1
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [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: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
2
|
Yan L, Li R, Li D, Zhu Y, Lv Z, Wang B. Development of a novel vasculogenic mimicry-associated gene signature for the prognostic assessment of osteosarcoma patients. Clin Transl Oncol 2023; 25:3501-3518. [PMID: 37219824 DOI: 10.1007/s12094-023-03218-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 05/06/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Osteosarcoma (OS) is a form of primary bone malignancy associated with poor prognostic outcomes. Recent work has highlighted vasculogenic mimicry (VM) as a key mechanism that supports aggressive tumor growth. The patterns of VM-associated gene expression in OS and the relationship between these genes and patient outcomes, however, have yet to be defined. METHODS Here, 48 VM-related genes were systematically assessed to examine correlations between the expression of these genes and OS patient prognosis in the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) cohort. Patients were classified into three OS subtypes. Differentially expressed genes for these three OS subtypes were then compared with hub genes detected in a weighted gene co-expression network analysis, leading to the identification of 163 overlapping genes that were subject to further biological activity analyses. A three-gene signature (CGREF1, CORT, and GALNT14) was ultimately constructed through a least absolute shrinkage and selection operator Cox regression analysis, and this signature was used to separate patients into low- and high-risk groups. The K-M survival analysis, receiver operating characteristic analysis, and decision curve analysis were adopted to evaluate the prognostic prediction performance of the signature. Furthermore, the expression patterns of three genes derived from the prognostic model were validated by quantitative real-time polymerase chain reaction (RT-qPCR). RESULTS VM-associated gene expression patterns were successfully established, and three VM subtypes of OS that were associated with patient prognosis and copy number variants were defined. The developed three-gene signature was constructed, which served as independent prognostic markers and prediction factors for the clinicopathological features of OS. Finally, lastly, the signature may also have a guiding effect on the sensitivity of different chemotherapeutic drugs. CONCLUSION Overall, these analyses facilitated the development of a prognostic VM-associated gene signature capable of predicting OS patient outcomes. This signature may be of value for both studies of the mechanistic basis for VM and clinical decision-making in the context of OS patient management.
Collapse
Affiliation(s)
- Lei Yan
- Department of Orthopaedic Surgery, The First Affliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, China
| | - Ruoqi Li
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Dijun Li
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, China
| | - Yuanyuan Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Zhi Lv
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, China.
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, China.
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Therapeutic Effect of a Latent Form of Cortistatin in Experimental Inflammatory and Fibrotic Disorders. Pharmaceutics 2022; 14:pharmaceutics14122785. [PMID: 36559278 PMCID: PMC9784182 DOI: 10.3390/pharmaceutics14122785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/01/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Cortistatin is a cyclic neuropeptide that recently emerged as an attractive therapeutic factor for treating inflammatory, autoimmune, fibrotic, and pain disorders. Despite of its efficiency and apparent safety in experimental preclinical models, its short half-life in body fluids and its potential pleiotropic effects, due to its promiscuity for several receptors expressed in various cells and tissues, represent two major drawbacks for the clinical translation of cortistatin-based therapies. Therefore, the design of new strategies focused on increasing the stability, bioavailability, and target specificity of cortistatin are lately demanded by the industry. Here, we generated by molecular engineering a new cortistatin-based prodrug formulation that includes, beside the bioactive cortistatin, a molecular-shield provided by the latency-associated protein of the transforming growth factor-β1 and a cleavage site specifically recognized by metalloproteinases, which are abundant in inflammatory/fibrotic foci. Using different models of sepsis, inflammatory bowel disease, scleroderma, and pulmonary fibrosis, we demonstrated that this latent form of cortistatin was a highly effective protection against these severe disorders. Noteworthy, from a therapeutic point of view, is that latent cortistatin seems to require significantly lower doses and fewer administrations than naive cortistatin to reach the same efficacy. Finally, the metalloproteinase-cleavage site was essential for the latent molecule to exert its therapeutic action. In summary, latent cortistatin emerges as a promising innovative therapeutic tool for treating chronic diseases of different etiologies with difficult clinical solutions and as a starting point for a rational development of prodrugs based on the use of bioactive peptides.
Collapse
|
4
|
Structure-based design of a Cortistatin analogue with immunomodulatory activity in models of inflammatory bowel disease. Nat Commun 2021; 12:1869. [PMID: 33767180 PMCID: PMC7994712 DOI: 10.1038/s41467-021-22076-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Ulcerative colitis and Crohn’s disease are forms of inflammatory bowel disease whose incidence and prevalence are increasing worldwide. These diseases lead to chronic inflammation of the gastrointestinal tract as a result of an abnormal response of the immune system. Recent studies positioned Cortistatin, which shows low stability in plasma, as a candidate for IBD treatment. Here, using NMR structural information, we design five Cortistatin analogues adopting selected native Cortistatin conformations in solution. One of them, A5, preserves the anti-inflammatory and immunomodulatory activities of Cortistatin in vitro and in mouse models of the disease. Additionally, A5 displays an increased half-life in serum and a unique receptor binding profile, thereby overcoming the limitations of the native Cortistatin as a therapeutic agent. This study provides an efficient approach to the rational design of Cortistatin analogues and opens up new possibilities for the treatment of patients that fail to respond to other therapies. Inflammatory bowel disease is caused by chronic inflammation of the gastrointestinal tract and disturbed immune responses. Here the authors present examination of Cortistatin analogues that display enhanced half-life stability whilst maintaining immunomodulatory functionality.
Collapse
|
5
|
Tostivint H, Gaillard AL, Mazan S, Pézeron G. Revisiting the evolution of the somatostatin family: Already five genes in the gnathostome ancestor. Gen Comp Endocrinol 2019; 279:139-147. [PMID: 30836103 DOI: 10.1016/j.ygcen.2019.02.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/10/2019] [Accepted: 02/28/2019] [Indexed: 11/21/2022]
Abstract
The somatostatin (SST) family members are a group of neuropeptides that are best known for their role in the regulation of growth, development and metabolism. The occurrence of six paralogous SST genes named SST1, SST2, SST3, SST4, SST5 and SST6 has been reported in vertebrates. It has been proposed that SST1, SST2 and SST5 arose in 2R from a common ancestral gene. SST3 and SST6 would have been subsequently generated by tandem duplications of the SST1 and SST2 genes respectively, at the base of the actinopterygian lineage. SST4 is thought to have appeared more recently from SST1, in teleost-specific 3R. In order to gain more insights into the SST gene family in vertebrates, we sought to identify which paralogs of this family are present in cartilaginous fish. For this purpose, we first searched the recently available genome and transcriptome databases from the catshark Scyliorhinus canicula. In a previous study, three S. canicula SST genes, called at that time SSTa, SSTb and SSTc, were identified and proposed to correspond to SST1, SST5 and SST2 respectively. In the present work, two additional SST genes, called SSTd and SSTe, were found in S. canicula plus two other chondrichtyan species, elephant shark (Callorhinchus milii) and whale shark (Rhincodon typus). Phylogeny and synteny analyses were then carried out in order to reveal the evolutionary relationships of SSTd and SSTe with other vertbrates SSTs. We showed that SSTd and SSTe correspond to SST2 and SST3 respectively, while SSTc corresponds to SST6 and not to SST2 as initially proposed. Our investigations in other vertebrate species also led us to find that the so-called SST2 gene in chicken, lungfish, sturgeons and teleosts actually corresponds to SST6. Conversely, the so-called SST6 gene in actinopterygians corresponds to SST2. Taken together, our results suggest that: i) SST3 and SST6 were already present in the gnathostome ancestor, much earlier than previously thought; ii) SST6 was also present in the tetrapod ancestor and still occurs in living birds; with this respect, it is likely that SST6 was independently lost several times during evolution: in amphibians, squamates and mammals; iii) SST2, SST3 and SST5 were probably lost in euteleosts, sarcopterygians and tetrapods, respectively.
Collapse
Affiliation(s)
- Hervé Tostivint
- Physiologie moléculaire et adaptation UMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, France.
| | - Anne-Laure Gaillard
- Physiologie moléculaire et adaptation UMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Mazan
- Biologie intégrative des organismes marins, UMR 7232 CNRS, Observatoire Océanologique, Sorbonne Université, Banyuls-sur-Mer, France
| | - Guillaume Pézeron
- Physiologie moléculaire et adaptation UMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, France
| |
Collapse
|
6
|
Günther T, Tulipano G, Dournaud P, Bousquet C, Csaba Z, Kreienkamp HJ, Lupp A, Korbonits M, Castaño JP, Wester HJ, Culler M, Melmed S, Schulz S. International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature. Pharmacol Rev 2019; 70:763-835. [PMID: 30232095 PMCID: PMC6148080 DOI: 10.1124/pr.117.015388] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.
Collapse
Affiliation(s)
- Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Giovanni Tulipano
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Pascal Dournaud
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Corinne Bousquet
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Zsolt Csaba
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Kreienkamp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Márta Korbonits
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Justo P Castaño
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Wester
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Michael Culler
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Shlomo Melmed
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| |
Collapse
|
7
|
Chen W, Fu Y, Yin X, Liu Y. Circulating levels of cortistatin are correlated with metabolic parameters in patients with newly diagnosed type 2 diabetes mellitus. Peptides 2017; 94:86-90. [PMID: 28526556 DOI: 10.1016/j.peptides.2017.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/27/2017] [Accepted: 05/15/2017] [Indexed: 11/23/2022]
Abstract
Cortistatin (CST) is a recently discovered cyclic neuropeptide with multiple bioactive effects. The aim of this study was to investigate the relationship between plasma CST and various metabolic markers in patients with newly diagnosed type 2 diabetes mellitus (T2DM). For this study, 60 patients with newly diagnosed T2DM and 38 age- and gender-matched healthy controls were recruited. Fasting plasma glucose (FPG), serum insulin and hemoglobin A1c (HbA1c) levels and a blood lipid profile were obtained with commercially available diagnostic reagents. CST plasma levels were determined using an enzyme immunoassay kit. The results showed that the plasma levels of CST were substantially lower in patients with newly diagnosed T2DM compared with the healthy controls. Plasma CST levels were positively correlated with high-density lipoprotein and negatively related to FPG, serum insulin, the homeostasis model assessment of insulin resistance (HOMA-IR) and HbA1c in all subjects. Further analysis showed that CST levels were positively correlated with systolic blood pressure and negatively correlated with FPG, serum insulin, HOMA-IR and HbA1c in patients with newly diagnosed T2DM. Moreover, logistic regression analyses indicated that plasma CST was correlated with newly diagnosed T2DM. In conclusion, patients with newly diagnosed T2DM had significantly lower plasma levels of CST than healthy controls, and plasma CST was associated with glucose metabolism and insulin resistance, indicating a potential role of CST in the development of T2DM.
