1
|
Gharib SA, Vemireddy R, Castillo JJ, Fountaine BS, Bammler TK, MacDonald JW, Hull-Meichle RL, Zraika S. Cystic fibrosis-related diabetes is associated with reduced islet protein expression of GLP-1 receptor and perturbation of cell-specific transcriptional programs. Sci Rep 2024; 14:25689. [PMID: 39463434 PMCID: PMC11514218 DOI: 10.1038/s41598-024-76722-1] [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: 08/16/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Insulin secretion is impaired in individuals with cystic fibrosis (CF), contributing to high rates of CF-related diabetes (CFRD) and substantially increasing disease burden. To develop improved therapies for CFRD, better knowledge of pancreatic pathology in CF is needed. Glucagon like peptide-1 (GLP-1) from islet α cells potentiates insulin secretion by binding GLP-1 receptors (GLP-1Rs) on β cells. We determined whether expression of GLP-1 and/or its signaling components are reduced in CFRD, thereby contributing to impaired insulin secretion. Immunohistochemistry of pancreas from humans with CFRD versus no-CF/no-diabetes revealed no difference in GLP-1 immunoreactivity per islet area, whereas GLP-1R immunoreactivity per islet area or per insulin-positive islet area was reduced in CFRD. Using spatial transcriptomics, we observed several differentially expressed α- and/or β-cell genes between CFRD and control pancreas. In CFRD, we found upregulation of α-cell PCSK1 which encodes the enzyme (PC1/3) that generates GLP-1, and downregulation of α-cell PCSK1N which inhibits PC1/3. Gene set enrichment analysis also revealed α and β cell-specific pathway dysregulation in CFRD. Together, our data suggest intra-islet GLP-1 is not limiting in CFRD, but its action may be restricted due to reduced GLP-1R protein levels. Thus, restoring β-cell GLP-1R protein expression may improve β-cell function in CFRD.
Collapse
Affiliation(s)
- Sina A Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Computational Medicine Core at Center for Lung Biology, University of Washington, Seattle, Washington, USA
| | - Rachna Vemireddy
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joseph J Castillo
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
- Research and Development Service, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
| | - Brendy S Fountaine
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Rebecca L Hull-Meichle
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
- Research and Development Service, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
- Alberta Diabetes Institute, Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA.
- Research and Development Service, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.
- Veterans Affairs Puget Sound Health Care System, 1660 South Columbian Way (151), Seattle, WA, 98108, USA.
| |
Collapse
|
2
|
Zhang W, Wang R, Yi Z, Guo R, Li Y, Xu Y, Li X, Song J. Investigation of the Expression and Regulation of SCG5 in the Context of the Chromogranin-Secretogranin Family in Malignant Tumors. Protein Pept Lett 2024; 31:657-666. [PMID: 39219421 DOI: 10.2174/0109298665325956240819064853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024]
Abstract
The SCG5 gene has been demonstrated to play an essential role in the development and progression of a range of malignant neoplasms. The regulation of SCG5 expression involves multiple biological pathways. According to relevant studies, SCG5 is differentially expressed in different cancers, and its up- or down-regulation may even affect tumour growth, invasion, and migration, which caught our attention. Therefore, we summarise the regulatory roles played by the SCG5 gene in a variety of cancers and the biological regulatory mechanisms associated with its possible promotion or inhibition of tumour biological behavior, to further explore the potential of SCG5 as a new tumour marker and hopefully provide theoretical guidance for subsequent disease research and treatment.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Rui Wang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Zhongquan Yi
- Central laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
| | - Rongqi Guo
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Yangyang Li
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Yanhan Xu
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Xia Li
- Department of General Medicine, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
| | - Jianxiang Song
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
| |
Collapse
|
3
|
Wu W, Ma M, Ibarra AE, Lu G, Bakshi VP, Li L. Global Neuropeptidome Profiling in Response to Predator Stress in Rat: Implications for Post-Traumatic Stress Disorder. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:1549-1558. [PMID: 37405781 PMCID: PMC11731200 DOI: 10.1021/jasms.3c00027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Traumatic stress triggers or exacerbates multiple psychiatric illnesses, including post-traumatic stress disorder (PTSD). Nevertheless, the neurophysiological mechanisms underlying stress-induced pathology remain unclear, in part due to the limited understanding of neuronal signaling molecules, such as neuropeptides, in this process. Here, we developed mass spectrometry (MS)-based qualitative and quantitative analytical strategies to profile neuropeptides in rats exposed to predator odor (an ethologically relevant analogue of trauma-like stress) versus control subjects (no odor) to determine peptidomic alterations induced by trauma. In total, 628 unique neuropeptides were identified across 5 fear-circuitry-related brain regions. Brain-region-specific changes of several neuropeptide families, including granin, ProSAAS, opioids, cholecystokinin, and tachykinin, were also observed in the stressed group. Neuropeptides from the same protein precursor were found to vary across different brain regions, indicating the site-specific effects of predator stress. This study reveals for the first time the interaction between neuropeptides and traumatic stress, providing insights into the molecular mechanisms of stress-induced psychopathology and suggesting putative novel therapeutic strategies for disorders such as PTSD.
Collapse
Affiliation(s)
- Wenxin Wu
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Min Ma
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Angel Erbey Ibarra
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Gaoyuan Lu
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Vaishali P. Bakshi
- Department of Psychiatry, University of Wisconsin–Madison, Madison, WI 53719, United States
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53705, United States
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States
| |
Collapse
|
4
|
Shakya M, Yildirim T, Lindberg I. Increased expression and retention of the secretory chaperone proSAAS following cell stress. Cell Stress Chaperones 2020; 25:929-941. [PMID: 32607937 PMCID: PMC7591655 DOI: 10.1007/s12192-020-01128-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 11/25/2022] Open
Abstract
The secretory pathway of neurons and endocrine cells contains a variety of mechanisms designed to combat cellular stress. These include not only the unfolded protein response pathways but also diverse chaperone proteins that collectively work to ensure proteostatic control of secreted and membrane-bound molecules. One of the least studied of these chaperones is the neural- and endocrine-specific molecule known as proSAAS. This small chaperone protein acts as a potent anti-aggregant both in vitro and in cellulo and also represents a cerebrospinal fluid biomarker in Alzheimer's disease. In the present study, we have examined the idea that proSAAS, like other secretory chaperones, might represent a stress-responsive protein. We find that exposure of neural and endocrine cells to the cell stressors tunicamycin and thapsigargin increases cellular proSAAS mRNA and protein in Neuro2A cells. Paradoxically, proSAAS secretion is inhibited by these same drugs. Exposure of Neuro2A cells to low concentrations of the hypoxic stress inducer cobalt chloride, or to sodium arsenite, an oxidative stressor, also increases cellular proSAAS content and reduces its secretion. We conclude that the cellular levels of the small secretory chaperone proSAAS are positively modulated by cell stress.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF2, S267, Baltimore, MD, 21201, USA
| | - Taha Yildirim
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF2, S267, Baltimore, MD, 21201, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF2, S267, Baltimore, MD, 21201, USA.
