Kallmann syndrome, thus implicating this pathway in GnRH neuronal migration. However, PROK2/PROKR2 mutations are also seen in normosmic GnRH deficiency, suggesting a role for the prokineticin signaling system in GnRH biology that is beyond neuronal migration. This observation is particularly surprising because mature GnRH neurons do not express PROKR2. Moreover, mutations in both PROK2 and PROKR2 are predominantly detected in the heterozygous state with incomplete penetrance or variable expressivity frequently seen within and across pedigrees. In some of these pedigrees, a "second hit" or oligogenicity has been documented. Besides reproduction, a pleiotropic physiological role for PROK2 is now recognized, including regulation of pain perception, circadian rhythms, hematopoiesis, and immune response. Therefore, further detailed clinical studies of patients with PROK2/PROKR2 mutations will help to map the broader biological role of the PROK2/PROKR2 pathway and identify other interacting genes/proteins that mediate its molecular effects in humans.
Collapse
Affiliation(s)
- Cecilia Martin
- Harvard Center for Reproductive Endocrine Sciences, Reproductive Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Peng Z, Tang Y, Luo H, Jiang F, Yang J, Sun L, Li JD. Disease-causing mutation in PKR2 receptor reveals a critical role of positive charges in the second intracellular loop for G-protein coupling and receptor trafficking. J Biol Chem 2011; 286:16615-22. [PMID: 21454486 DOI: 10.1074/jbc.m111.223784] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prokineticins are a pair of signal factors involved in many physiological processes by binding to two closely related G-protein-coupled receptors, PKR1 and PKR2. Recently, mutations in prokineticin 2 (PK2) and PKR2 are found to be associated with Kallmann syndrome and/or idiopathic hypogonadotropic hypogonadism, disorders characterized by delayed puberty and infertility. However, little is known how PKRs interact and activate G-proteins to elicit signal transduction. In the present study, we took advantage of one disease-associated mutation (R164Q) located in the second intracellular (IL2) loop of PKR2, to investigate the role of IL2 loop in the cell signaling, G-protein binding and receptor trafficking. R164Q mutant PKR2 showed normal cell surface expression and ligand binding capacity. However, the PKR2 signaling was abolished by R164Q mutation. We demonstrated that R164Q mutation disrupted the interaction of IL2 loop to the Gα(q), Gα(i), and Gα(16)-proteins. A positive-charged amino acid at this position is required for proper function, and the signaling efficacy and potency depend on the net amount of positive charges. We also demonstrated that the interactive partner of Arg-164 may localize in the C-terminal five residues of Gα(q)-protein. A series of mutation analysis indicated that the basic amino acids at the C terminus of IL2 loop may function cooperatively in GPCRs. Furthermore, R164Q mutation also results in minimal ligand-induced endocytosis of PKR2. As many GPCRs share structural homology in the C terminus of IL2 loop, our findings may have general application in understanding structure and function of GPCRs.
Collapse
Affiliation(s)
- Zhen Peng
- State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Prokineticins in angiogenesis and cancer. Cancer Lett 2010; 296:144-9. [DOI: 10.1016/j.canlet.2010.06.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 06/14/2010] [Accepted: 06/25/2010] [Indexed: 01/18/2023]
|
37
|
Miele R, Lattanzi R, Bonaccorsi di Patti MC, Paiardini A, Negri L, Barra D. Expression of Bv8 in Pichia pastoris to identify structural features for receptor binding. Protein Expr Purif 2010; 73:10-4. [DOI: 10.1016/j.pep.2010.04.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 03/19/2010] [Accepted: 04/13/2010] [Indexed: 11/30/2022]
|
38
|
Morales RAV, Daly NL, Vetter I, Mobli M, Napier IA, Craik DJ, Lewis RJ, Christie MJ, King GF, Alewood PF, Durek T. Chemical Synthesis and Structure of the Prokineticin Bv8. Chembiochem 2010; 11:1882-8. [DOI: 10.1002/cbic.201000330] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
39
|