Collapse
Affiliation(s)
- Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xinhua Yin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yue Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China.
| |
Collapse
|
8
|
Gruber HE, Hoelscher GL, Bethea SF, Menscher EA, Ingram JA, Templin MA, Hanley EN. Cortistatin is endogenous to the human intervertebral disc and exerts in vitro mitogenic effects on annulus cells and a downregulatory effect on TNF-α expression. Spine J 2014; 14:2995-3001. [PMID: 24912122 DOI: 10.1016/j.spinee.2014.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/29/2014] [Accepted: 06/02/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Cortistatin (CST) is a recently discovered cyclic neuropeptide with biologic anti-inflammatory properties relevant to disc degeneration. PURPOSE To test whether CST is present in the disc tissue, whether its expression is influenced by tumor necrosis factor-α (TNF-α), and whether it influences cell proliferation. STUDY DESIGN Institutional review board-approved study using immunohistochemistry on human disc tissue, in vitro annulus cultures to determine the effect of CST on cell proliferation, and the effect of TNF-α on CST gene expression. PATIENT SAMPLE Discs from 12 subjects used for immunohistochemistry, four annulus specimens used for cell culture with proinflammatory cytokines, and 11 used for cell proliferation analyses. OUTCOME MEASURES Immunohistochemical localization of CST, gene expression of CST, and cell proliferation analyses. METHODS Immunohistochemistry localized CST in disc tissue. Microarray analysis measured CST gene expression. Human annulus cells were exposed to CST for proliferation tests or cultured for the effect of TNF-α on CST expression. Standard statistical analyses were performed. RESULTS Immunohistochemistry identified CST in outer annulus, inner annulus, and nucleus tissue. Annulus cells exposed to TNF-α revealed significantly lower CST expression (p=.013). Exposure to CST significantly increased proliferation. Quantitative real-time polymerase chain reaction also confirmed expression of CST in vitro. CONCLUSIONS Data provide the first evidence that CST is present in the human disc. Addition of CST significantly increased cell proliferation. Cortistatin expression was significantly downregulated by TNF-α exposure in vitro. Findings suggest possible in vivo reduction of the anti-inflammatory actions of CST because of elevated proinflammatory cytokines during degenerating disc.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA.
| | - Gretchen L Hoelscher
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA
| | - Synthia F Bethea
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA
| | - Evan A Menscher
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA
| | - Jane A Ingram
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA
| | - Megan A Templin
- Dickson Advanced Analytics, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA
| | - Edward N Hanley
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, NC 28232, USA
| |
Collapse
|
9
|
Tostivint H, Ocampo Daza D, Bergqvist CA, Quan FB, Bougerol M, Lihrmann I, Larhammar D. Molecular evolution of GPCRs: Somatostatin/urotensin II receptors. J Mol Endocrinol 2014; 52:T61-86. [PMID: 24740737 DOI: 10.1530/jme-13-0274] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Somatostatin (SS) and urotensin II (UII) are members of two families of structurally related neuropeptides present in all vertebrates. They exert a large array of biological activities that are mediated by two families of G-protein-coupled receptors called SSTR and UTS2R respectively. It is proposed that the two families of peptides as well as those of their receptors probably derive from a single ancestral ligand-receptor pair. This pair had already been duplicated before the emergence of vertebrates to generate one SS peptide with two receptors and one UII peptide with one receptor. Thereafter, each family expanded in the three whole-genome duplications (1R, 2R, and 3R) that occurred during the evolution of vertebrates, whereupon some local duplications and gene losses occurred. Following the 2R event, the vertebrate ancestor is deduced to have possessed three SS (SS1, SS2, and SS5) and six SSTR (SSTR1-6) genes, on the one hand, and four UII (UII, URP, URP1, and URP2) and five UTS2R (UTS2R1-5) genes, on the other hand. In the teleost lineage, all these have been preserved with the exception of SSTR4. Moreover, several additional genes have been gained through the 3R event, such as SS4 and a second copy of the UII, SSTR2, SSTR3, and SSTR5 genes, and through local duplications, such as SS3. In mammals, all the genes of the SSTR family have been preserved, with the exception of SSTR6. In contrast, for the other families, extensive gene losses occurred, as only the SS1, SS2, UII, and URP genes and one UTS2R gene are still present.
Collapse
Affiliation(s)
- Hervé Tostivint
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Daniel Ocampo Daza
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Christina A Bergqvist
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Feng B Quan
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Marion Bougerol
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Isabelle Lihrmann
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Dan Larhammar
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| |
Collapse
|
10
|
Tostivint H, Quan FB, Bougerol M, Kenigfest NB, Lihrmann I. Impact of gene/genome duplications on the evolution of the urotensin II and somatostatin families. Gen Comp Endocrinol 2013; 188:110-7. [PMID: 23313073 DOI: 10.1016/j.ygcen.2012.12.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 12/22/2012] [Accepted: 12/26/2012] [Indexed: 12/12/2022]
Abstract
The present review describes the molecular evolution of two phylogenetically related families of neuropeptides, the urotensin II (UII) and somatatostatin (SS) families. The UII family consists of four paralogous genes called UII, URP, URP1 and URP2 and the SS family is composed of six paralogous genes named SS1, SS2, SS3, SS4, SS5 and SS6. All these paralogs are present in teleosts, while only four of them, UII, URP, SS1 and SS2 are detected in tetrapods. Comparative genomics showed that most of these genes, namely UII, URP, URP1 and URP2 on the one hand and SS1, SS2 and SS5 on the other hand arose through the 2R. In contrast, the teleost-specific 3R had a much more moderate impact since it only concerned the UII and SS1 genes, which once duplicated, generated a second UII copy and SS4, respectively. The two remaining genes, SS3 and SS6, arose through tandem duplications of the SS1 and SS2 genes respectively, probably in the stem lineage of actinopterygians, before the emergence of teleosts. The history of the UII and SS families has also been marked by massive gene lost, both in tetrapods and in teleosts, but only after the 3R in this latter lineage. Finally, ancestral UII and SS genes are thought to have arisen through tandem duplication of a single ancestral gene, largely before the 1R. An important challenge for the future will be to understand the physiological significance of the molecular diversity of these two families.
Collapse
Affiliation(s)
- Hervé Tostivint
- Evolution des Régulations Endocriniennes, UMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, France.
| | | | | | | | | |
Collapse
|
11
|
Peptide receptor targeting in cancer: the somatostatin paradigm. INTERNATIONAL JOURNAL OF PEPTIDES 2013; 2013:926295. [PMID: 23476673 PMCID: PMC3582104 DOI: 10.1155/2013/926295] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 12/10/2012] [Accepted: 12/28/2012] [Indexed: 02/06/2023]
Abstract
Peptide receptors involved in pathophysiological processes represent promising therapeutic targets. Neuropeptide somatostatin (SST) is produced by specialized cells in a large number of human organs and tissues. SST primarily acts as inhibitor of endocrine and exocrine secretion via the activation of five G-protein-coupled receptors, named sst1–5, while in central nervous system, SST acts as a neurotransmitter/neuromodulator, regulating locomotory and cognitive functions. Critical points of SST/SST receptor biology, such as signaling pathways of individual receptor subtypes, homo- and heterodimerization, trafficking, and cross-talk with growth factor receptors, have been extensively studied, although functions associated with several pathological conditions, including cancer, are still not completely unraveled. Importantly, SST exerts antiproliferative and antiangiogenic effects on cancer cells in vitro, and on experimental tumors in vivo. Moreover, SST agonists are clinically effective as antitumor agents for pituitary adenomas and gastro-pancreatic neuroendocrine tumors. However, SST receptors being expressed by tumor cells of various tumor histotypes, their pharmacological use is potentially extendible to other cancer types, although to date no significant results have been obtained. In this paper the most recent findings on the expression and functional roles of SST and SST receptors in tumor cells are discussed.
Collapse
|
12
|
Ocampo Daza D, Sundström G, Bergqvist CA, Larhammar D. The evolution of vertebrate somatostatin receptors and their gene regions involves extensive chromosomal rearrangements. BMC Evol Biol 2012. [PMID: 23194088 PMCID: PMC3560075 DOI: 10.1186/1471-2148-12-231] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Somatostatin and its related neuroendocrine peptides have a wide variety of physiological functions that are mediated by five somatostatin receptors with gene names SSTR1-5 in mammals. To resolve their evolution in vertebrates we have investigated the SSTR genes and a large number of adjacent gene families by phylogeny and conserved synteny analyses in a broad range of vertebrate species. Results We find that the SSTRs form two families that belong to distinct paralogons. We observe not only chromosomal similarities reflecting the paralogy relationships between the SSTR-bearing chromosome regions, but also extensive rearrangements between these regions in teleost fish genomes, including fusions and translocations followed by reshuffling through intrachromosomal rearrangements. These events obscure the paralogy relationships but are still tractable thanks to the many genomes now available. We have identified a previously unrecognized SSTR subtype, SSTR6, previously misidentified as either SSTR1 or SSTR4. Conclusions Two ancestral SSTR-bearing chromosome regions were duplicated in the two basal vertebrate tetraploidizations (2R). One of these ancestral SSTR genes generated SSTR2, -3 and -5, the other gave rise to SSTR1, -4 and -6. Subsequently SSTR6 was lost in tetrapods and SSTR4 in teleosts. Our study shows that extensive chromosomal rearrangements have taken place between related chromosome regions in teleosts, but that these events can be resolved by investigating several distantly related species.
Collapse
Affiliation(s)
- Daniel Ocampo Daza
- Department of Neuroscience, Science for Life Laboratory, Uppsala Universitet, Box 593, SE-75124 Uppsala, Sweden.
| | | | | | | |
Collapse
|
13
|
Abstract
RNA-seq is a powerful tool for the study of alternative splicing and other forms of alternative isoform expression. Understanding the regulation of these processes requires sensitive and specific detection of differential isoform abundance in comparisons between conditions, cell types, or tissues. We present DEXSeq, a statistical method to test for differential exon usage in RNA-seq data. DEXSeq uses generalized linear models and offers reliable control of false discoveries by taking biological variation into account. DEXSeq detects with high sensitivity genes, and in many cases exons, that are subject to differential exon usage. We demonstrate the versatility of DEXSeq by applying it to several data sets. The method facilitates the study of regulation and function of alternative exon usage on a genome-wide scale. An implementation of DEXSeq is available as an R/Bioconductor package.