| |
Collapse
|
5
|
Sharker MR, Nou IS, Kho KH. Molecular characterization and spatiotemporal expression of prohormone convertase 2 in the Pacific abalone, Haliotis discus hannai. PLoS One 2020; 15:e0231353. [PMID: 32271824 PMCID: PMC7144994 DOI: 10.1371/journal.pone.0231353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/20/2020] [Indexed: 11/18/2022] Open
Abstract
Prohormone convertases (PCs) are subtilisin-like proteases responsible for the intracellular processing of prohormones and proneuropeptides in vertebrates and invertebrates. The full-length PC2 cDNA sequence was cloned from pleuropedal ganglion of Haliotis discus hannai, consisted of 2254-bp with an open reading frame of 1989-bp and encoded a protein of 662 amino acid residues. The architecture of Hdh PC2 displayed key features of PCs, including a signal peptide, a pro-segment domain with sites for autocatalytic activation, a catalytic domain, and a pro-protein domain (P-domain). It shares the highest homology of its amino acid sequence with the PC2 from H. asinina and to lesser extent with that of Homo sapiens and Rana catesbeiana PC2. Sequence alignment analysis indicated that Hdh PC2 was highly conserved in the catalytic domain, including a catalytic triad of serine proteinases of the subtilisin family at positions Asp-195, His-236, and Ser-412. The cloned sequence contained a canonical integrin binding sequence, and four cysteine residues involved in the formation of an intramolecular disulfide link. Phylogenetic analysis revealed that the Hdh PC2 is robustly clustered with the Has PC2. Quantitative PCR assay demonstrated that the Hdh PC2 was predominantly expressed in the pleuropedal ganglion rather than in other examined tissues. Although PC2 mRNA was expressed throughout the gametogenetic cycle of male and female abalone, the expression level was significantly higher in the ripening stage of female abalone. Also, a significantly higher expression was observed in the pleuropedal ganglion and gonadal tissues at a higher effective accumulative temperature (1000°C). In situ hybridization revealed that the PC2 mRNA expressing neurosecretory cells were distributed in the cortex region of the pleuropedal ganglion. According to the results, it can be concluded that pleuropedal ganglion is the highest site of PC2 activity, and this enzyme might be involved in the abalone reproduction process.
Collapse
Affiliation(s)
- Md Rajib Sharker
- Department of Fisheries Science, College of Fisheries and Ocean Sciences, Chonnam National University, Jeonnam, Republic of Korea
| | - Ill-Sup Nou
- Department of Horticulture, College of Life Science and Natural Resources, Sunchon National University, Jeollanam-do, Republic of Korea
| | - Kang Hee Kho
- Department of Fisheries Science, College of Fisheries and Ocean Sciences, Chonnam National University, Jeonnam, Republic of Korea
| |
Collapse
|
6
|
Carmon O, Laguerre F, Riachy L, Delestre-Delacour C, Wang Q, Tanguy E, Jeandel L, Cartier D, Thahouly T, Haeberlé AM, Fouillen L, Rezazgui O, Schapman D, Haefelé A, Goumon Y, Galas L, Renard PY, Alexandre S, Vitale N, Anouar Y, Montero-Hadjadje M. Chromogranin A preferential interaction with Golgi phosphatidic acid induces membrane deformation and contributes to secretory granule biogenesis. FASEB J 2020; 34:6769-6790. [PMID: 32227388 DOI: 10.1096/fj.202000074r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/28/2020] [Accepted: 03/14/2020] [Indexed: 12/14/2022]
Abstract
Chromogranin A (CgA) is a key luminal actor of secretory granule biogenesis at the trans-Golgi network (TGN) level but the molecular mechanisms involved remain obscure. Here, we investigated the possibility that CgA acts synergistically with specific membrane lipids to trigger secretory granule formation. We show that CgA preferentially interacts with the anionic glycerophospholipid phosphatidic acid (PA). In accordance, bioinformatic analysis predicted a PA-binding domain (PABD) in CgA sequence that effectively bound PA (36:1) or PA (40:6) in membrane models. We identified PA (36:1) and PA (40:6) as predominant species in Golgi and granule membranes of secretory cells, and we found that CgA interaction with these PA species promotes artificial membrane deformation and remodeling. Furthermore, we demonstrated that disruption of either CgA PABD or phospholipase D (PLD) activity significantly alters secretory granule formation in secretory cells. Our findings show for the first time the ability of CgA to interact with PLD-generated PA, which allows membrane remodeling and curvature, key processes necessary to initiate secretory granule budding.
Collapse
Affiliation(s)
- Ophélie Carmon
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Fanny Laguerre
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Lina Riachy
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Charlène Delestre-Delacour
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Qili Wang
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Lydie Jeandel
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Dorthe Cartier
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Tamou Thahouly
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Anne-Marie Haeberlé
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Laetitia Fouillen
- Laboratoire de Biogénèse Membranaire, CNRS, Plateforme Métabolome, Université de Bordeaux, UMR-5200, Villenave D'Ornon, France
| | - Olivier Rezazgui
- INSA Rouen, CNRS, Normandie University, UNIROUEN, COBRA, UMR 6014 and FR 3038, Rouen, France
| | - Damien Schapman
- Normandie University, UNIROUEN, INSERM, PRIMACEN, Rouen, France
| | - Alexandre Haefelé
- INSA Rouen, CNRS, Normandie University, UNIROUEN, COBRA, UMR 6014 and FR 3038, Rouen, France
| | - Yannick Goumon
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Ludovic Galas
- Normandie University, UNIROUEN, INSERM, PRIMACEN, Rouen, France
| | - Pierre-Yves Renard
- INSA Rouen, CNRS, Normandie University, UNIROUEN, COBRA, UMR 6014 and FR 3038, Rouen, France
| | - Stéphane Alexandre
- Polymères, Biopolymères, Surfaces Laboratory, CNRS, Normandie University, UNIROUEN, UMR 6270, Rouen, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Youssef Anouar
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| | - Maité Montero-Hadjadje
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, Normandie University, UNIROUEN, INSERM, U1239, Rouen, France
| |
Collapse
|
7
|
Schernthaner-Reiter MH, Trivellin G, Stratakis CA. Chaperones, somatotroph tumors and the cyclic AMP (cAMP)-dependent protein kinase (PKA) pathway. Mol Cell Endocrinol 2020; 499:110607. [PMID: 31586652 DOI: 10.1016/j.mce.2019.110607] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 02/08/2023]
Abstract
The cAMP-PKA pathway plays an essential role in the pituitary gland, governing cell differentiation and survival, and maintenance of endocrine function. Somatotroph growth hormone transcription and release as well as cell proliferation are regulated by the cAMP-PKA pathway; cAMP-PKA pathway abnormalities are frequently detected in sporadic as well as in hereditary somatotroph tumors and more rarely in other pituitary tumors. Inactivating variants of the aryl hydrocarbon receptor-interacting protein (AIP)-coding gene are the genetic cause of a subset of familial isolated pituitary adenomas (FIPA). Multiple functional links between the co-chaperone AIP and the cAMP-PKA pathway have been described. This review explores the role of chaperones including AIP in normal pituitary function as well as in somatotroph tumors, and their interaction with the cAMP-PKA pathway.