Negri L, Lattanzi R, Giannini E, Melchiorri P. Modulators of pain: Bv8 and prokineticins. Curr Neuropharmacol 2010; 4:207-15. [PMID: 18615143 DOI: 10.2174/157015906778019518] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Revised: 03/06/2006] [Accepted: 03/07/2006] [Indexed: 12/14/2022] Open
Abstract
Bv8 is a small protein secreted by frog skin. Mammalian homologues of Bv8, the prokineticins PK1 and PK2, and their G-protein coupled receptors PKR1 and PKR2 have been identified and linked to several biological effects. Bv8 elicits a dose-dependent reduction in nociceptive threshold to thermal and mechanical stimuli applied to the skin of tail and paw of rats and mice and increases the sensitivity to nociceptive mediators as capsaicin and prostaglandins. The receptors for Bv8/PKs are present in a fraction of peptidergic population of C-fibre neurons, and in a fraction of A myelinated-fibre neurons. In mouse and rat dorsal root ganglia, PKR-expressing neurons also express TRPV1 and the activation of PKRs sensitises TPRV1 to the action of capsaicin. Mice lacking PKR1 gene exhibit impaired Bv8-induced hyperalgesia, develop deficient responses to noxious heat, capsaicin and protons and show reduced thermal and mechanical hypersensitivity to paw inflammation, indicating a requirement for PKR1 signalling associated with activation and sensitisation of primary afferent fibres. PKs are highly expressed by neutrophils and other inflammatory cells and must be considered as new pronociceptive mediators in inflammatory tissues. Bv8-like hyperalgesic activity was demonstrated in extracts of rat inflammatory granulocytes. Bv8 stimulate macrophage and T lymphocyte to differentiate between an inflammatory and Th1 profile indicating that Bv8/PK proteins play a role in immuno-inflammatory responses. Blockade of PKRs may represent a novel therapeutic strategy in acute and inflammatory pain conditions.
Collapse
Affiliation(s)
- Lucia Negri
- Department of Human Physiology and Pharmacology, University of Rome "La Sapienza" Roma, Italia.
| | | | | | | |
Collapse
|
40
|
Guilini C, Urayama K, Turkeri G, Dedeoglu DB, Kurose H, Messaddeq N, Nebigil CG. Divergent roles of prokineticin receptors in the endothelial cells: angiogenesis and fenestration. Am J Physiol Heart Circ Physiol 2010; 298:H844-52. [DOI: 10.1152/ajpheart.00898.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Prokineticins are secreted peptides that activate two G protein-coupled receptors: PKR1 and PKR2. Prokineticins induce angiogenesis and fenestration, but the cognate receptors involved in these functions are unknown. We hypothesized a role for prokineticin receptor signaling pathways and expression profiles in determining the selective effects of prokineticins on coronary endothelial cells (H5V). Activation of the PKR1/MAPK/Akt signaling pathway stimulates proliferation, migration, and angiogenesis in H5V cells, in which PKR1 predominates over PKR2. PKR1 was colocalized with Gα11 and was internalized following the stimulation of these cells with prokineticin-2. Knock down of PKR1 or Gα11 expression in H5V cells effectively inhibited prokineticin-2-induced vessel formation and MAPK/Akt activation, indicating a role for PKR1/Gα11 in this process. However, in conditions in which PKR2 predominated over PKR1, these cells displayed a fenestrated endothelial cell phenotype. H5V cells overexpressing PKR2 displayed large numbers of multivesicular bodies and caveolar clusters and a disruption of the distribution of zonula occluden-1 tight junction protein. Prokineticin-2 induced the colocalization of PKR2 with Gα12, and activated Gα12, which bound to zonula occluden-1 to trigger the degradation of this protein in these cells. Prokineticin-2 induced the formation of vessel-like structures by human aortic endothelial cells expressing only PKR1, and disorganized the tight junctions in human hepatic sinusoidal endothelial cells expressing only PKR2, confirming the divergent roles of these receptors. Our findings show the functional characteristics of coronary endothelial cells depend on the expression of PKR1 and PKR2 levels and the divergent signaling pathways used by these receptors.