Collapse
Affiliation(s)
- Simon Anders
- European Molecular Biology Laboratory, 69111 Heidelberg, Germany.
| | | | | |
Collapse
|
14
|
Anderson LL, Scanes CG. Nanobiology and physiology of growth hormone secretion. Exp Biol Med (Maywood) 2012; 237:126-42. [DOI: 10.1258/ebm.2011.011306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Growth hormone (GH) secretion is controlled by hypothalamic releasing hormones from the median eminence together with hormones and neuropeptides produced by peripheral organs. Secretion of GH involves movement of secretory vesicles along microtubules, transient ‘docking’ with the porosome in the cell membrane and subsequent release of GH. Release of GH is stimulated by GH releasing hormone (GHRH) and inhibited by somatostatin (SRIF). Ghrelin may be functioning to stimulate GH release from somatotropes acting via the GH secretagogue (GHS) receptor (GHSR). However, recent physiological studies militate against this. In addition, ghrelin does influence GH release acting within the hypothalamus. Release of GH from the somatotropes involves the GH-containing secretory granules moving close to the cell surface followed by transitory fusion of the secretory granules with the porosomes located in multiple secretory pits in the cell membrane. Other peptides/proteins can influence GH secretion, particularly in species of non-mammalian vertebrates.
Collapse
Affiliation(s)
- Lloyd L Anderson
- Department of Animal Science
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011
| | - Colin G Scanes
- Department of Biological Sciences, University of Wisconsin, Milwaukee, WI 53211, USA
| |
Collapse
|
15
|
Stengel A, Taché Y. Activation of somatostatin 2 receptors in the brain and the periphery induces opposite changes in circulating ghrelin levels: functional implications. Front Endocrinol (Lausanne) 2012; 3:178. [PMID: 23335913 PMCID: PMC3542632 DOI: 10.3389/fendo.2012.00178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 12/17/2012] [Indexed: 12/26/2022] Open
Abstract
Somatostatin is an important modulator of neurotransmission in the central nervous system and acts as a potent inhibitor of hormone and exocrine secretion and regulator of cell proliferation in the periphery. These pleiotropic actions occur through interaction with five G protein-coupled somatostatin receptor subtypes (sst(1) (-) (5)) that are widely expressed in the brain and peripheral organs. The characterization of somatostatin's effects can be investigated by pharmacological or genetic approaches using newly developed selective sst agonists and antagonists and mice lacking specific sst subtypes. Recent evidence points toward a divergent action of somatostatin in the brain and in the periphery to regulate circulating levels of ghrelin, an orexigenic hormone produced by the endocrine X/A-like cells in the rat gastric mucosa. Somatostatin interacts with the sst(2) in the brain to induce an increase in basal ghrelin plasma levels and counteracts the visceral stress-related decrease in circulating ghrelin. By contrast, stimulation of peripheral somatostatin-sst(2) signaling results in the inhibition of basal ghrelin release and mediates the postoperative decrease in circulating ghrelin. The peripheral sst(2)-mediated reduction of plasma ghrelin is likely to involve a paracrine action of D cell-derived somatostatin acting on sst(2) bearing X/A-like ghrelin cells in the gastric mucosa. The other member of the somatostatin family, named cortistatin, in addition to binding to sst(1) (-) (5) also directly interacts with the ghrelin receptor and therefore may simultaneously modulate ghrelin release and actions at target sites bearing ghrelin receptors representing a link between the ghrelin and somatostatin systems.
Collapse
Affiliation(s)
- Andreas Stengel
- Division Psychosomatic Medicine and Psychotherapy, Department of Medicine, Obesity Center Berlin, Charité, Universitätsmedizin BerlinBerlin, Germany
- *Correspondence: Andreas Stengel, Division Psychosomatic Medicine and Psychotherapy, Department of Medicine, Obesity Center Berlin, Charité, Universitätsmedizin Berlin, Luisenstr. 13a, 10117 Berlin, Germany. e-mail: ; Yvette Taché, Digestive Diseases Division, CURE: Digestive Diseases Research Center, Center for Neurobiology of Stress and Women’s Health, Department of Medicine, VA Greater Los Angeles Health Care System, University of California at Los Angeles, CURE Building 115, Room 117, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA. e-mail:
| | - Yvette Taché
- Digestive Diseases Division, CURE: Digestive Diseases Research Center, Center for Neurobiology of Stress and Women’s Health, Department of Medicine, VA Greater Los Angeles Health Care System, University of California at Los AngelesLos Angeles, CA, USA
- *Correspondence: Andreas Stengel, Division Psychosomatic Medicine and Psychotherapy, Department of Medicine, Obesity Center Berlin, Charité, Universitätsmedizin Berlin, Luisenstr. 13a, 10117 Berlin, Germany. e-mail: ; Yvette Taché, Digestive Diseases Division, CURE: Digestive Diseases Research Center, Center for Neurobiology of Stress and Women’s Health, Department of Medicine, VA Greater Los Angeles Health Care System, University of California at Los Angeles, CURE Building 115, Room 117, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA. e-mail:
| |
Collapse
|
16
|
Martinowich K, Schloesser RJ, Jimenez DV, Weinberger DR, Lu B. Activity-dependent brain-derived neurotrophic factor expression regulates cortistatin-interneurons and sleep behavior. Mol Brain 2011; 4:11. [PMID: 21388527 PMCID: PMC3061911 DOI: 10.1186/1756-6606-4-11] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/09/2011] [Indexed: 01/19/2023] Open
Abstract
Background Sleep homeostasis is characterized by a positive correlation between sleep length and intensity with the duration of the prior waking period. A causal role for brain-derived neurotrophic factor (BDNF) in sleep homeostasis has been suggested, but the underlying mechanisms remain unclear. Cortistatin, a neuropeptide expressed primarily in a subset of cortical GABAergic interneurons, is another molecule implicated in sleep homeostasis. Results We confirmed that sleep deprivation leads to an increase in cortical cortistatin mRNA expression. Disruption of activity-dependent BDNF expression in a genetically modified mouse line impairs both baseline levels of cortistatin mRNA as well as its levels following sleep deprivation. Disruption of activity-dependent BDNF also leads to a decrease in sleep time during the active (dark) phase. Conclusion Our studies suggest that regulation of cortistatin-expressing interneurons by activity-dependent BDNF expression may contribute to regulation of sleep behavior.
Collapse
Affiliation(s)
- Keri Martinowich
- Genes, Cognition and Psychosis Program (GCAP), National Institute of Mental Health (NIMH), Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
17
|
Molecular and expression characterization of two somatostatin genes in the Chinese sturgeon, Acipenser sinensis. Comp Biochem Physiol A Mol Integr Physiol 2009; 154:127-34. [DOI: 10.1016/j.cbpa.2009.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 05/07/2009] [Accepted: 05/19/2009] [Indexed: 11/23/2022]
|
18
|
Van Op den Bosch J, Adriaensen D, Van Nassauw L, Timmermans JP. The role(s) of somatostatin, structurally related peptides and somatostatin receptors in the gastrointestinal tract: a review. ACTA ACUST UNITED AC 2009; 156:1-8. [PMID: 19362110 DOI: 10.1016/j.regpep.2009.04.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 04/05/2009] [Indexed: 12/19/2022]
Abstract
Extensive functional and morphological research has demonstrated the pivotal role of somatostatin (SOM) in the regulation of a wide variety of gastrointestinal activities. In addition to its profound inhibitory effects on gastrointestinal motility and exocrine and endocrine secretion processes along the entire gastrointestinal tract, SOM modulates several organ-specific activities. In contrast to these well-known SOM-dependent effects, knowledge on the SOM receptors (SSTR) involved in these effects is much less conclusive. Experimental data on the identities of the SSTRs, although species- and tissue-dependent, point towards the involvement of multiple receptor subtypes in the vast majority of gastrointestinal SOM-mediated effects. Recent evidence demonstrating the role of SOM in intestinal pathologies has extended the interest of gastrointestinal research in this peptide even further. More specifically, SOM is supposed to suppress intestinal inflammatory responses by interfering with the extensive bidirectional communication between mucosal mast cells and neurons. This way, SOM not only acts as a powerful inhibitor of the inflammatory cascade at the site of inflammation, but exerts a profound antinociceptive effect through the modulation of extrinsic afferent nerve fibres. The combination of these physiological and pathological activities opens up new opportunities to explore the potential of stable SOM analogues in the treatment of GI inflammatory pathologies.
Collapse
Affiliation(s)
- Joeri Van Op den Bosch
- Laboratory of Cell Biology & Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | |
Collapse
|
19
|
Dalm VASH, Hofland LJ, Lamberts SWJ. Future clinical prospects in somatostatin/cortistatin/somatostatin receptor field. Mol Cell Endocrinol 2008; 286:262-77. [PMID: 17942217 DOI: 10.1016/j.mce.2007.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 08/13/2007] [Accepted: 09/07/2007] [Indexed: 01/31/2023]
Abstract
Somatostatin receptors (sst), somatostatin (SS) and cortistatin (CST) are widely expressed in the various systems in the human and rodent organisms and are "responsible" for maintaining homeostasis, which is essential for survival. Because of their broad expression pattern sst, SS and CST interactions may play regulatory roles in both physiology and pathophysiology in mammalian organisms. SS analogue treatment strategies as well as the use of SS analogues for diagnostic purposes have been established in diseases of different origins. This review focuses on the currently determined role for SS analogues in today's clinical practice and the potential clinical prospects for SS, CST and sst interactions in the future, with a focus on neuroendocrine and non-neuroendocrine tumours and immune-mediated diseases. Moreover, the role of new SS analogues and new insights in sst physiology will be discussed.
Collapse
Affiliation(s)
- V A S H Dalm
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | | | | |
Collapse
|
20
|
Volante M, Rosas R, Allìa E, Granata R, Baragli A, Muccioli G, Papotti M. Somatostatin, cortistatin and their receptors in tumours. Mol Cell Endocrinol 2008; 286:219-29. [PMID: 18248880 DOI: 10.1016/j.mce.2007.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 07/30/2007] [Accepted: 12/01/2007] [Indexed: 01/13/2023]
Abstract
Somatostatin (SS) and its synthetic analogs have a role in the treatment of neuroendocrine tumours both in terms of symptoms control and antiproliferative activities. These effects are mediated by five SS receptors, widely expressed in both human neuroendocrine and non-neuroendocrine tumours, which were demonstrated to be diagnostically and therapeutically valuable targets. Cortistatin (CST), a brain cortex peptide, partially homologous to SS and having similar functions is also expressed in peripheral tissues and tumours. CST binds all SS receptors, and, differently from SS, also the ghrelin receptor GHSR1a and the CST specific receptor MrgX2. The expression profile of CST is mostly restricted to neuroendocrine tumours (gastrointestinal, pancreas, lung, parathyroid, thyroid, adrenal). In these tumours, CST probably acts via the SS or ghrelin receptor, the MrgX2 receptor being absent. Thus, in comparison to SS analogs, CST synthetic analogs may represent additional diagnostic/therapeutic tools in those tumours expressing the receptors for SS, for ghrelin or for both peptides.