Collapse
Affiliation(s)
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| |
Collapse
|
8
|
Böttcher-Friebertshäuser E, Garten W, Klenk HD. Characterization of Proprotein Convertases and Their Involvement in Virus Propagation. ACTIVATION OF VIRUSES BY HOST PROTEASES 2018. [PMCID: PMC7122180 DOI: 10.1007/978-3-319-75474-1_9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Wolfgang Garten
- Institut für Virologie, Philipps Universität, Marburg, Germany
| | | |
Collapse
|
9
|
Maeda Y, Kudo S, Tsushima K, Sato E, Kubota C, Kayamori A, Bochimoto H, Koga D, Torii S, Gomi H, Watanabe T, Hosaka M. Impaired Processing of Prohormones in Secretogranin III-Null Mice Causes Maladaptation to an Inadequate Diet and Stress. Endocrinology 2018; 159:1213-1227. [PMID: 29281094 DOI: 10.1210/en.2017-00636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 12/15/2017] [Indexed: 11/19/2022]
Abstract
Secretogranin III (SgIII), a member of the granin family, binds both to another granin, chromogranin A (CgA), and to a cholesterol-rich membrane that is destined for secretory granules (SGs). The knockdown of SgIII in adrenocorticotropic hormone (ACTH)-producing AtT-20 cells largely impairs the regulated secretion of CgA and ACTH. To clarify the physiological roles of SgIII in vivo, we analyzed hormone secretion and SG biogenesis in newly established SgIII-knockout (KO) mice. Although the SgIII-KO mice were viable and fertile and exhibited no overt abnormalities under ordinary rearing conditions, a high-fat/high-sucrose diet caused pronounced obesity in the mice. Furthermore, in the SgIII-KO mice compared with wild-type (WT) mice, the stimulated secretion of active insulin decreased substantially, whereas the storage of proinsulin increased in the islets. The plasma ACTH was also less elevated in the SgIII-KO mice than in the WT mice after chronic restraint stress, whereas the storage level of the precursor proopiomelanocortin in the pituitary gland was somewhat increased. These findings suggest that the lack of SgIII causes maladaptation of endocrine cells to an inadequate diet and stress by impairing the proteolytic conversion of prohormones in SGs, whereas SG biogenesis and the basal secretion of peptide hormones under ordinary conditions are ensured by the compensatory upregulation of other residual granins or factors.
Collapse
Affiliation(s)
- Yoshinori Maeda
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Saki Kudo
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Ken Tsushima
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Eri Sato
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Chisato Kubota
- Biosignal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Aika Kayamori
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Hiroki Bochimoto
- Health Care Administration Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Daisuke Koga
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Japan
| | - Seiji Torii
- Biosignal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hiroshi Gomi
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Tsuyoshi Watanabe
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Japan
| | - Masahiro Hosaka
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| |
Collapse
|
10
|
Fricker LD. Carboxypeptidase E and the Identification of Novel Neuropeptides as Potential Therapeutic Targets. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:85-102. [PMID: 29413529 DOI: 10.1016/bs.apha.2017.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peptides and small molecules that bind to peptide receptors are important classes of drugs that are used for a wide variety of different applications. The search for novel neuropeptides traditionally involved a time-consuming approach to purify each peptide to homogeneity and determine its amino acid sequence. The discovery in the 1980s of enkephalin convertase/carboxypeptidase E (CPE), and the observation that this enzyme was involved in the production of nearly every known neuropeptide led to the idea for a one-step affinity purification of CPE substrates. This approach was successfully used to isolate hundreds of known neuropeptides in mouse brain, as well as over a dozen novel peptides. Some of the novel peptides found using this approach are among the most abundant peptides present in brain, but had not been previously identified by traditional approaches. Recently, receptors for two of the novel peptides have been identified, confirming their role as neuropeptides that function in cell-cell signaling. Small molecules that bind to one of these receptors have been developed and found to significantly reduce food intake and anxiety-like behavior in an animal model. This review describes the entire project, from discovery of CPE to the novel peptides and their receptors.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
11
|
Majerova P, Barath P, Michalicova A, Katina S, Novak M, Kovac A. Changes of Cerebrospinal Fluid Peptides due to Tauopathy. J Alzheimers Dis 2017; 58:507-520. [DOI: 10.3233/jad-170110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- AXON Neuroscience R&D, Bratislava, Slovak Republic
| | - Peter Barath
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Alena Michalicova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- AXON Neuroscience R&D, Bratislava, Slovak Republic
| | - Stanislav Katina
- AXON Neuroscience R&D, Bratislava, Slovak Republic
- Institute of Mathematics and Statistics, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- AXON Neuroscience R&D, Bratislava, Slovak Republic
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- AXON Neuroscience R&D, Bratislava, Slovak Republic
- Department of Pharmacology and Toxicology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovak Republic
| |
Collapse
|
12
|
Segerstolpe Å, Palasantza A, Eliasson P, Andersson EM, Andréasson AC, Sun X, Picelli S, Sabirsh A, Clausen M, Bjursell MK, Smith DM, Kasper M, Ämmälä C, Sandberg R. Single-Cell Transcriptome Profiling of Human Pancreatic Islets in Health and Type 2 Diabetes. Cell Metab 2016; 24:593-607. [PMID: 27667667 PMCID: PMC5069352 DOI: 10.1016/j.cmet.2016.08.020] [Citation(s) in RCA: 1025] [Impact Index Per Article: 113.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/13/2016] [Accepted: 08/26/2016] [Indexed: 12/25/2022]
Abstract
Hormone-secreting cells within pancreatic islets of Langerhans play important roles in metabolic homeostasis and disease. However, their transcriptional characterization is still incomplete. Here, we sequenced the transcriptomes of thousands of human islet cells from healthy and type 2 diabetic donors. We could define specific genetic programs for each individual endocrine and exocrine cell type, even for rare δ, γ, ε, and stellate cells, and revealed subpopulations of α, β, and acinar cells. Intriguingly, δ cells expressed several important receptors, indicating an unrecognized importance of these cells in integrating paracrine and systemic metabolic signals. Genes previously associated with obesity or diabetes were found to correlate with BMI. Finally, comparing healthy and T2D transcriptomes in a cell-type resolved manner uncovered candidates for future functional studies. Altogether, our analyses demonstrate the utility of the generated single-cell gene expression resource.
Collapse
Affiliation(s)
- Åsa Segerstolpe
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, 171 77 Stockholm, Sweden; Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Athanasia Palasantza
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Pernilla Eliasson
- Cardiovascular and Metabolic Diseases (CVMD), Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, 431 83 Mölndal, Sweden
| | - Eva-Marie Andersson
- Cardiovascular and Metabolic Diseases (CVMD), Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, 431 83 Mölndal, Sweden
| | - Anne-Christine Andréasson
- Cardiovascular and Metabolic Diseases (CVMD), Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, 431 83 Mölndal, Sweden
| | - Xiaoyan Sun
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Novum, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Simone Picelli
- Ludwig Institute for Cancer Research, 171 77 Stockholm, Sweden
| | - Alan Sabirsh
- Cardiovascular and Metabolic Diseases (CVMD), Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, 431 83 Mölndal, Sweden
| | - Maryam Clausen
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, 431 83 Mölndal, Sweden
| | | | - David M Smith
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, Cambridge Science Park, Milton Road, Cambridge CB4 0WG, UK
| | - Maria Kasper
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Novum, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Carina Ämmälä
- Cardiovascular and Metabolic Diseases (CVMD), Innovative Medicines and Early Development Biotech Unit (iMed), AstraZeneca, 431 83 Mölndal, Sweden
| | - Rickard Sandberg
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, 171 77 Stockholm, Sweden; Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, 141 57 Huddinge, Sweden; Ludwig Institute for Cancer Research, 171 77 Stockholm, Sweden.