Collapse
Affiliation(s)
- Célia Guilini
- Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, FRE3211, Centre National de la Recherche Scientifique, Université de Strasbourg, and Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Kyoji Urayama
- Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, FRE3211, Centre National de la Recherche Scientifique, Université de Strasbourg, and Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Gulen Turkeri
- Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, FRE3211, Centre National de la Recherche Scientifique, Université de Strasbourg, and Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Deniz B. Dedeoglu
- Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, FRE3211, Centre National de la Recherche Scientifique, Université de Strasbourg, and Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Kyushu University, Fukuoka, Japan; and
| | - Nadia Messaddeq
- Institut de Génétique et de Biologie Moléculaire, Illkirch, France
| | - Canan G. Nebigil
- Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, FRE3211, Centre National de la Recherche Scientifique, Université de Strasbourg, and Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
41
|
Abreu AP, Kaiser UB, Latronico AC. The role of prokineticins in the pathogenesis of hypogonadotropic hypogonadism. Neuroendocrinology 2010; 91:283-90. [PMID: 20502053 PMCID: PMC2968764 DOI: 10.1159/000308880] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 03/17/2010] [Indexed: 11/19/2022]
Abstract
The prokineticin system comprises two multifunctional secreted proteins, prokineticin-1 (PROK1) and prokineticin-2 (PROK2), and their cognate G protein-coupled receptors. The prokineticins were originally identified as endogenous regulators of gastrointestinal motility. Currently, these bioactive peptides are involved in a wide spectrum of biological functions, including angiogenesis, neurogenesis, circadian rhythms, nociception, hematopoiesis and immune response. Mice homozygous for null mutations in Prokr2 or Prok2 recapitulate the human phenotype of Kallmann syndrome, exhibiting severe atrophy of the reproductive system and hypoplastic olfactory bulbs. Indeed, the evidence from several naturally inactivating mutations in the PROK2 and PROKR2 genes in patients with Kallmann syndrome and normosmic hypogonadotropic hypogonadism also indicate the essential role of PROK2 in olfactory bulb morphogenesis and GnRH secretion in humans.
Collapse
Affiliation(s)
- Ana Paula Abreu
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular, LIM/42, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass., USA
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass., USA
| | - Ana Claudia Latronico
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular, LIM/42, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
- Ana C. Latronico, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Disciplina de Endocrinologia e Metabologia, Av. Dr. Eneas de Carvalho Aguiar, 155 2°andar Bloco 6, São Paulo, SP 05403 900 (Brasil), Tel./Fax +55 11 30697519, E-Mail
| |
Collapse
|
42
|
Balasubramanian R, Dwyer A, Seminara SB, Pitteloud N, Kaiser UB, Crowley WF. Human GnRH deficiency: a unique disease model to unravel the ontogeny of GnRH neurons. Neuroendocrinology 2010; 92:81-99. [PMID: 20606386 PMCID: PMC3214927 DOI: 10.1159/000314193] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 04/21/2010] [Indexed: 11/19/2022]
Abstract
Evolutionary survival of a species is largely a function of its reproductive fitness. In mammals, a sparsely populated and widely dispersed network of hypothalamic neurons, the gonadotropin-releasing hormone (GnRH) neurons, serve as the pilot light of reproduction via coordinated secretion of GnRH. Since it first description, human GnRH deficiency has been recognized both clinically and genetically as a heterogeneous disease. A spectrum of different reproductive phenotypes comprised of congenital GnRH deficiency with anosmia (Kallmann syndrome), congenital GnRH deficiency with normal olfaction (normosmic idiopathic hypogonadotropic hypogonadism), and adult-onset hypogonadotropic hypogonadism has been described. In the last two decades, several genes and pathways which govern GnRH ontogeny have been discovered by studying humans with GnRH deficiency. More importantly, detailed study of these patients has highlighted the emerging theme of oligogenicity and genotypic synergism, and also expanded the phenotypic diversity with the documentation of reversal of GnRH deficiency later in adulthood in some patients. The underlying genetic defect has also helped understand the associated nonreproductive phenotypes seen in some of these patients. These insights now provide practicing clinicians with targeted genetic diagnostic strategies and also impact on clinical management.