Collapse
Affiliation(s)
- M Volante
- Department of Clinical & Biological Sciences, University of Turin at San Luigi Hospital, Orbassano, University of Turin, Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
21
|
Siehler S, Nunn C, Hannon J, Feuerbach D, Hoyer D. Pharmacological profile of somatostatin and cortistatin receptors. Mol Cell Endocrinol 2008; 286:26-34. [PMID: 18243519 DOI: 10.1016/j.mce.2007.12.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 12/06/2007] [Accepted: 12/12/2007] [Indexed: 11/24/2022]
Abstract
Somatostatin (SRIF) and cortistatin (CST) are two endogenous peptides with high sequence similarities that act as hormones/neurotransmitters both in the CNS and the periphery; their genes although distinct result from gene duplication. Their receptors appear to be common, since the five known SRIF receptors (sst1-sst5) have similar subnanomolar affinity for SRIF and CST, whether the short (SRIF-14, CST-14, CST-17) or the long versions (SRIF-28, CST-29) of the peptides. Whether CST targets specific receptors not shared by SRIF, is still debated: MrgX2 has been described as a selective CST receptor, with submicromolar affinity for CST but devoid of affinity for SRIF; however the distribution of CST and MrgX2 is largely different, and there is no MrgX2 in rodents. A similar situation arises with the GHS receptor GHS-R1a, which displays some preferential affinity for CST over SRIF, but for which there is no evidence that it is activated by CST in vivo. In both cases, one may argue that submicromolar affinity is not the norm of a GPCR for its endogenous neuropeptide. On the other hand, all receptors known to bind SRIF have similar high affinity for CST and both peptides act as potent agonists at the sst1-sst5 receptors, whichever transduction pathway is considered. In addition, [(125)I][Tyr(10)]CST(14) labels sst1-sst5 receptors with subnanomolar affinity, and [(125)I][Tyr(10)]CST(14) binding in the brain is overlapping with that of [(125)I][Tyr(0)]SRIF(14). The functional differences reported that distinguish CST from SRIF, have not been explained convincingly and may relate to ligand-driven transductional selectivity, and other complicating factors such as receptor dimerisation, (homo or heterodimerisation), and/or the influence of accessory proteins (GIPs, RAMPS), which remain to be studied in more detail.
Collapse
Affiliation(s)
- Sandra Siehler
- Neuroscience Research, Novartis Institutes for BioMedical Research Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Tostivint H, Lihrmann I, Vaudry H. New insight into the molecular evolution of the somatostatin family. Mol Cell Endocrinol 2008; 286:5-17. [PMID: 18406049 DOI: 10.1016/j.mce.2008.02.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 02/26/2008] [Accepted: 02/28/2008] [Indexed: 12/11/2022]
Abstract
The present review describes the molecular evolution of the somatostatin family and its relationships with that of the urotensin II family. Most of the somatostatin sequences collected from different vertebrate species can be grouped as the products of at least four loci. The somatostatin 1 (SS1) gene is present in all vertebrate classes from agnathans to mammals. The SS1 gene has given rise to the somatostatin 2 (SS2) gene by a segment/chromosome duplication that is probably the result of a tetraploidization event according to the 2R hypothesis. The somatostatin-related peptide cortistatin, first identified in rodents and human, is the counterpart of SS2 in placental mammals. In fish, the existence of two additional somatostatin genes has been reported. The first gene, which encodes a peptide usually named somatostatin II (SSII), exists in almost all teleost species investigated so far and is thought to have arisen through local duplication of the SS1 gene. The second gene, which has been characterized in only a few teleost species, encodes a peptide also named SSII that exhibits a totally atypical structure. The origin of this gene is currently unknown. Nevertheless, because the two latter genes are clearly paralogous genes, we propose to rename them SS3 and SS4, respectively, in order to clarify the current confusing nomenclature. The urotensin II family consists of two genes, namely the urotensin II (UII) gene and the UII-related peptide (URP) gene. Both UII and URP exhibit limited structural identity to somatostatin so that UII was originally described as a "somatostatin-like peptide". Recent comparative genomics studies have revealed that the SS1 and URP genes, on the one hand, and the SS2 and UII genes, on the other hand, are closely linked on the same chromosomes, thus confirming that the SS1/SS2 and the UII/URP genes belong to the same superfamily. According to these data, it appears that an ancestral somatostatin/urotensin II gene gave rise by local duplication to a somatostatin ancestor and a urotensin II ancestor, whereupon this pair was duplicated (presumably by a segment/chromosome duplication) to give rise to the SS1-UII pair and the SS2-URP pair.
Collapse
Affiliation(s)
- Hervé Tostivint
- INSERM U413, Laboratory of Cellular and Molecular Neuroendocrinology, University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | |
Collapse
|
23
|
Broglio F, Grottoli S, Arvat E, Ghigo E. Endocrine actions of cortistatin: in vivo studies. Mol Cell Endocrinol 2008; 286:123-7. [PMID: 18281148 DOI: 10.1016/j.mce.2007.12.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 12/11/2007] [Accepted: 12/18/2007] [Indexed: 11/20/2022]
Abstract
Cortistatin (CST) shares high structural homology with somatostatin (SST) and binds all SST-receptors (SST-R) subtypes with similar affinity. However, CST actions, tissue expression patterns and regulation do not fully overlap with those of SST, and, moreover, CST, but not SST, also binds and activates proadrenomedullin N-terminal peptide receptor (MrgX2) and shows binding affinity to ghrelin receptor (GHS-R1a). Several studies performed to clarify the endocrine actions of CST, compared with SST, showed that, in humans, CST and SST share the same endocrine actions, i.e. inhibition of GH and insulin secretion in physiological conditions and in acromegaly. A similar inhibitory effect on PRL and ACTH secretion was shown in acromegaly, prolactinoma or in Cushing's disease. This identity of endocrine actions by CST and SST suggests that SST-R activation by CST overrides any other independent action of this peptide mediated by other receptors. Thus, in terms of endocrine actions, CST can well be considered a natural alternative to SST.
Collapse
Affiliation(s)
- Fabio Broglio
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Turin, Torino, Italy.
| | | | | | | |
Collapse
|
24
|
de Lecea L. Cortistatin--functions in the central nervous system. Mol Cell Endocrinol 2008; 286:88-95. [PMID: 18374474 DOI: 10.1016/j.mce.2007.12.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 11/29/2007] [Accepted: 12/19/2007] [Indexed: 11/22/2022]
Abstract
Cortistatin (CST) is a neuropeptide from the somatostatin (SRIF)/urotensin (UII) family named after its predominantly cortical expression and ability to depress cortical activity, which was discovered a decade ago. In vitro assays show CST is able to bind all five cloned somatostatin receptors and shares many pharmacological and functional properties with SRIF. However, distinct from SRIF, CST has been shown to induce slow-wave sleep, reduce locomotor activity, and activate cation selective currents not responsive to somatostatin. Different lines of evidence also indicate that CST, like SRIF, is involved in learning and memory processes. CST-14 may also function as an endogenous anti-convulsant. In addition to its role in cortical synchronization, CST-14 has emerged as an important mediator of immunity and inflammation. This review will cover some of the basic properties of CST in the brain, and will discuss new data on the role of CST in cortical activity.
Collapse
Affiliation(s)
- Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94304, USA.
| |
Collapse
|
25
|
Padova H, Rubinfeld H, Hadani M, Cohen ZR, Nass D, Taylor JE, Culler MD, Shimon I. Effects of selective somatostatin analogs and cortistatin on cell viability in cultured human non-functioning pituitary adenomas. Mol Cell Endocrinol 2008; 286:214-8. [PMID: 18276067 DOI: 10.1016/j.mce.2007.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 12/07/2007] [Accepted: 12/18/2007] [Indexed: 11/16/2022]
Abstract
Clinically "non-functioning" human pituitary adenomas (NFPA) constitute about 35% of pituitary adenomas. Somatostatin receptors (SSTR) expression in these adenomas has previously been described both in vitro and in vivo, without evidence for a correlation with tumor volume or the therapeutic efficacy of somatostatin analogs. This study was performed on 13 surgically removed pituitary macroadenomas, diagnosed before surgery as "non-functioning". In addition, 3 growth hormone (GH)-secreting adenomas served as controls. A specimen from each tumor was dispersed and digested to isolate and culture the tumor cells, and the in vitro effects of SSTR2 and SSTR5 selective analogs and Cortistatin (CST) (100nM) on cell viability were studied. The quantity of viable cells was estimated using the XTT method. RNA purification of tumor samples and subsequent RT-PCR studies for SSTR2 and SSTR5 expression were performed. Somatostatin analog with high affinity for SSTR2 reduced cell viability by 20-80% in 8 of 13 NFPAs studied, all expressing the SSTR2. The inhibitory effect on cell viability of SSTR5-selective analog was 15-80% in 10 of 13 NFPAs studied, all but three expressing the SSTR5. CST, however, effectively reduced cell viability in only 6 NFPAs. Cell viability was inhibited by all peptides studied in 2 out of 3 GH-secreting adenomas, expressing both receptors. The third adenoma responded to SSTR2 analog and expressed only SSTR2. These results suggest the involvement of SSTR2 and SSTR5 in the anti-proliferative effects of somatostatin; however, CST is less potent in reducing cell viability in these tumors.
Collapse
Affiliation(s)
- H Padova
- Institute of Endocrinology, Rabin Medical Center, Petah-Tiqva, Tel Aviv University, Sackler School of Medicine, Israel
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hofland LJ. Somatostatin and somatostatin receptors in Cushing's disease. Mol Cell Endocrinol 2008; 286:199-205. [PMID: 18221833 DOI: 10.1016/j.mce.2007.10.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 10/04/2007] [Accepted: 10/24/2007] [Indexed: 11/15/2022]
Abstract
Cushing's disease is caused by an ACTH secreting pituitary adenoma. Surgery is the treatment of choice and cure rates between 60 and 90% are reported. For patients in which surgery fails, effective medical treatment options are needed. Somatostatin (SS) receptors (sst) are expressed on normal and tumoral corticotroph cells. However, the role of somatostatin and in particular the current clinically available sst(2)-preferring SS analogs in the regulation of normal ACTH secretion, as well as in lowering ACTH and cortisol hypersecretion in patients with Cushing's disease, has been shown to be limited. Recent studies have provided renewed insights into the expression of sst subtypes, as well as into the functional role of SS-analogs in the regulation of ACTH secretion by corticotroph tumors. Sst(2) and sst(5) seem the predominantly expressed sst in corticotroph adenoma cells and targeting both these receptors with a new generation of multiligand SS analogs showed promising effects in terms of lowering ACTH release and urinary free cortisol (UFC) levels in patients with Cushing's disease. In this review an overview of the current insights into the role of SS and sst in the regulation of normal and pathological ACTH secretion is provided.