| |
Collapse
|
13
|
Ramos-Molina B, Lindberg I. Phosphorylation and Alternative Splicing of 7B2 Reduce Prohormone Convertase 2 Activation. Mol Endocrinol 2015; 29:756-64. [PMID: 25811241 DOI: 10.1210/me.2014-1394] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
FAM20C is a secretory kinase responsible for the phosphorylation of multiple secreted proteins in mammalian cells; it has been shown to phosphorylate serine residues within a variety of different bone proteins. In this work we demonstrate that FAM20C also phosphorylates threonines, specifically those within the N-terminal domain of the neuroendocrine chaperone 7B2. Analysis of the primary sequence of 7B2 revealed that three threonine residues in its N-terminal domain are located within FAM20C consensus motifs: Thr73, Thr99, and Thr111. The individual substitution of Thr73 and Thr111 residues by neutral alanines caused a marked decrease in the total phosphorylation of 7B2. Furthermore, the phosphomimetic substitution of Thr111 by Glu clearly diminished the ability of 7B2 to activate pro-prohormone convertase 2 (PC2) in 7B2-lacking SK-N-MC neuroblastoma cells, suggesting that the phosphorylation of this residue critically impacts the 7B2-proPC2 interaction. However, the phosphomimetic mutation did not alter 7B2's ability to function as an antiaggregant for human islet amyloid polypeptide. FAM20C-mediated phosphorylation of a common alternatively spliced variant of human 7B2 that lacks Ala100 (thus eliminating the Thr99 phosphorylation consensus site) was similar to the Ala-containing protein, but this variant did not activate proPC2 as efficiently as the Ala-containing protein. Although threonines within 7B2 were phosphorylated efficiently, FAM20C was incapable of performing the well-known regulatory threonine phosphorylation of the molecular chaperone binding immunoglobulin protein. Taken together, these results indicate that FAM20C plays a role in 7B2-mediated proPC2 activation by phosphorylating residue Thr111; and that 7B2 function is regulated by alternative splicing.
Collapse
Affiliation(s)
- Bruno Ramos-Molina
- Department of Anatomy and Neurobiology, School of Medicine, University of Maryland-Baltimore, Baltimore, Maryland 21201
| | | |
Collapse
|
14
|
Ferro ES, Rioli V, Castro LM, Fricker LD. Intracellular peptides: From discovery to function. EUPA OPEN PROTEOMICS 2014. [DOI: 10.1016/j.euprot.2014.02.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Hoshino A, Helwig M, Rezaei S, Berridge C, Eriksen JL, Lindberg I. A novel function for proSAAS as an amyloid anti-aggregant in Alzheimer's disease. J Neurochem 2013; 128:419-30. [PMID: 24102330 DOI: 10.1111/jnc.12454] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 01/23/2023]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD) are characterized by an abnormal aggregation of misfolded beta-sheet rich proteins such as β-amyloid (Aβ). Various ubiquitously expressed molecular chaperones control the correct folding of cellular proteins and prevent the accumulation of harmful species. We here describe a novel anti-aggregant chaperone function for the neuroendocrine protein proSAAS, an abundant secretory polypeptide that is widely expressed within neural and endocrine tissues and which has previously been associated with neurodegenerative disease in various proteomics studies. In the brains of 12-month-old APdE9 mice, and in the cortex of a human AD-affected brain, proSAAS immunoreactivity was highly colocalized with amyloid pathology. Immunoreactive proSAAS co-immunoprecipitated with Aβ immunoreactivity in lysates from APdE9 mouse brains. In vitro, proSAAS efficiently prevented the fibrillation of Aβ(1-42) at molar ratios of 1 : 10, and this anti-aggregation effect was dose dependent. Structure-function studies showed that residues 97-180 were sufficient for the anti-aggregation function against Aβ. Finally, inclusion of recombinant proSAAS in the medium of Neuro2a cells, as well as lentiviral-mediated proSAAS over-expression, blocked the neurocytotoxic effect of Aβ(1-42) in Neuro2a cells. Taken together, our results suggest that proSAAS may play a role in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Akina Hoshino
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
16
|
Peinado JR, Sami F, Rajpurohit N, Lindberg I. Blockade of islet amyloid polypeptide fibrillation and cytotoxicity by the secretory chaperones 7B2 and proSAAS. FEBS Lett 2013; 587:3406-11. [PMID: 24042052 DOI: 10.1016/j.febslet.2013.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/03/2013] [Accepted: 09/06/2013] [Indexed: 10/26/2022]
Abstract
The deposition of fibrillated human islet β-cell peptide islet amyloid polypeptide (hIAPP) into amyloid plaques is characteristic of the pathogenesis of islet cell death during type 2 diabetes. We investigated the effects of the neuroendocrine secretory proteins 7B2 and proSAAS on hIAPP fibrillation in vitro and on cytotoxicity. In vitro, 21-kDa 7B2 and proSAAS blocked hIAPP fibrillation. Structure-function studies showed that a central region within 21-kDa 7B2 is important in this effect and revealed the importance of the N-terminal region of proSAAS. Both chaperones blocked the cytotoxic effects of exogenous hIAPP on Rin5f cells; 7B2 generated by overexpression was also effective. ProSAAS and 7B2 may perform a chaperone role as secretory anti-aggregants in normal islet cell function and in type 2 diabetes.
Collapse
Affiliation(s)
- Juan R Peinado
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore, Baltimore, MD 21201, United States
| | | | | | | |
Collapse
|
17
|
Venetikou MS, Meleagros L, Ghatei MA, Bloom SR. Pituitary protein 7B2 plasma levels in patients with liver disease: Comparisons with other hormones and neuropeptides. Oncol Lett 2013; 6:499-506. [PMID: 24137355 PMCID: PMC3789099 DOI: 10.3892/ol.2013.1384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 04/30/2013] [Indexed: 11/29/2022] Open
Abstract
7B2, a protein initially isolated from the porcine pituitary gland, has been identified in numerous animal and human tissues, with the highest concentrations in the pituitary and hypothalamus. The 7B2 molecule is highly evolutionarily conserved and is considered to be indispensable in the function and regulation of proprotein convertase 2 (PC2). In the present study, the plasma 7B2 immunoreactivity (7B2-IR) of 18 patients with liver disease was studied. Of these patients, seven (three male and four female), aged 37–67 [54.6±13.5 (SD)] years, suffered from liver cirrhosis of cryptogenic (n=2) or alcoholic (n=5) aetiology. The remaining 11 patients (four male and seven female), aged 22–76 [56.1±17.6 (SD)] years, suffered from miscellaneous liver abnormalities. The clinical diagnosis was confirmed in the majority of patients by the histological examination of a percutaneous liver biopsy or by appropriate radiological investigations. Plasma bilirubin, alkaline phosphatase, aspartate aminotransferase, albumin, prothrombin time, electrolytes, urea and creatinine were measured. The plasma 7B2-IR levels were estimated using a sensitive radioimmunoassay (RIA), and the elution position of 7B2-IR was verified by gel chromatography. The mean plasma 7B2-IR concentration in patients with liver disease was 99.44±15.9 pmol/l. In the patients with hepatocellular damage due to metastatic tumours [Ca bronchus, carcinoid (n=6)], the 7B2-IR concentrations were significantly higher [185±36.9 pmol/l, (P<0.05)] compared with the overall subjects with liver damage. The results of the present study demonstrate that 7B2-IR is increased in liver disease, with the highest levels detected in patients with tumourous liver conditions.