Collapse
MESH Headings
- Animals
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/genetics
- Female
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Gastrointestinal Hormones/genetics
- Gastrointestinal Hormones/metabolism
- Gonadotropin-Releasing Hormone/deficiency
- Gonadotropin-Releasing Hormone/genetics
- Humans
- Hypogonadism/genetics
- Hypothalamus/growth & development
- Kallmann Syndrome/genetics
- Male
- Mice
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Olfaction Disorders/genetics
- Phenotype
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Kisspeptin-1
- Receptors, LHRH/genetics
- Receptors, LHRH/metabolism
- Receptors, Neurokinin-3/genetics
- Receptors, Neurokinin-3/metabolism
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
| | | | | | | | | | - William F. Crowley
- *William F. Crowley, Jr., Harvard Reproductive Endocrine Sciences Center of Excellence, Massachusetts General Hospital, Bartlett Hall Extension 5th Floor, 55, Fruit Street, Boston, MA 02114 (USA), Tel. +1 617 726 5390, Fax +1 617 726 5357, E-Mail
| |
Collapse
|
43
|
Abstract
Idiopathic hypogonadotropic hypogonadism (IHH) has an incidence of 1-10 cases per 100,000 births. About 60% of patients with IHH present with associated anosmia, also known as Kallmann syndrome, characterized by total or partial loss of olfaction. Many of the gene mutations associated with Kallmann syndrome have been mapped to KAL1 or FGFR1. However, together, these mutations account for only about 15% of Kallmann syndrome cases. More recently, mutations in PROK2 and PROKR2 have been linked to the syndrome and may account for an additional 5-10% of cases. The remaining 40% of patients with IHH have a normal sense of smell. Prior to 2003, the only gene linked to normosmic IHH was the gonadotropin-releasing hormone receptor gene. However, mutations in this receptor are believed to account for only 10% of cases. Subsequently, mutations in KISS1R, TAC3 and TACR3 were identified as causes of normosmic IHH. Certain genes, including PROK2 and FGFR1, are associated with both anosmic and normosmic IHH. Despite recent advances in the field, the genetic causes of the majority of cases of IHH remain unknown. This Review discusses genes associated with hypogonadotropic disorders and the molecular mechanisms by which mutations in these genes may result in IHH.
Collapse
Affiliation(s)
- Suzy D C Bianco
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | | |
Collapse
|
44
|
The chemokine Bv8/prokineticin 2 is up-regulated in inflammatory granulocytes and modulates inflammatory pain. Proc Natl Acad Sci U S A 2009; 106:14646-51. [PMID: 19667192 DOI: 10.1073/pnas.0903720106] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neutrophil migration into injured tissues is invariably accompanied by pain. Bv8/prokineticin 2 (PK2), a chemokine characterized by a unique structural motif comprising five disulfide bonds, is highly expressed in inflamed tissues associated to infiltrating cells. Here, we demonstrate the fundamental role of granulocyte-derived PK2 (GrPK2) in initiating inflammatory pain and driving peripheral sensitization. In animal models of complete Freund's adjuvant-induced paw inflammation the development and duration of pain temporally correlated with the expression levels of PK2 in the inflamed sites. Such an increase in PK2 mRNA depends mainly on a marked up-regulation of PK2 gene transcription in granulocytes. A substantially lower up-regulation was also detected in macrophages. From a pool of peritoneal granulocytes, elicited in rats by oyster glycogen, we purified the GrPK2 protein, which displayed high affinity for the prokineticin receptors (PKRs) and, when injected into the rat paw, induced hypersensitivity to noxious stimuli as the amphibian prokineticin Bv8 did. Mice lacking PKR1 or PKR2 developed significantly less inflammation-induced hyperalgesia in comparison with WT mice, confirming the involvement of both PKRs in inflammatory pain. The inflammation-induced up-regulation of PK2 was significantly less in pkr1 null mice than in WT and pkr2 null mice, demonstrating a role of PKR1 in setting PK2 levels during inflammation. Pretreatment with a nonpeptide PKR antagonist, which preferentially binds PKR1, dose-dependently reduced and eventually abolished both prokineticin-induced hypernociception and inflammatory hyperalgesia. Inhibiting PK2 formation or antagonizing PKRs may represent another therapeutic approach for controlling inflammatory pain.