Collapse
Affiliation(s)
- Leo J Hofland
- Department of Internal Medicine, Division Endocrinology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Prodam F, Benso A, Gramaglia E, Lucatello B, Riganti F, van der Lely AJ, Deghenghi R, Muccioli G, Ghigo E, Broglio F. Cortistatin-8, a synthetic cortistatin-derived ghrelin receptor ligand, does not modify the endocrine responses to acylated ghrelin or hexarelin in humans. Neuropeptides 2008; 42:89-93. [PMID: 18061663 DOI: 10.1016/j.npep.2007.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 08/30/2007] [Accepted: 09/21/2007] [Indexed: 10/22/2022]
Abstract
Cortistatin (CST), a neuropeptide with high structural homology with somatostatin (SST), binds all SST receptor (SST-R) subtypes but, unlike SST, also shows high binding affinity to ghrelin receptor (GHS-R1a). CST exerts the same endocrine activities of SST in humans, suggesting that the activation of the SST-R might mask the potential interaction with ghrelin system. CST-8, a synthetic CST-analogue devoid of any binding affinity to SST-R but capable to bind the GHS-R1a, has been reported able to exert antagonistic effects on ghrelin actions either in vitro or in vivo in animals. We studied the effects of CST-8 (2.0 microg/kg i.v. as a bolus or 2.0 microg/kg/h i.v. as infusion) on both spontaneous and ghrelin- or hexarelin- (1.0 microg/kg i.v. as bolus) stimulated GH, PRL, ACTH and cortisol secretion in 6 normal volunteers. During saline, no change occurred in GH and PRL levels while a spontaneous ACTH and cortisol decrease was observed. As expected, both ghrelin and hexarelin stimulated GH, PRL, ACTH and cortisol secretion (p<0.05). CST-8, administered either as bolus or as continuous infusion, did not modify both spontaneous and ghrelin- or hexarelin-stimulated GH, PRL, ACTH and cortisol secretion. In conclusion, CST-8 seems devoid of any modulatory action on either spontaneous or ghrelin-stimulated somatotroph, lactotroph and corticotroph secretion in humans in vivo. These negative results do not per se exclude that, even at these doses, CST-8 might have some neuroendocrine effects after prolonged treatment or that, at higher doses, may be able to effectively antagonize ghrelin action in humans. However, these data strongly suggest that CST-8 is not a promising candidate as GHS-R1a antagonist for human studies to explore the functional interaction between ghrelin and cortistatin systems.
Collapse
Affiliation(s)
- F Prodam
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Broglio F, Papotti M, Muccioli G, Ghigo E. Brain-gut communication: cortistatin, somatostatin and ghrelin. Trends Endocrinol Metab 2007; 18:246-51. [PMID: 17632010 DOI: 10.1016/j.tem.2007.06.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 05/29/2007] [Accepted: 06/20/2007] [Indexed: 12/11/2022]
Abstract
Although cortistatin (CST) shares great structural homology with somatostatin (SST) and binds to all SST receptor subtypes with similar affinity, these neurohormones have divergent biological roles, as evidenced by their different patterns of tissue expression and biological actions. Moreover, CST, but not SST, can bind to the proadrenomedullin N-terminal peptide (PAMP) receptor MrgX2 and type 1a growth hormone secretagogue (GHS) receptor (GHSR-1a), also known as the 'ghrelin' receptor. These findings suggest that CST-specific actions could be mediated by the GHSR-1a and CST might represent a link between the ghrelin and the SST systems. Here, we review the data leading to this working hypothesis and discuss the in vitro, in vivo and clinical implications of potential SST-receptor-independent, GHSR-1a-mediated neuroendocrine and metabolic effects of CST.
Collapse
Affiliation(s)
- Fabio Broglio
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Turin, corso Dogliotti 14, 10126 Turin, Italy.
| | | | | | | |
Collapse
|
29
|
Xidakis C, Mastrodimou N, Notas G, Renieri E, Kolios G, Kouroumalis E, Thermos K. RT-PCR and immunocytochemistry studies support the presence of somatostatin, cortistatin and somatostatin receptor subtypes in rat Kupffer cells. ACTA ACUST UNITED AC 2007; 143:76-82. [PMID: 17481746 DOI: 10.1016/j.regpep.2007.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Revised: 03/14/2007] [Accepted: 03/18/2007] [Indexed: 01/31/2023]
Abstract
The present study investigated the presence of somatostatin receptor subtypes (ssts) and the endogenous peptides somatostatin and cortistatin in rat Kupffer cells, since modulation of these cells by somatostatin may be important for the beneficial effect of somatostatin analogues in a selected group of hepatocellular carcinoma patients. Kupffer cells were isolated from rat liver in agreement with national and EU guidelines. RT-PCR was employed to assess the expression of somatostatin, cortistatin and ssts in Kupffer cells. Western blot analysis and immunocytochemistry were employed to assess the expression and the localization of the receptors, respectively. Quiescent Kupffer cells were found to express sst(1-4) mRNA, while immunocytochemical studies supported the presence of only the sst(3) and sst(4) receptors, which were found to be internalized. However, sst1 and sst(2A) receptors were detected by western blotting. RT-PCR and RIA measurements support the presence of both somatostatin and cortistatin. Stimulation of the cells with LPS activated the expression of the sst(2), sst(3) and sst(4) receptors. The present data provide evidence to support the presence of ssts and the endogenous neuropeptides somatostatin and CST in rat Kupffer cells. Both peptides may act in an autocrine manner to regulate sst receptor distribution. Studies are in progress in order to further characterize the role of ssts in Kupffer cells and in hepatic therapeutics.
Collapse
Affiliation(s)
- C Xidakis
- Laboratory of Pharmacology, Department Basic Sciences, University of Crete, Faculty of Medicine, Heraklion, Crete, Greece
| | | | | | | | | | | | | |
Collapse
|
30
|
Canosa LF, Chang JP, Peter RE. Neuroendocrine control of growth hormone in fish. Gen Comp Endocrinol 2007; 151:1-26. [PMID: 17286975 DOI: 10.1016/j.ygcen.2006.12.010] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 12/12/2006] [Accepted: 12/22/2006] [Indexed: 10/23/2022]
Abstract
The biological actions of growth hormone (GH) are pleiotropic, including growth promotion, energy mobilization, gonadal development, appetite, and social behavior. Accordingly, the regulatory network for GH is complex and includes many endocrine and environmental factors. In fish, the neuroendocrine control of GH is multifactorial with multiple inhibitors and stimulators of pituitary GH secretion. In fish, GH release is under a tonic negative control exerted mainly by somatostatin. Sex steroid hormones and nutritional status influence the level of brain expression and effectiveness of some of these GH neuroendocrine regulatory factors, suggesting that their relative importance differs under different physiological conditions. At the pituitary level, some, if not all, somatotropes can respond to multiple regulators. Therefore, ligand- and function-specificity, as well as the integrative responses to multiple signals must be achieved at the level of signal transduction mechanisms. Results from investigations on a limited number of stimulatory and inhibitory GH-release regulators indicate that activation of different but convergent intracellular pathways and the utilization of specific intracellular Ca(2+) stores are some of the strategies utilized. However, more work remains to be done in order to better understand the integrative mechanisms of signal transduction at the somatotrope level and the relevance of various GH regulators in different physiological circumstances.
Collapse
Affiliation(s)
- Luis Fabián Canosa
- Department of Biological Sciences, University of Alberta, Edmonton, Alta., Canada T6G 2E9
| | | | | |
Collapse
|
31
|
Abstract
Insomnia and hypersomnia are frequent sleep disorders, and they are most often treated pharmacologically with hypnotics and wake-promoting compounds. These compounds act on classical neurotransmitter systems, such as benzodiazepines on GABA-A receptors, and amfetamine-like stimulants on monoaminergic terminals to modulate neurotransmission. In addition, acetylcholine, amino acids, lipids and proteins (cytokines) and peptides, are known to significantly modulate sleep and are, therefore, possibly involved in the pathophysiology of some sleep disorders. Due to the recent developments of molecular biological techniques, many neuropeptides have been newly identified, and some are found to significantly modulate sleep. It was also discovered that the impairment of the hypocretin/orexin neurotransmission (a recently isolated hypothalamic neuropeptide system) is the major pathophysiology of narcolepsy, and hypocretin replacement therapy is anticipated to treat the disease in humans. In this article, the authors briefly review the history of neuropeptide research, followed by the sleep modulatory effects of various neuropeptides. Finally, general strategies for the pharmacological therapeutics targeting the peptidergic systems for sleep disorders are discussed.
Collapse
Affiliation(s)
- Seiji Nishino
- Stanford University School of Medicine, Department of Psychiatry and Behavioural Sciences, Sleep and Circadian Neurobiology Laboratory and Center for Narcolepsy Research, Palo Alto, CA 94304-5489, USA.
| | | |
Collapse
|
32
|
Abstract
The neuropeptide somatostatin (SST) is expressed in a discrete population of interneurons in the dentate gyrus. These interneurons have their soma in the hilus and project to the outer molecular layer onto dendrites of dentate granule cells, adjacent to perforant path input. SST-containing interneurons are very sensitive to excitotoxicty, and thus are vulnerable to a variety of neurological diseases and insults, including epilepsy, Alzheimer's disease, traumatic brain injury, and ischemia. The SST gene contains a prototypical cyclic AMP response element (CRE) site. Such a regulatory site confers activity-dependence to the gene, such that it is turned on when neuronal activity is high. Thus SST expression is increased by pathological conditions such as seizures and by natural stimulation such as environmental enrichment. SST may play an important role in cognition by modulating the response of neurons to synaptic input. In the dentate, SST and the related peptide cortistatin (CST) reduce the likelihood of generating long-term potentiation, a cellular process involved in learning and memory. Thus these neuropeptides would increase the threshold of input required for acquisition of new memories, increasing "signal to noise" to filter out irrelevant environmental cues. The major mechanism through which SST inhibits LTP is likely through inhibition of voltage-gated Ca(2+) channels on dentate granule cell dendrites. Transgenic overexpression of CST in the dentate leads to profound deficits in spatial learning and memory, validating its role in cognitive processing. A reduction of synaptic potentiation by SST and CST in dentate may also contribute to the well-characterized antiepileptic properties of these neuropeptides. Thus SST and CST are important neuromodulators in the dentate gyrus, and disruption of this signaling system may have major impact on hippocampal function.