Collapse
Affiliation(s)
- Maria S Venetikou
- Department of Medical Sciences, Faculty of Health and Caring Professions, Technological Educational Institute (TEI), Athens, Greece
| | | | | | | |
Collapse
|
18
|
Yuan B, Feng JQ, Bowman S, Liu Y, Blank RD, Lindberg I, Drezner MK. Hexa-D-arginine treatment increases 7B2•PC2 activity in hyp-mouse osteoblasts and rescues the HYP phenotype. J Bone Miner Res 2013; 28:56-72. [PMID: 22886699 PMCID: PMC3523095 DOI: 10.1002/jbmr.1738] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 07/31/2012] [Accepted: 08/02/2012] [Indexed: 12/24/2022]
Abstract
Inactivating mutations of the "phosphate regulating gene with homologies to endopeptidases on the X chromosome" (PHEX/Phex) underlie disease in patients with X-linked hypophosphatemia (XLH) and the hyp-mouse, a murine homologue of the human disorder. Although increased serum fibroblast growth factor 23 (FGF-23) underlies the HYP phenotype, the mechanism(s) by which PHEX mutations inhibit FGF-23 degradation and/or enhance production remains unknown. Here we show that treatment of wild-type mice with the proprotein convertase (PC) inhibitor, decanoyl-Arg-Val-Lys-Arg-chloromethyl ketone (Dec), increases serum FGF-23 and produces the HYP phenotype. Because PC2 is uniquely colocalized with PHEX in osteoblasts/bone, we examined if PC2 regulates PHEX-dependent FGF-23 cleavage and production. Transfection of murine osteoblasts with PC2 and its chaperone protein 7B2 cleaved FGF-23, whereas Signe1 (7B2) RNA interference (RNAi) transfection, which limited 7B2 protein production, decreased FGF-23 degradation and increased Fgf-23 mRNA and protein. The mechanism by which decreased 7B2•PC2 activity influences Fgf-23 mRNA was linked to reduced conversion of the precursor to bone morphogenetic protein 1 (proBMP1) to active BMP1, which resulted in limited cleavage of dentin matrix acidic phosphoprotein 1 (DMP1), and consequent increased Fgf-23 mRNA. The significance of decreased 7B2•PC2 activity in XLH was confirmed by studies of hyp-mouse bone, which revealed significantly decreased Sgne1 (7B2) mRNA and 7B2 protein, and limited cleavage of proPC2 to active PC2. The expected downstream effects of these changes included decreased FGF-23 cleavage and increased FGF-23 synthesis, secondary to decreased BMP1-mediated degradation of DMP1. Subsequent Hexa-D-Arginine treatment of hyp-mice enhanced bone 7B2•PC2 activity, normalized FGF-23 degradation and production, and rescued the HYP phenotype. These data suggest that decreased PHEX-dependent 7B2•PC2 activity is central to the pathogenesis of XLH.
Collapse
Affiliation(s)
- Baozhi Yuan
- Department of Medicine, University of Wisconsin-Madison and Geriatric Research and Education Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53792
| | - Jian Q. Feng
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX 75246
| | - Stephen Bowman
- Department of Medicine, University of Wisconsin-Madison and Geriatric Research and Education Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53792
| | - Ying Liu
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX 75246
| | - Robert D. Blank
- Department of Medicine, University of Wisconsin-Madison and Geriatric Research and Education Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53792
| | - Iris Lindberg
- Dept. of Anatomy and Neurobiology, University of Maryland Baltimore, Baltimore, MD 21201
| | - Marc K. Drezner
- Department of Medicine, University of Wisconsin-Madison and Geriatric Research and Education Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53792
| |
Collapse
|
19
|
Helwig M, Hoshino A, Berridge C, Lee SN, Lorenzen N, Otzen DE, Eriksen JL, Lindberg I. The neuroendocrine protein 7B2 suppresses the aggregation of neurodegenerative disease-related proteins. J Biol Chem 2012; 288:1114-24. [PMID: 23172224 PMCID: PMC3542996 DOI: 10.1074/jbc.m112.417071] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer (AD) and Parkinson (PD) are characterized by abnormal aggregation of misfolded β-sheet-rich proteins, including amyloid-β (Aβ)-derived peptides and tau in AD and α-synuclein in PD. Correct folding and assembly of these proteins are controlled by ubiquitously expressed molecular chaperones; however, our understanding of neuron-specific chaperones and their involvement in the pathogenesis of neurodegenerative diseases is limited. We here describe novel chaperone-like functions for the secretory protein 7B2, which is widely expressed in neuronal and endocrine tissues. In in vitro experiments, 7B2 efficiently prevented fibrillation and formation of Aβ1–42, Aβ1–40, and α-synuclein aggregates at a molar ratio of 1:10. In cell culture experiments, inclusion of recombinant 7B2, either in the medium of Neuro-2A cells or intracellularly via adenoviral 7B2 overexpression, blocked the neurocytotoxic effect of Aβ1–42 and significantly increased cell viability. Conversely, knockdown of 7B2 by RNAi increased Aβ1–42-induced cytotoxicity. In the brains of APP/PSEN1 mice, a model of AD amyloidosis, immunoreactive 7B2 co-localized with aggregation-prone proteins and their respective aggregates. Furthermore, in the hippocampus and substantia nigra of human AD- and PD-affected brains, 7B2 was highly co-localized with Aβ plaques and α-synuclein deposits, strongly suggesting physiological association. Our data provide insight into novel functions of 7B2 and establish this neural protein as an anti-aggregation chaperone associated with neurodegenerative disease.
Collapse
Affiliation(s)
- Michael Helwig
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Dasgupta I, Sanglas L, Enghild JJ, Lindberg I. The neuroendocrine protein 7B2 is intrinsically disordered. Biochemistry 2012; 51:7456-64. [PMID: 22947085 PMCID: PMC3457758 DOI: 10.1021/bi300871k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The small neuroendocrine protein 7B2 has been shown to
be required
for the productive maturation of proprotein convertase 2 (proPC2)
to an active enzyme form; this action is accomplished via its ability
to block aggregation of proPC2 into nonactivatable forms. Recent data
show that 7B2 can also act as a postfolding chaperone to block the
aggregation of a number of other proteins, for example, α-synuclein.
To gain insight into the mechanism of action of 7B2 in blocking protein
aggregation, we performed structural studies of this protein using
gel filtration chromatography, intrinsic tryptophan fluorescence,
1-anilino-8-naphthalenesulfonate (ANS) binding, circular dichroism
(CD), and nuclear magnetic resonance (NMR) spectroscopy. Gel filtration
studies indicated that 7B2 exists as an extended monomer, eluting
at a molecular mass higher than that expected for a globular protein
of similar size. However, chemical cross-linking showed that 7B2 exhibits
concentration-dependent oligomerization. CD experiments showed that
both full-length 27 kDa 7B2 and the C-terminally truncated 21 kDa
form lack appreciable secondary structure, although the longer protein
exhibited more structural content than the latter, as demonstrated
by intrinsic and ANS fluorescence studies. NMR spectra confirmed the
lack of structure in native 7B2, but a disorder-to-order transition
was observed upon incubation with one of its client proteins, α-synuclein.
We conclude that 7B2 is a natively disordered protein whose function
as an antiaggregant chaperone is likely facilitated by its lack of
appreciable secondary structure and tendency to form oligomers.
Collapse
Affiliation(s)
- Indrani Dasgupta
- Department of Anatomy and Neurobiology, School of Medicine, University of Maryland-Baltimore, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
21
|
Jansen EJR, van Bakel NHM, Olde Loohuis NFM, Hafmans TGM, Arentsen T, Coenen AJM, Scheenen WJJM, Martens GJM. Identification of domains within the V-ATPase accessory subunit Ac45 involved in V-ATPase transport and Ca2+-dependent exocytosis. J Biol Chem 2012; 287:27537-46. [PMID: 22736765 DOI: 10.1074/jbc.m112.356105] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The vacuolar (H(+))-ATPase (V-ATPase) is crucial for maintenance of the acidic microenvironment in intracellular organelles, whereas its membrane-bound V(0)-sector is involved in Ca(2+)-dependent membrane fusion. In the secretory pathway, the V-ATPase is regulated by its type I transmembrane and V(0)-associated accessory subunit Ac45. To execute its function, the intact-Ac45 protein is proteolytically processed to cleaved-Ac45 thereby releasing its N-terminal domain. Here, we searched for the functional domains within Ac45 by analyzing a set of deletion mutants close to the in vivo situation, namely in transgenic Xenopus intermediate pituitary melanotrope cells. Intact-Ac45 was poorly processed and accumulated in the endoplasmic reticulum of the transgenic melanotrope cells. In contrast, cleaved-Ac45 was efficiently transported through the secretory pathway, caused an accumulation of the V-ATPase at the plasma membrane and reduced dopaminergic inhibition of Ca(2+)-dependent peptide secretion. Surprisingly, removal of the C-tail from intact-Ac45 caused cellular phenotypes also found for cleaved-Ac45, whereas C-tail removal from cleaved-Ac45 still allowed its transport to the plasma membrane, but abolished V-ATPase recruitment into the secretory pathway and left dopaminergic inhibition of the cells unaffected. We conclude that domains located in the N- and C-terminal portions of the Ac45 protein direct its trafficking, V-ATPase recruitment and Ca(2+)-dependent-regulated exocytosis.