Collapse
|
45
|
Lin X, Kim YA, Lee BL, Söderhäll K, Söderhäll I. Identification and properties of a receptor for the invertebrate cytokine astakine, involved in hematopoiesis. Exp Cell Res 2009; 315:1171-80. [PMID: 19353765 DOI: 10.1016/j.yexcr.2009.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We have recently isolated an invertebrate cytokine from a freshwater crayfish, which we named astakine 1. Interestingly this protein is expressed exclusively in hemocytes and hematopoietic tissue and is essential for the release of new hemocytes into the open circulatory system of these animals. This astakine has a prokineticin (PK) domain but lacks the N-terminal AVIT amino acids and hence receptor binding may differ from vertebrate PKs. Accordingly, here we report that a receptor for astakine 1 on hematopoietic tissue (Hpt) cells is identical to the beta-subunit of F1ATP synthase. In this study we have used several different methods to clearly demonstrate that ATP-synthase is located on the plasma membrane of a subpopulation of Hpt cells and there may function as a receptor for astakine, whereas mature blood cells (hemocytes) do not have any ATP-synthase on the outside of their plasma membranes. Our results clearly show that ATP synthase beta subunits are present on the cell surface of Hpt cells and highlight the need for more detailed studies on intracellular traffic connections between mitochondria and other membrane compartments.
Collapse
Affiliation(s)
- Xionghui Lin
- Department of Comparative Physiology, Uppsala University, Uppsala, Sweden
| | | | | | | | | |
Collapse
|
46
|
Balboni G, Lazzari I, Trapella C, Negri L, Lattanzi R, Giannini E, Nicotra A, Melchiorri P, Visentin S, Nuccio CD, Salvadori S. Triazine compounds as antagonists at Bv8-prokineticin receptors. J Med Chem 2009; 51:7635-9. [PMID: 19006379 DOI: 10.1021/jm800854e] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
On the basis of a Janssen's patent, we approached a new synthesis of some 1,3,5-triazin-4,6-diones as potential non peptidic prokineticin receptor antagonists, containing the following substitutions: (N(1) and N(5) link a 4-methoxybenzyl and a 4-ethylbenzyl, respectively; C(2) can link an amino-ethyl-guanidine (reference compound 1) or an ethylendiamine (2) or an amino-ethyl-amino-2-imidazoline (3). New compounds were assessed for PKR1 and PKR2 affinity. Antagonist properties were evaluated as inhibition of 1 nM Bv8-induced intracellular Ca2+ mobilization.
Collapse
Affiliation(s)
- Gianfranco Balboni
- Department of Pharmaceutical Sciences, University of Ferrara, I-44100 Ferrara, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ji Y, Li X. Cloning and developmental expression analysis of prokineticin 2 and its receptor PKR2 in the Syrian hamster surpachiasmatic nucleus. Brain Res 2009; 1271:18-26. [PMID: 19327346 DOI: 10.1016/j.brainres.2009.03.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 02/24/2009] [Accepted: 03/05/2009] [Indexed: 10/21/2022]
Abstract
Prokineticin 2 (PK2) and its receptor (PKR2) may play important roles in transmitting circadian rhythm information from the suprachiasmatic nucleus (SCN) to downstream neural targets. In this study, we identified PK2 and PKR2 genes in the Syrian hamster through a homologous cloning approach by performing 5' and 3'-RACE. Sequence alignments indicate significant homology between the mouse, rat and Syrian hamster sequences. In situ hybridization experiments indicate that PK2 and PKR2 are expressed in the SCN, with the expression of PK2 with a pronounced amplitude change across the daily cycle under DD conditions. Recent studies indicate that the molecular machinery of the central clock in the suprachiasmatic nucleus only mature gradually during development. We thus studied the developmental expression of PK2 and PKR2 in the surpachiasmatic nuclei of the perinatal animals. PKR2 is expressed in the Syrian hamster SCN at all time points examined, while the expression of PK2 remains undetectable at prenatal time points. Expression of PK2 begins to be detectable on postnatal day 3 and onwards on day 5 and day 7. The developmental appearance of the PK2 expression rhythm may reflect the maturation process of the central clock's molecular machinery.