Collapse
Affiliation(s)
- Melanie K Tallent
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15 St., Philadelphia, PA 19102, USA.
| |
Collapse
|
33
|
Abstract
Cortistatin (CST) is a recently discovered neuropeptide from the somatostatin gene family, named after its predominantly cortical expression and ability to depress cortical activity. CST shows many remarkable structural and functional similarities to its related neuropeptide somatostatin, or somatotropin release-inhibiting factor. However, the many physiological differences between CST and somatostatin are just as remarkable as the similarities. CST-29 has recently been shown to prevent inflammation in rodent models for human diseases, raising novel therapeutic properties to this neuropeptide. In this review, the authors address a new possible role for CST in the immune system and evaluate the possible therapeutic use of CST to treat disorders associated with inflammation.
Collapse
Affiliation(s)
- Alicia Rubio
- Universidad Autónoma de Madrid, Centro de Biología Molecular, Madrid, Spain
| | | | | |
Collapse
|
34
|
Tostivint H, Joly L, Lihrmann I, Parmentier C, Lebon A, Morisson M, Calas A, Ekker M, Vaudry H. Comparative genomics provides evidence for close evolutionary relationships between the urotensin II and somatostatin gene families. Proc Natl Acad Sci U S A 2006; 103:2237-42. [PMID: 16467151 PMCID: PMC1413727 DOI: 10.1073/pnas.0510700103] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although urotensin II (UII) and somatostatin 1 (SS1) exhibit some structural similarities, their precursors do not show any appreciable sequence identity and, thus, it is widely accepted that the UII and SS1 genes do not derive from a common ancestral gene. The recent characterization of novel isoforms of these two peptides, namely urotensin II-related peptide (URP) and somatostatin 2 (SS2)/cortistatin (CST), provides new opportunity to revisit the phylogenetic relationships of UII and SS1 using a comparative genomics approach. In the present study, by radiation hybrid mapping and in silico sequence analysis, we have determined the chromosomal localization of the genes encoding UII- and somatostatin-related peptides in several vertebrate species, including human, chicken, and zebrafish. In most of the species investigated, the UII and URP genes are closely linked to the SS2/CST and SS1 genes, respectively. We also found that the UII-SS2/CST locus and the URP/SS1 locus are paralogous. Taken together, these data indicate that the UII and URP genes, on the one hand, and the SS1 and SS2/CST genes, on the other hand, arose through a segmental duplication of two ancestral genes that were already physically linked to each other. Our results also suggest that these two genes arose themselves through a tandem duplication of a single ancestral gene. It thus appears that the genes encoding UII- and somatostatin-related peptides belong to the same superfamily.
Collapse
Affiliation(s)
- Hervé Tostivint
- *Institut National de la Santé et de la Recherche Médicale Unité 413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, 76821 Mont-Saint-Aignan, France
| | - Lucille Joly
- Center for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Isabelle Lihrmann
- *Institut National de la Santé et de la Recherche Médicale Unité 413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, 76821 Mont-Saint-Aignan, France
| | - Caroline Parmentier
- Laboratoire de Neurobiologie des Signaux Intercellulaires, Centre National de la Recherche Scientifique Unité Mixte Recherche 7101, Université Pierre et Marie Curie, 75252 Paris, France; and
| | - Alexis Lebon
- *Institut National de la Santé et de la Recherche Médicale Unité 413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, 76821 Mont-Saint-Aignan, France
| | - Mireille Morisson
- Laboratoire de Génétique Cellulaire, Institut National de la Recherche Agronomique, 31326 Castanet-Tolosan, France
| | - André Calas
- Laboratoire de Neurobiologie des Signaux Intercellulaires, Centre National de la Recherche Scientifique Unité Mixte Recherche 7101, Université Pierre et Marie Curie, 75252 Paris, France; and
| | - Marc Ekker
- Center for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Hubert Vaudry
- *Institut National de la Santé et de la Recherche Médicale Unité 413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, 76821 Mont-Saint-Aignan, France
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
35
|
Tostivint H, Joly L, Lihrmann I, Conlon JM, Ekker M, Vaudry H. Linkage Mapping of the [Pro2]Somatostatin-14 Gene in Zebrafish: Evolutionary Perspectives. Ann N Y Acad Sci 2006; 1040:486-9. [PMID: 15891097 DOI: 10.1196/annals.1327.098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Radiation hybrid mapping assigned the zebrafish [Pro(2)]somatostatin-14 (also termed somatostatin 2; SS2) gene to linkage group 23 of the zebrafish genome, close to the marker nadl1.2. Comparative genomic analysis revealed conserved syntenies of the SS2 gene locus with part of the human 1p36 region, where the cortistatin gene is located. This observation strongly suggests that the SS2 gene in nonmammalian species and the cortistatin gene in mammals are orthologous.
Collapse
Affiliation(s)
- Hervé Tostivint
- European Institute for Peptide Research (IFRMP 23), Laboratory of Cellular and Molecular Neuroendocrinology, INSERM U-413, University of Rouen, Mont-Saint-Aignan, France.
| | | | | | | | | | | |
Collapse
|
36
|
Vallarino M, Bruzzone F, Mathieu M, Chartrel N, Vieau D, Ciarlo M, Fournier A, Vaudry H. Ontogeny of the somatostatin variant [Pro2,Met13]somatostatin-14 in the brain, pituitary, and sensory organs of the frogRana esculenta. J Comp Neurol 2006; 497:717-33. [PMID: 16786560 DOI: 10.1002/cne.20986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Two forms of somatostatin are expressed in the frog brain, i.e., somatostatin-14 (SS1) and the [Pro(2), Met(13)]somatostatin-14 variant (SS2). We have previously described the ontogeny of SS1-immunoreactive cells in the brain of Rana esculenta. In the present study, we have investigated the distribution of prepro-SS2 (PSS2)-expressing neurons in the brain of the same species during development by using antibodies directed against the N-flanking region of SS2 (PSS2(54-66)). Immunoreactive perikarya first appeared in the ventral hypothalamus at stages IV-VII. Subsequently, positive neurons were seen in the nucleus of the diagonal band of Broca, the anterior preoptic area, the posterior tuberculum (stages VIII-XII), as well as the dorsal (stages XIII-XV) and medial (stages XIX-XX) periventricular preoptic nucleus. At metamorphic climax and in newly metamorphosed frogs, positive perikarya were found in the striatum and in the interpeduncular nucleus. PSS2(54-66)-immunoreactive fibers were already widely distributed during the first stages of development, indicating that SS2 may act as a neuromodulator and/or neurotransmitter during ontogeny. The presence of PSS2(54-66)-positive nerve fibers in olfactory structures suggests that, in tadpoles, SS2 may be involved in the processing of olfactory information. The occurrence of PSS2(54-66)-like immunoreactivity in taste buds, and in the olfactory and vomeronasal organs indicates that SS2 may mediate the unconditioned and reinforcing properties of natural chemicals. Finally, the intenseexpression of PSS2(54-66)-like immunoreactivity in melanotrope cells of the pituitary suggests that SS2 may diffuse toward the pars distalis to regulate the activity of adenohypophysial cells during tadpole development.
Collapse
Affiliation(s)
- Mauro Vallarino
- Department of Experimental Biology, DIBISAA, University of Genova, 16132 Genova, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Otal R, Burgaya F, Frisén J, Soriano E, Martínez A. Ephrin-A5 modulates the topographic mapping and connectivity of commissural axons in murine hippocampus. Neuroscience 2006; 141:109-21. [PMID: 16690216 DOI: 10.1016/j.neuroscience.2006.03.052] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 02/17/2006] [Accepted: 03/20/2006] [Indexed: 10/24/2022]
Abstract
Entorhinal and commissural/associational projections show a non-overlapping distribution in the hippocampus proper and the dentate gyrus. The expression of Ephrins and their Eph receptors in the developing hippocampus indicates that this family of axonal guidance molecules may modulate the formation of these connections. Here we focused on the role of the ephrin-A5 ligand in the development of the main hippocampal afferents. In situ hybridization showed that ephrin-A5 mRNA was detected mainly in the principal cells of the hippocampus proper and in the dentate gyrus throughout postnatal development. Immunocytochemical analyses revealed prominent expression of the EphA3 receptor, a putative receptor for ephrin-A5, in the main cells and the neuropil of the developing hippocampus. Tracing experiments in ephrin-A5(-/-) mice showed that commissural projections were transiently altered in the hippocampus proper at P5, but they were mistargeted throughout the postnatal development in the dentate gyrus. Immunocytochemistry with anti-calbindin antibodies revealed that the dentate mossy fiber projection was not altered in ephrin-A5(-/-) mice. Electron microscopy studies showed alterations in the density of synapses and spines in commissural/associational layers, but not in entorhinal layers, and in the mossy fibers in these animals. Taken together, these findings indicate that ephrin-A5 signaling is involved in the formation and maturation of synapses in the hippocampus.
Collapse
Affiliation(s)
- R Otal
- Department of Cell Biology, University of Barcelona and Institut de Recerca Biomèdica, Parc Científic de Barcelona, E-08028 Barcelona, Spain
| | | | | | | | | |
Collapse
|
38
|
Thermos K, Bagnoli P, Epelbaum J, Hoyer D. The somatostatin sst1 receptor: an autoreceptor for somatostatin in brain and retina? Pharmacol Ther 2005; 110:455-64. [PMID: 16274747 DOI: 10.1016/j.pharmthera.2005.09.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 09/20/2005] [Indexed: 11/15/2022]
Abstract
The sst1 receptor was the first of the 5 somatostatin receptors to be cloned by homology with the glucagon receptor 13 years ago. It is a 7-transmembrane domain G-protein-coupled receptor that is negatively coupled to adenylyl cyclase, but can also trigger other transduction pathways. The distribution of sst1 mRNA, immunolabeling, and radioligand binding are not entirely overlapping, but the recent availability of knockout (KO) mice and a (still limited) number of selective agonists/antagonists has increased our knowledge about this receptor. These new tools have helped to reveal a role for the sst1 receptor in hippocampal, hypothalamic, basal ganglia, and retinal functions. In at least the latter 3 structures, the sst1 receptor appears to act as an inhibitory autoreceptor located on somatostatin neurons, whereas in the hippocampus such a role is still based on circumstantial evidence.