Collapse
Affiliation(s)
- Eric J R Jansen
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition, and Behaviour and Nijmegen Centre for Molecular Life Sciences (NCMLS), Faculty of Science, Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Tamura Y, Shintani M, Inoue H, Monden M, Shiomi H. Regulatory mechanism of body temperature in the central nervous system during the maintenance phase of hibernation in Syrian hamsters: Involvement of β-endorphin. Brain Res 2012; 1448:63-70. [DOI: 10.1016/j.brainres.2012.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 01/19/2012] [Accepted: 02/02/2012] [Indexed: 01/17/2023]
|
23
|
|
24
|
Vázquez-Martínez R, Díaz-Ruiz A, Almabouada F, Rabanal-Ruiz Y, Gracia-Navarro F, Malagón MM. Revisiting the regulated secretory pathway: from frogs to human. Gen Comp Endocrinol 2012; 175:1-9. [PMID: 21907200 DOI: 10.1016/j.ygcen.2011.08.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 08/16/2011] [Accepted: 08/20/2011] [Indexed: 01/01/2023]
Abstract
The regulated secretory pathway is a hallmark of endocrine and neuroendocrine cells. This process comprises different sequential steps, including ER-associated protein synthesis, ER-to-Golgi protein transport, Golgi-associated posttranslational modification, sorting and packing of secretory proteins into carrier granules, cytoskeleton-based granule transport towards the plasma membrane and tethering, docking and fusion of granules with specialized releasing zones in the plasma membrane. Each one of these steps is tightly regulated by a large number of factors that function in a spatially and temporarily coordinated fashion. During the past three decades, much effort has been devoted to characterize the precise role of the yet-known proteins participating in the different steps of this process and to identify new regulatory factors in order to obtain a unifying picture of the secretory pathway. In spite of this and given the enormous complexity of the process, certain steps are not fully understood yet and many players remain to be identified. In this review, we offer a summary of the current knowledge on the main molecular mechanisms that govern and ensure the correct release of secretory proteins. In addition, we have integrated the advance on the field made possible by studies carried out in non-mammalian vertebrates, which, although not very numerous, have substantially contributed to acquire a mechanistic understanding of the regulated secretory pathway.
Collapse
Affiliation(s)
- Rafael Vázquez-Martínez
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica, University of Córdoba, 14014-Córdoba, Spain.
| | | | | | | | | | | |
Collapse
|
25
|
Bartolomucci A, Possenti R, Mahata SK, Fischer-Colbrie R, Loh YP, Salton SRJ. The extended granin family: structure, function, and biomedical implications. Endocr Rev 2011; 32:755-97. [PMID: 21862681 PMCID: PMC3591675 DOI: 10.1210/er.2010-0027] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The chromogranins (chromogranin A and chromogranin B), secretogranins (secretogranin II and secretogranin III), and additional related proteins (7B2, NESP55, proSAAS, and VGF) that together comprise the granin family subserve essential roles in the regulated secretory pathway that is responsible for controlled delivery of peptides, hormones, neurotransmitters, and growth factors. Here we review the structure and function of granins and granin-derived peptides and expansive new genetic evidence, including recent single-nucleotide polymorphism mapping, genomic sequence comparisons, and analysis of transgenic and knockout mice, which together support an important and evolutionarily conserved role for these proteins in large dense-core vesicle biogenesis and regulated secretion. Recent data further indicate that their processed peptides function prominently in metabolic and glucose homeostasis, emotional behavior, pain pathways, and blood pressure modulation, suggesting future utility of granins and granin-derived peptides as novel disease biomarkers.
Collapse
Affiliation(s)
- Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | |
Collapse
|
26
|
Helwig M, Lee SN, Hwang JR, Ozawa A, Medrano JF, Lindberg I. Dynamic modulation of prohormone convertase 2 (PC2)-mediated precursor processing by 7B2 protein: preferential effect on glucagon synthesis. J Biol Chem 2011; 286:42504-42513. [PMID: 22013069 DOI: 10.1074/jbc.m111.281980] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The small neuroendocrine protein 7B2 is required for the production of active prohormone convertase 2 (PC2), an enzyme involved in the synthesis of peptide hormones, such as glucagon and proopiomelanocortin-derived α-melanocyte-stimulating hormone. However, whether 7B2 can dynamically modulate peptide production through regulation of PC2 activity remains unclear. Infection of the pancreatic alpha cell line α-TC6 with 7B2-encoding adenovirus efficiently increased production of glucagon, whereas siRNA-mediated knockdown of 7B2 significantly decreased stored glucagon. Furthermore, rescue of 7B2 expression in primary pituitary cultures prepared from 7B2 null mice restored melanocyte-stimulating hormone production, substantiating the role of 7B2 as a regulatory factor in peptide biosynthesis. In anterior pituitary and pancreatic beta cell lines, however, overexpression of 7B2 affected neither production nor secretion of peptides despite increased release of active PC2. In direct contrast, 7B2 overexpression decreased the secretion and increased the activity of PC2 within α-TC6 cells; the increased intracellular concentration of active PC2 within these cells may therefore account for the enhanced production of glucagon. In line with these findings, we found elevated circulating glucagon levels in 7B2-overexpressing cast/cast mice in vivo. Surprisingly, when proopiomelanocortin and proglucagon were co-expressed in either pituitary or pancreatic alpha cell lines, proglucagon processing was preferentially decreased when 7B2 was knocked down. Taken together, these results suggest that proglucagon cleavage has a greater dependence on PC2 activity than other precursors and moreover that 7B2-dependent routing of PC2 to secretory granules is cell line-specific. The manipulation of 7B2 could therefore represent an effective way to selectively regulate synthesis of certain PC2-dependent peptides.
Collapse
Affiliation(s)
- Michael Helwig
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Sang-Nam Lee
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Jae Ryoung Hwang
- Molecular Therapy Research Center, Sungkyunkwan University, Seoul 135-710, Korea
| | - Akihiko Ozawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Juan F Medrano
- Department of Animal Science, University of California, Davis, California 95616-8521
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201.
| |
Collapse
|
27
|
Waha A, Felsberg J, Hartmann W, Hammes J, von dem Knesebeck A, Endl E, Pietsch T, Waha A. Frequent epigenetic inactivation of the chaperone SGNE1/7B2 in human gliomas. Int J Cancer 2011; 131:612-22. [PMID: 21901745 DOI: 10.1002/ijc.26416] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 08/16/2011] [Indexed: 11/08/2022]
Abstract
In a genome-wide screen using DMH (differential methylation hybridization) we have identified a CpG island within the 5' region and untranslated first exon of the secretory granule neuroendocrine protein 1 gene (SGNE1/7B2) that showed hypermethylation in low- and high-grade astrocytomas compared to normal brain tissue. Pyrosequencing was performed to confirm the methylation status of this CpG island in 89 astrocytic gliomas of different malignancy grades and six glioma cell lines. Hypermethylation of SGNE1/7B2 was significantly more frequent in diffuse low-grade astrocytomas as well as secondary glioblastomas and anaplastic astrocytomas as compared to primary glioblastomas. mRNA expression analysis by real-time RT-PCR indicates that SGNE1/7B2 expression is downregulated in astrocytic gliomas compared to white matter samples. Treatment of glioma cells with the demethylating agent 5-aza-2'-deoxycytidine restores the transcription of SGNE1/7B2. Overexpression of SGNE1/7B2 in T98G, A172 and U373MG glioblastoma cells significantly suppressed focus formation and led to a significant increase in apoptotic cells as determined by flow cytometric analysis in T98G cells. In summary, we have identified SGNE1/7B2 as a novel target silenced by DNA methylation in astrocytic gliomas. The high incidence of this alteration and the significant effects of SGNE1/7B2 on the growth and apoptosis of glioblastoma cells provide a first proof for a functional implication of SGNE1/7B2 inactivation in the molecular pathology of gliomas.