Collapse
Affiliation(s)
- Yaoting Ji
- College of Life Sciences, WuHan University, Room 6008, WuHan 430072, China
| | | |
Collapse
|
48
|
Monnier C, Dodé C, Fabre L, Teixeira L, Labesse G, Pin JP, Hardelin JP, Rondard P. PROKR2 missense mutations associated with Kallmann syndrome impair receptor signalling activity. Hum Mol Genet 2009; 18:75-81. [PMID: 18826963 PMCID: PMC3298864 DOI: 10.1093/hmg/ddn318] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Kallmann syndrome (KS) combines hypogonadism due to gonadotropin-releasing hormone deficiency, and anosmia or hyposmia, related to defective olfactory bulb morphogenesis. In a large series of KS patients, ten different missense mutations (p.R85C, p.R85H, p.R164Q, p.L173R, p.W178S, p.Q210R, p.R268C, p.P290S, p.M323I, p.V331M) have been identified in the gene encoding the G protein-coupled receptor prokineticin receptor-2 (PROKR2), most often in the heterozygous state. Many of these mutations were, however, also found in clinically unaffected individuals, thus raising the question of their actual implication in the KS phenotype. We reproduced each of the ten mutations in a recombinant murine Prokr2, and tested their effects on the signalling activity in transfected HEK-293 cells, by measuring intracellular calcium release upon ligand-activation of the receptor. We found that all mutated receptors except one (M323I) had decreased signalling activities. These could be explained by different defective mechanisms. Three mutations (L173R, W178S, P290S) impaired cell surface-targeting of the receptor. One mutation (Q210R) abolished ligand-binding. Finally, five mutations (R85C, R85H, R164Q, R268C, V331M) presumably impaired G protein-coupling of the receptor. In addition, when wild-type and mutant receptors were coexpressed in HEK-293 cells, none of the mutant receptors that were retained within the cells did affect cell surface-targeting of the wild-type receptor, and none of the mutant receptors properly addressed at the plasma membrane did affect wild-type receptor signalling activity. This argues against a dominant negative effect of the mutations in vivo.
Collapse
Affiliation(s)
- Carine Monnier
- CNRS UMR5203, Institut de Génomique Fonctionnelle, INSERM U661, Université Montpellier 1,2, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Negri L, Lattanzi R, Giannini E, Canestrelli M, Nicotra A, Melchiorri P. Chapter 11 Bv8/Prokineticins and their Receptors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 85:145-57. [DOI: 10.1016/s0074-7742(09)85011-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
50
|
Franchi S, Giannini E, Lattuada D, Lattanzi R, Tian H, Melchiorri P, Negri L, Panerai AE, Sacerdote P. The prokineticin receptor agonist Bv8 decreases IL-10 and IL-4 production in mice splenocytes by activating prokineticin receptor-1. BMC Immunol 2008; 9:60. [PMID: 18957080 PMCID: PMC2584092 DOI: 10.1186/1471-2172-9-60] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 10/28/2008] [Indexed: 12/03/2022] Open
Abstract
Background Bv8, prokineticin-1, or endocrine gland-vascular endothelial growth factor, and prokineticin-2 are recently isolated peptide agonists of two G protein-coupled receptors, prokineticin receptor-1 (PROKR 1) and PROKR 2, and have been described as affecting a number of myeloid cell functions. We evaluated the impact of Bv8 on lymphoid cells by investigating its ability to modulate T cell cytokine balance in mouse. Results The production of T-helper1 cytokines (IL-2, IFN-γ and IL-1β), the T-helper 2 cytokine IL-4, and the anti-inflammatory cytokine IL-10 by mouse splenocytes was evaluated after polyclonal stimulation or immunisation with the keyhole limpet hemocyanin protein antigen by measuring cytokine levels. When added in vitro to Con-A-stimulated splenocytes, Bv8 significantly increased IL-1β and decreased IL-4 and IL-10; IL-2 and IFN-γ were not affected. Similar results were obtained when Bv8 was administered in vivo. In KLH-immunised mice, splenocytes restimulated in vitro with KLH and Bv8 produced significantly smaller amounts of IL-4 and IL-10. KLH-induced IL-10 and IL-4 production was also significantly blunted in animals administered Bv8 in vivo at the time of KLH immunisation or two weeks later. The Bv8-induced effects were lost in mice lacking the PROKR 1 gene, thus indicating that PROKR 1 is the receptor involved in the modulation of cytokines. Conclusion These findings indicate that Bv8/prokineticin-1 is a novel modulator of lymphoid functions, and may be a suitable target for new immunopharmacological strategies.
Collapse
Affiliation(s)
- Silvia Franchi
- Department of Pharmacology, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|