Collapse
Affiliation(s)
- Kyriaki Thermos
- Laboratory of Pharmacology, Department of Basic Sciences, School of Medicine, University of Crete, GR-71110 Heraklion, Crete, Greece
| | | | | | | |
Collapse
|
39
|
Tallent MK, Fabre V, Qiu C, Calbet M, Lamp T, Baratta MV, Suzuki C, Levy CL, Siggins GR, Henriksen SJ, Criado JR, Roberts A, de Lecea L. Cortistatin overexpression in transgenic mice produces deficits in synaptic plasticity and learning. Mol Cell Neurosci 2005; 30:465-75. [PMID: 16182561 DOI: 10.1016/j.mcn.2005.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Revised: 07/18/2005] [Accepted: 08/17/2005] [Indexed: 11/15/2022] Open
Abstract
Cortistatin-14 (CST) is a neuropeptide expressed in cortical and hippocampal interneurons that shares 11 of 14 residues with somatostatin. In contrast to somatostatin, infusion of CST decreases locomotor activity and selectively enhances slow wave sleep. Here, we show that transgenic mice that overexpress cortistatin under the control of neuron-specific enolase promoter do not express long-term potentiation in the dentate gyrus. This blockade of dentate LTP correlates with profound impairment of hippocampal-dependent spatial learning. Exogenously applied CST to slices of wild-type mice also blocked induction of LTP in the dentate gyrus. Our findings implicate cortistatin in the modulation of synaptic plasticity and cognitive function. Thus, increases in hippocampal cortistatin expression during aging could have an impact on age-related cognitive deficits.
Collapse
Affiliation(s)
- Melanie K Tallent
- Department of Pharmacology and Physiology, Drexel University, University College of Medicine, Philadelphia, PA 19102, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Li M, Yan S, Fisher WE, Chen C, Yao Q. New Roles of a Neuropeptide Cortistatin in the Immune System and Cancer. World J Surg 2005; 29:354-6. [PMID: 15696397 DOI: 10.1007/s00268-004-7811-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Cortistatin (CST) is a neuropeptide that strongly resembles somatostatin (SS) structurally and functionally. CST binds to all five SS receptors (SSTR1-SSTR5) with high affinity and exerts its function mainly through SSTRs. Despite many similar functions between these two neuropeptides, they are products of different genes. Recently, some distinct functions and receptor usage of CST have been reported. Some of the interesting functions of CST were not found with SS. Therefore CST could have potential new roles in an ex-neuronal system that regulates immune responses as well as other cellular functions in the body. In this review, we discuss the new functions of CST in the immune system, cancer pathogenesis, and possible CST-specific receptors.
Collapse
Affiliation(s)
- Min Li
- Molecular Surgeon Research Center, Michael E. DeBakey Department of Surgery, Baylor College of Medicine and the Methodist Hospital, 6550 Fannin Street, Houston, Texas 77030, USA.
| | | | | | | | | |
Collapse
|
41
|
Spier AD, Fabre V, de Lecea L. Cortistatin radioligand binding in wild-type and somatostatin receptor-deficient mouse brain. ACTA ACUST UNITED AC 2005; 124:179-86. [PMID: 15544857 DOI: 10.1016/j.regpep.2004.07.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Revised: 07/10/2004] [Accepted: 07/12/2004] [Indexed: 10/26/2022]
Abstract
Cortistatin-14 (CST-14) is a recently discovered member of the somatostatin family of neuropeptides. It shares 11 of its 14 amino acids with somatostatin-14 (SRIF-14). In the present study, binding sites for cortistatin-14 in the mouse brain were examined and compared to those for somatostatin using iodinated cortistatin-14 and iodinated somatostatin-14. By in vitro receptor autoradiography, high densities of cortistatin-14 and somatostatin-14 specific binding sites were detected in the cortex, hippocampal formation, basolateral amygdala and medial habenula. Unlabeled 100 nM cortistatin-14 inhibited iodinated somatostatin-14 binding in the hippocampus, but not in the cortex or amygdaloid nuclei. In somatostatin receptor subtype-2 knock-out (KO) mice, autoradiographic iodinated somatostatin-14 binding was observed in the hippocampus and habenula but was removed in the cortex and amygdaloid nuclei, specific iodinated cortistatin-14 binding sites were found in the hippocampus, habenula and throughout the cortex. We conclude that the somatostatin receptor subtype-2 is responsible for somatostatin binding in cortical and amygdaloid regions and that cortistatin predominantly interacts with the same receptors as somatostatin.
Collapse
Affiliation(s)
- Avron D Spier
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
42
|
Allia E, Tarabra E, Volante M, Cerrato M, Ghigo E, Muccioli G, Papotti M. Expression of cortistatin and MrgX2, a specific cortistatin receptor, in human neuroendocrine tissues and related tumours. J Pathol 2005; 207:336-45. [PMID: 16161007 DOI: 10.1002/path.1839] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cortistatin (CST), a novel hormone originally described in the rat, mouse, and human cerebral cortex, displays structural and functional similarities to somatostatin (SRIF). CST binds to all five somatostatin receptors and, differently from SRIF, also binds to MrgX2, which has recently been identified as its specific receptor. Little is known about the distribution of CST and MrgX2 in peripheral non-tumour and neoplastic tissues. The aim of the present study was therefore to determine by immunohistochemistry and mRNA analysis (RT-PCR) the distribution of CST and MrgX2 in 56 human non-tumour and 108 tumour tissues, with special reference to neuroendocrine tissue types. Despite the high level of CST mRNA expression in non-tumour and tumour (both neuroendocrine and non-neuroendocrine) tissues, the presence of immunoreactive CST was confirmed in a subset of gastroenteropancreatic, parathyroid, and pituitary non-tumour cells only, and showed a predominantly focal pattern in most neuroendocrine tumours. Co-localization experiments in the gastroenteropancreatic system demonstrated that the normal CST-producing cells are delta cells, while in the adenohypophysis no preferential co-localization of CST with any of the pituitary hormones was observed. MrgX2 mRNA was variably detected in the hypothalamus, pituitary, thyroid, lung, gastroenteropancreatic tract, testis, and ovary, and was negative in the cerebral cortex, parathyroid, and adrenal, as well as in a variety of tumour types. Conversely, immunolocalization of MrgX2 protein was restricted to neurohypophysis and testis, whilst all tumours analysed were negative. A possible explanation for the discrepancy between RT-PCR and immunohistochemistry is that MrgX2 protein was widely detected in blood vessels, scattered lymphocytes, and gastrointestinal ganglia in both normal and neoplastic samples. The findings demonstrate a selective distribution of CST in normal and neoplastic neuroendocrine tissues, suggesting that CST might have a broader functional role than previously assumed, whereas possible autocrine/paracrine actions via its recently described specific receptor MrgX2 are restricted to selected tissues.
Collapse
Affiliation(s)
- Elena Allia
- Department of Biomedical Sciences and Oncology, University of Turin, Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
43
|
Winsky-Sommerer R, Spier AD, Fabre V, de Lecea L, Criado JR. Overexpression of the human β-amyloid precursor protein downregulates cortistatin mRNA in PDAPP mice. Brain Res 2004; 1023:157-62. [PMID: 15364032 DOI: 10.1016/j.brainres.2004.04.082] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2004] [Indexed: 11/29/2022]
Abstract
We measured preprocortistatin mRNA expression in young and aged transgenic (Tg) mice overexpressing the human beta-amyloid precursor protein (hbetaAPP) under the platelet-derived growth factor-beta promoter. Our findings suggest that the significant increase in hippocampal cortistatin mRNA expression during normal aging is significantly attenuated in Tg mice at an age known to exhibit beta-amyloid protein (Abeta) deposition. These deficits in cortistatin expression may play a role in the deficits in hippocampal-dependent spatial learning and sleep/wake states previously demonstrated in aged Tg mice.
Collapse
|
44
|
Gottero C, Prodam F, Destefanis S, Benso A, Gauna C, Me E, Filtri L, Riganti F, Van Der Lely AJ, Ghigo E, Broglio F. Cortistatin-17 and -14 exert the same endocrine activities as somatostatin in humans. Growth Horm IGF Res 2004; 14:382-387. [PMID: 15336231 DOI: 10.1016/j.ghir.2004.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2004] [Revised: 04/27/2004] [Accepted: 04/28/2004] [Indexed: 11/21/2022]
Abstract
Cortistatin (CST) is a neuropeptide, which binds with high affinity all somatostatin (SS) receptor subtypes and shows high structural homology with SS itself. A receptor specific for CST only, i.e., not recognized by SS, has been recently described in agreement with data reporting that not all CST actions are shared by SS. Interestingly, CST but not SS also binds ghrelin receptor (GHS-R1a) in vitro, suggesting a potential interplay between CST and ghrelin system. The aim of this study was to investigate in humans the endocrine and metabolic activities of human CST-17 in comparison with rat CST-14 that has previously been shown to exert the same endocrine actions of SS in healthy volunteers. To this aim, in six healthy male volunteers (age [median, 3rd-97th centiles]: 28.5; 23.6-34.3 years; Body Mass Index: 23.5; 21.0-25.1 kg/m(2)), we studied the effects of human CST-17 (2.0 microg/kg/h iv over 120 min), rat CST-14 (2.0 microg/kg/h iv over 120 min) and SS-14 (2.0 microg/kg/h iv over 120 min) on: (a) spontaneous GH, ACTH, PRL, cortisol, insulin and glucose levels; (b) the GH responses to GHRH (1.0 microg/kg iv at 0 min); (c) the GH, PRL, ACTH, cortisol, insulin and glucose responses to ghrelin (1.0 microg/kg iv at 0 min). CST-17 inhibited (p < 0.01) basal GH secretion to the same extent of CST-14 and SS-14. Spontaneous PRL, ACTH and cortisol secretion were not significantly modified by CST-17, CST-14 or SS-14. CST-17 as well as CST-14 and SS-14 also inhibited (p < 0.05) spontaneous insulin secretion to a similar extent. None of these peptides modified glucose levels. The GH response to GHRH was inhibited to the same extent by CST-17 (p < 0.01), CST-14 (p < 0.01) and SS-14 (p < 0.05 ). The ghrelin-induced GH response was higher than that elicited by GHRH (p < 0.01) and inhibited by CST-17 (p < 0.05) as well as by CST-14 (p < 0.05) and SS-14 (p < 0.01). The PRL, ACTH and cortisol responses to ghrelin were unaffected by CST-17, CST-14 or SS-14. On the other hand, the inhibitory effect of ghrelin on insulin levels was abolished by CST-17, CST-14 or SS-14 (p < 0.05) that, in turn, did not modify the ghrelin-induced increase in glucose levels. In conclusion, this study demonstrates that human CST-17 and rat CST-14 exert the same endocrine activities of SS in humans. The endocrine actions of human and rat CST therefore are likely to reflect activation of classical SS receptors.