Collapse
Affiliation(s)
- Anke Waha
- Department of Neuropathology, University of Bonn, Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Abstract
We know neuropeptides now for over 40 years as chemical signals in the brain. The discovery of neuropeptides is founded on groundbreaking research in physiology, endocrinology, and biochemistry during the last century and has been built on three seminal notions: (1) peptide hormones are chemical signals in the endocrine system; (2) neurosecretion of peptides is a general principle in the nervous system; and (3) the nervous system is responsive to peptide signals. These historical lines have contributed to how neuropeptides can be defined today: "Neuropeptides are small proteinaceous substances produced and released by neurons through the regulated secretory route and acting on neural substrates." Thus, neuropeptides are the most diverse class of signaling molecules in the brain engaged in many physiological functions. According to this definition almost 70 genes can be distinguished in the mammalian genome, encoding neuropeptide precursors and a multitude of bioactive neuropeptides. In addition, among cytokines, peptide hormones, and growth factors there are several subfamilies of peptides displaying most of the hallmarks of neuropeptides, for example neural chemokines, cerebellins, neurexophilins, and granins. All classical neuropeptides as well as putative neuropeptides from the latter families are presented as a resource.
Collapse
Affiliation(s)
- J Peter H Burbach
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
30
|
Patel YC, Liu J, Galanopoulou A, Papachristou DN. Production, Action, and Degradation of Somatostatin. Compr Physiol 2011. [DOI: 10.1002/cphy.cp070209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
31
|
Kuhla B, Albrecht D, Bruckmaier R, Viergutz T, Nürnberg G, Metges CC. Proteome and radioimmunoassay analyses of pituitary hormones and proteins in response to feed restriction of dairy cows. Proteomics 2010; 10:4491-500. [DOI: 10.1002/pmic.201000383] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
32
|
Jansen EJR, Hafmans TGM, Martens GJM. V-ATPase-mediated granular acidification is regulated by the V-ATPase accessory subunit Ac45 in POMC-producing cells. Mol Biol Cell 2010; 21:3330-9. [PMID: 20702583 PMCID: PMC2947469 DOI: 10.1091/mbc.e10-04-0274] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The regulation of the V-ATPase, the proton pump mediating intraorganellar acidification, is still elusive. We find that excess of the neuroendocrine V-ATPase accessory subunit Ac45 reduces the intragranular pH and consequently disturbs prohormone convertase activation and prohormone processing. Thus, Ac45 represents the first V-ATPase regulator. The vacuolar (H+)-ATPase (V-ATPase) is an important proton pump, and multiple critical cell-biological processes depend on the proton gradient provided by the pump. Yet, the mechanism underlying the control of the V-ATPase is still elusive but has been hypothesized to involve an accessory subunit of the pump. Here we studied as a candidate V-ATPase regulator the neuroendocrine V-ATPase accessory subunit Ac45. We transgenically manipulated the expression levels of the Ac45 protein specifically in Xenopus intermediate pituitary melanotrope cells and analyzed in detail the functioning of the transgenic cells. We found in the transgenic melanotrope cells the following: i) significantly increased granular acidification; ii) reduced sensitivity for a V-ATPase-specific inhibitor; iii) enhanced early processing of proopiomelanocortin (POMC) by prohormone convertase PC1; iv) reduced, neutral pH–dependent cleavage of the PC2 chaperone 7B2; v) reduced 7B2-proPC2 dissociation and consequently reduced proPC2 maturation; vi) decreased levels of mature PC2 and consequently reduced late POMC processing. Together, our results show that the V-ATPase accessory subunit Ac45 represents the first regulator of the proton pump and controls V-ATPase-mediated granular acidification that is necessary for efficient prohormone processing.
Collapse
Affiliation(s)
- Eric J R Jansen
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition, and Behaviour and Nijmegen Centre for Molecular Life Sciences (NCMLS), Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands
| | | | | |
Collapse
|
33
|
Distribution of 7B2 (secretogranin V)-like immunoreactivity in the Japanese red-bellied newt (Cynops pyrrhogaster) pituitary. Tissue Cell 2010; 42:176-80. [DOI: 10.1016/j.tice.2010.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Revised: 03/29/2010] [Accepted: 03/29/2010] [Indexed: 10/19/2022]
|
34
|
Devreese B, Sergeant K, Van Bakel NHM, Debyser G, Van Beeumen J, Martens GJM, Van Herp F. A proteome map of the pituitary melanotrope cell activated by black-background adaptation ofXenopus laevis. Proteomics 2010; 10:574-80. [DOI: 10.1002/pmic.200900281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
35
|
Koo BH, Apte SS. Cell-surface processing of the metalloprotease pro-ADAMTS9 is influenced by the chaperone GRP94/gp96. J Biol Chem 2009; 285:197-205. [PMID: 19875450 DOI: 10.1074/jbc.m109.039677] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A disintegrin-like and metalloprotease domain with thrombospondin type 1 motifs 9 (ADAMTS9) is a highly conserved metalloprotease that has been identified as a tumor suppressor gene and is required for normal mouse development. The secreted ADAMTS9 zymogen undergoes proteolytic excision of its N-terminal propeptide by the proprotein convertase furin. However, in contrast to other metalloproteases, propeptide excision occurs at the cell surface and leads to decreased activity of the zymogen. Here, we investigated the potential cellular mechanisms regulating ADAMTS9 biosynthesis and cell-surface processing by analysis of molecular complexes formed by a construct containing the propeptide and catalytic domain of pro-ADAMTS9 (Pro-Cat) in HEK293F cells. Cross-linking of cellular proteins bound to Pro-Cat followed by mass spectrometric analysis identified UDP-glucose:glycoprotein glucosyltransferase I, heat shock protein gp96 (GRP94), BiP (GRP78), and ERdj3 (Hsp40 homolog) as associated proteins. gp96 and BiP were present at the cell surface in an immunoprecipitable complex with pro-ADAMTS9 and furin. Treatment with geldanamycin, an inhibitor of the HSP90alpha family (including gp96), led to decreased furin processing of pro-ADAMTS9 and accumulation of the unprocessed pro-ADAMTS9 at the cell surface. gp96 siRNA down-regulated the levels of cell-surface pro-ADAMTS9 and furin, whereas the levels of cell-surface pro-ADAMTS9, but not of cell-surface furin, were decreased upon treatment with BiP siRNA. These data identify for the first time the cellular chaperones associated with secretion of an ADAMTS protease and suggest a role for gp96 in modulating pro-ADAMTS9 processing.