Collapse
Affiliation(s)
- C Gottero
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Turin, c.so Dogliotti 14, 10126 Turin, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Canosa LF, Cerdá-Reverter JM, Peter RE. Brain mapping of three somatostatin encoding genes in the goldfish. J Comp Neurol 2004; 474:43-57. [PMID: 15156578 DOI: 10.1002/cne.20097] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In the present study the brain distribution of three somatostatin (SRIF)-encoding genes, PSS-I, PSS-II, and PSS-III, was analyzed by in situ hybridization (ISH) in the goldfish. The PSS-I mRNA showed the widest distribution throughout the brain, whereas PSS-II transcripts were restricted to some hypothalamic nuclei. On the other hand, PSS-III presents an intermediate distribution pattern. All SRIF encoding genes are expressed in hypophysiotropic nuclei supporting the idea that, in addition to SRIF-14, [Pro(2)] SRIF-14, and gSRIF-28 have pituitary-controlling functions. Moreover, each of the genes is expressed in nuclei directly associated with feeding behavior, suggesting a role for SRIF peptides in the central control of food intake and energy balance. Alternatively, they might have a role in processing sensory information related with feeding behavior, since PSS genes are expressed in the main gustatory, olfactory, and visual centers, which project to the hypothalamic feeding center in teleost fish.
Collapse
Affiliation(s)
- Luis Fabián Canosa
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | | | | |
Collapse
|
46
|
van der Lely AJ, Tschöp M, Heiman ML, Ghigo E. Biological, physiological, pathophysiological, and pharmacological aspects of ghrelin. Endocr Rev 2004; 25:426-57. [PMID: 15180951 DOI: 10.1210/er.2002-0029] [Citation(s) in RCA: 802] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ghrelin is a peptide predominantly produced by the stomach. Ghrelin displays strong GH-releasing activity. This activity is mediated by the activation of the so-called GH secretagogue receptor type 1a. This receptor had been shown to be specific for a family of synthetic, peptidyl and nonpeptidyl GH secretagogues. Apart from a potent GH-releasing action, ghrelin has other activities including stimulation of lactotroph and corticotroph function, influence on the pituitary gonadal axis, stimulation of appetite, control of energy balance, influence on sleep and behavior, control of gastric motility and acid secretion, and influence on pancreatic exocrine and endocrine function as well as on glucose metabolism. Cardiovascular actions and modulation of proliferation of neoplastic cells, as well as of the immune system, are other actions of ghrelin. Therefore, we consider ghrelin a gastrointestinal peptide contributing to the regulation of diverse functions of the gut-brain axis. So, there is indeed a possibility that ghrelin analogs, acting as either agonists or antagonists, might have clinical impact.
Collapse
Affiliation(s)
- Aart J van der Lely
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
47
|
Dalm VASH, Van Hagen PM, de Krijger RR, Kros JM, Van Koetsveld PM, Van Der Lely AJ, Lamberts SWJ, Hofland LJ. Distribution pattern of somatostatin and cortistatin mRNA in human central and peripheral tissues. Clin Endocrinol (Oxf) 2004; 60:625-9. [PMID: 15104567 DOI: 10.1111/j.1365-2265.2004.02024.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Somatostatin receptors (sst) and their endogenous ligand, somatostatin (SS), are widely expressed throughout the human body. Recently, the cDNA of a novel SS-like peptide, named cortistatin (CST), has been cloned. This CST was found to be expressed in more restricted areas, like brain cortex, testes, kidney, stomach and leucocytes. Further studies demonstrated a selective expression of CST in tissues and cells of the human immune system, while SS was not expressed. OBJECTIVE In the present study we investigated the expression pattern of both SS mRNA and CST mRNA in various human central and peripheral tissues by quantitative RT-PCR (Q-PCR), in order to evaluate whether CST is more widely expressed in man than described to date and to investigate the relationship between SS and CST expression levels in various tissues. Previously, two different CST mRNA isoforms have been described. Therefore, we investigated the expression of both isoforms by RT-PCR in the different tissues as well. RESULTS We demonstrate for the first time that CST mRNA is widely expressed in the human body. Interestingly, a selective expression of CST mRNA and not SS mRNA was only found in isolated cells of the human immune system, whereas different tissues expressed both SS and CST mRNA. CONCLUSION CST may have a broader functional role than previously assumed.
Collapse
Affiliation(s)
- Virgil A S H Dalm
- Department of Internal Medicine, Josephine Nefkens Institute, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Notas G, Kolios G, Mastrodimou N, Kampa M, Vasilaki A, Xidakis C, Castanas E, Thermos K, Kouroumalis E. Cortistatin production by HepG2 human hepatocellular carcinoma cell line and distribution of somatostatin receptors. J Hepatol 2004; 40:792-8. [PMID: 15094227 DOI: 10.1016/j.jhep.2004.01.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2003] [Revised: 12/07/2003] [Accepted: 01/08/2004] [Indexed: 12/19/2022]
Abstract
BACKGROUND/AIMS Recently, trials of octreotide have shown a significant survival benefit in the treatment of advanced hepatocellular carcinoma but new data are controversial. We, therefore, examined the production of somatostatin and cortistatin, the expression and distribution of somatostatin receptors (sst) in HepG2 human hepatocellular carcinoma cells, and the possible antiproliferative effect of octreotide on these cells. METHODS Radioimmunoassay and RT-PCR studies were performed for the detection of somatostatin and cortistatin. RT-PCR, radioligand binding and immunocytochemistry assays were employed for the detection of the ssts. Growth and viability of cells were measured by the tetrazolium salt assay. RESULTS HepG2 cells were found to express sst(2), sst(3) and sst(5) receptors. Immunocytochemistry revealed a mainly intracellular distribution of all ssts with unique patterns for each of them. Membrane binding sites for somatostatin were mainly of the sst(3) (39+/-8%) and sst(5) (59+/-5%) types, while only minor sst(2) binding could be detected (5+/-12%). Octreotide was found to inhibit the proliferation of HepG2 cells (IC(50) 1.25 x 10(-9)M) via protein tyrosine phosphatases. HepG2 cells produced cortistatin while somatostatin expression was not detected. CONCLUSIONS In conclusion, HepG2 cells express cortistatin, which regulates somatostatin receptors. Cell proliferation was reduced by octreotide via a protein tyrosine phosphatase dependent mechanism.
Collapse
MESH Headings
- Antineoplastic Agents, Hormonal/pharmacology
- Base Sequence
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- DNA/genetics
- Gene Expression
- Humans
- Immunohistochemistry
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Neuropeptides/biosynthesis
- Neuropeptides/genetics
- Octreotide/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Receptors, Somatostatin/genetics
- Receptors, Somatostatin/metabolism
- Somatostatin/biosynthesis
- Somatostatin/genetics
Collapse
Affiliation(s)
- George Notas
- Laboratory of Gastroenterology and Hepatology, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Møller LN, Stidsen CE, Hartmann B, Holst JJ. Somatostatin receptors. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2003; 1616:1-84. [PMID: 14507421 DOI: 10.1016/s0005-2736(03)00235-9] [Citation(s) in RCA: 255] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In 1972, Brazeau et al. isolated somatostatin (somatotropin release-inhibiting factor, SRIF), a cyclic polypeptide with two biologically active isoforms (SRIF-14 and SRIF-28). This event prompted the successful quest for SRIF receptors. Then, nearly a quarter of a century later, it was announced that a neuropeptide, to be named cortistatin (CST), had been cloned, bearing strong resemblance to SRIF. Evidence of special CST receptors never emerged, however. CST rather competed with both SRIF isoforms for specific receptor binding. And binding to the known subtypes with affinities in the nanomolar range, it has therefore been acknowledged to be a third endogenous ligand at SRIF receptors. This review goes through mechanisms of signal transduction, pharmacology, and anatomical distribution of SRIF receptors. Structurally, SRIF receptors belong to the superfamily of G protein-coupled (GPC) receptors, sharing the characteristic seven-transmembrane-segment (STMS) topography. Years of intensive research have resulted in cloning of five receptor subtypes (sst(1)-sst(5)), one of which is represented by two splice variants (sst(2A) and sst(2B)). The individual subtypes, functionally coupled to the effectors of signal transduction, are differentially expressed throughout the mammalian organism, with corresponding differences in physiological impact. It is evident that receptor function, from a physiological point of view, cannot simply be reduced to the accumulated operations of individual receptors. Far from being isolated functional units, receptors co-operate. The total receptor apparatus of individual cell types is composed of different-ligand receptors (e.g. SRIF and non-SRIF receptors) and co-expressed receptor subtypes (e.g. sst(2) and sst(5) receptors) in characteristic proportions. In other words, levels of individual receptor subtypes are highly cell-specific and vary with the co-expression of different-ligand receptors. However, the question is how to quantify the relative contributions of individual receptor subtypes to the integration of transduced signals, ultimately the result of collective receptor activity. The generation of knock-out (KO) mice, intended as a means to define the contributions made by individual receptor subtypes, necessarily marks but an approximation. Furthermore, we must now take into account the stunning complexity of receptor co-operation indicated by the observation of receptor homo- and heterodimerisation, let alone oligomerisation. Theoretically, this phenomenon adds a novel series of functional megareceptors/super-receptors, with varied pharmacological profiles, to the catalogue of monomeric receptor subtypes isolated and cloned in the past. SRIF analogues include both peptides and non-peptides, receptor agonists and antagonists. Relatively long half lives, as compared to those of the endogenous ligands, have been paramount from the outset. Motivated by theoretical puzzles or the shortcomings of present-day diagnostics and therapy, investigators have also aimed to produce subtype-selective analogues. Several have become available.
Collapse
Affiliation(s)
- Lars Neisig Møller
- Department of Medical Physiology, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | | | | |
Collapse
|
50
|
Robas N, Mead E, Fidock M. MrgX2 is a high potency cortistatin receptor expressed in dorsal root ganglion. J Biol Chem 2003; 278:44400-4. [PMID: 12915402 DOI: 10.1074/jbc.m302456200] [Citation(s) in RCA: 182] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MrgX2 is a recently identified orphan G-protein-coupled receptor whose ligand and physiological function were unknown. Here we describe cortistatin, a neuropeptide for which no specific receptor has been identified previously, as a high potency ligand at MrgX2. Cortistatin has several biological functions including roles in sleep regulation, locomotor activity, and cortical function. Using a "reverse pharmacology" approach, we have identified a number of additional cyclic peptide agonists for MrgX2, determined their rank order of potency, and demonstrated that this receptor has a pharmacological profile distinct from the other characterized members of the Mrg (Mas-related genes) family. In MrgX2-expressing cells, cortistatin-stimulated increases in intracellular Ca2+ but had no effect on basal or forskolin-stimulated cAMP levels, suggesting that this receptor is Gq-coupled. Immunohistochemical and quantitative PCR studies show MrgX2 to have a limited expression profile, both peripheral and within the central nervous system, with highest levels in dorsal root ganglion.
Collapse
Affiliation(s)
- Nicola Robas
- Department of Target Genomics, Pfizer Global Research and Development, Sandwich, Kent, CT13 N9J, United Kingdom
| | | | | |
Collapse
|