Collapse
Affiliation(s)
- Bon-Hun Koo
- Department of Biochemistry, College of Science, Yonsei University, Seoul 120-749, Korea
| | | |
Collapse
|
36
|
Abstract
Insulin, the major secreted product of the beta-cells of the islets of Langerhans, is initially synthesized as a precursor (preproinsulin), from which the mature hormone is excised by a series of proteolytic cleavages. This review provides a personal narrative of some of the key research projects leading to the identification of the central processing enzymes as proprotein convertase 1, proprotein convertase 2, and carboxypeptidase E. It also discusses the central roles of the intragranular environment and chaperone-like proteins in modulating processing activity.
Collapse
Affiliation(s)
- Howard W Davidson
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver, CO, USA.
| |
Collapse
|
37
|
Kudo H, Liu J, Jansen EJR, Ozawa A, Panula P, Martens GJM, Lindberg I. Identification of proSAAS homologs in lower vertebrates: conservation of hydrophobic helices and convertase-inhibiting sequences. Endocrinology 2009; 150:1393-9. [PMID: 18948394 PMCID: PMC2654743 DOI: 10.1210/en.2008-1301] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The prohormone convertases (PCs) 1/3 and 2 accomplish the major proteolytic cleavage events in neuroendocrine tissues; each of these convertases has a small associated binding protein that inhibits convertase action in the secretory pathway. The proSAAS protein binds to PC1/3, whereas the 7B2 protein binds to PC2. However, both convertase-binding proteins are more widely expressed than their cognate enzymes, suggesting that they may perform other functions as well. All known mammalian proSAASs are over 85% conserved; thus, identifying functionally important segments has been impossible. Here, we report the first identification of nonmammalian proSAAS molecules, from Xenopus and zebrafish (Danio rerio). Although these two proteins show an overall amino acid sequence identity of only 29 and 30% with mouse proSAAS, two 14-16 residue hydrophobic segments (predicted to form alpha-helices) and two, nine through 11 residue sequences containing basic convertase cleavage sites are highly conserved; therefore, these sequences may be of functional importance. Confidence that these nonmammalian molecules represent authentic proSAAS is supported by the finding that both inhibit mouse PC1/3 with nanomolar inhibition constants; human furin was not inhibited. In vitro, the two proteins were cleaved by PC2 and furin to three or more peptide products. Both zebrafish and Xenopus proSAAS exhibited neural and endocrine distributions, as assessed by in situ and PCR experiments, respectively. In summary, the identification of proSAAS molecules in lower vertebrates provides clues as to functional regions within this widely expressed neuroendocrine protein.
Collapse
Affiliation(s)
- H Kudo
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Pax6 is important in the development of the pancreas and was previously shown to regulate pancreatic endocrine differentiation, as well as the insulin, glucagon, and somatostatin genes. Prohormone convertase 2 (PC2) is the main processing enzyme in pancreatic alpha cells, where it processes proglucagon to produce glucagon under the spatial and temporal control of 7B2, which functions as a molecular chaperone. To investigate the role of Pax6 in glucagon biosynthesis, we studied potential target genes in InR1G9 alpha cells transfected with Pax6 small interfering RNA and in InR1G9 clones expressing a dominant-negative form of Pax6. We now report that Pax6 controls the expression of the PC2 and 7B2 genes. By binding and transactivation studies, we found that Pax6 indirectly regulates PC2 gene transcription through cMaf and Beta2/NeuroD1 while it activates the 7B2 gene both directly and indirectly through the same transcription factors, cMaf and Beta2/NeuroD1. We conclude that Pax6 is critical for glucagon biosynthesis and processing by directly and indirectly activating the glucagon gene through cMaf and Beta2/NeuroD1, as well as the PC2 and 7B2 genes.
Collapse
|
39
|
Wang S, Han J, Wang Y, Lu W, Chi C. Design of peptide inhibitors for furin based on the C-terminal fragment of histone H1.2. Acta Biochim Biophys Sin (Shanghai) 2008. [PMID: 18850049 PMCID: PMC7110110 DOI: 10.1111/j.1745-7270.2008.00470.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The mammalian proprotein convertase furin has been found to play an important role in diverse physiological and pathological events, such as the activation of viral glycoproteins and bacterial exotoxins. Small, non‐toxic and highly active, furin inhibitors are considered to be attractive drug candidates for diseases caused by virus and bacteria. In this study, a series of peptide inhibitors were designed and synthesized based on the C‐terminal fragment of histone H1.2, which has an inhibitory effect on furin. Replacing the reactive site of inhibitors with the consensus substrate recognition sequence of furin has been found to increase inhibitory activity greatly. The most potent inhibitor, I4, with 14 amino acid residues has a Ki value of 17 nM for furin. Although most of the synthesized peptides were temporary inhibitors, the inhibitor I5, with nine amino acids, retained its full potency, even after a 3 h incubation period with furin at 37 °C. These inhibitors may potentially lead to the development of anti‐viral and antibacterial drug compounds.
Collapse
Affiliation(s)
- Suming Wang
- School of Life Sciences, University of Science and Technology of China, Anhui 230027, China
| | | | | | | | | |
Collapse
|
40
|
Chrétien M, Seidah NG, Basak A, Mbikay M. Proprotein convertases as therapeutic targets. Expert Opin Ther Targets 2008; 12:1289-300. [DOI: 10.1517/14728222.12.10.1289] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
Morvan J, Tooze SA. Discovery and progress in our understanding of the regulated secretory pathway in neuroendocrine cells. Histochem Cell Biol 2008; 129:243-52. [PMID: 18197413 PMCID: PMC2248607 DOI: 10.1007/s00418-008-0377-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2008] [Indexed: 01/24/2023]
Abstract
In this review we start with a historical perspective beginning with the early morphological work done almost 50 years ago. The importance of these pioneering studies is underscored by our brief summary of the key questions addressed by subsequent research into the mechanism of secretion. We then highlight important advances in our understanding of the formation and maturation of neuroendocrine secretory granules, first using in vitro reconstitution systems, then most recently biochemical approaches, and finally genetic manipulations in vitro and in vivo.
Collapse
Affiliation(s)
- Joëlle Morvan
- London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London, UK
| | | |
Collapse
|
42
|
Bontemps Y, Scamuffa N, Calvo F, Khatib A. Potential opportunity in the development of new therapeutic agents based on endogenous and exogenous inhibitors of the proprotein convertases. Med Res Rev 2007; 27:631-48. [PMID: 17019676 PMCID: PMC7168524 DOI: 10.1002/med.20072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The proprotein convertases (PCs) are responsible for the endoproteolytic processing of various protein precursors (e.g., growth factors, receptors, adhesion molecules, and matrix metalloproteinases) implicated in several diseases such as obesity, diabetes, atherosclerosis, cancer, and Alzheimer disease. The potential clinical and pharmacological role of the PCs has fostered the development of various PC‐inhibitors. In this review we summarized the recent findings on PCs inhibitors, their mode of actions and potential use in the therapy of various diseases. © 2006 Wiley Periodicals, Inc. Med Res Rev, 27, No. 5, 631–648, 2007
Collapse
Affiliation(s)
- Yannick Bontemps
- INSERM, U 716, Equipe AVENIR, Institut de Génétique Moléculaire, Paris 75010, France
- Université Paris 7, Paris 75251, France
| | - Nathalie Scamuffa
- INSERM, U 716, Equipe AVENIR, Institut de Génétique Moléculaire, Paris 75010, France
- Université Paris 7, Paris 75251, France
| | - Fabien Calvo
- INSERM, U 716, Equipe AVENIR, Institut de Génétique Moléculaire, Paris 75010, France
- Université Paris 7, Paris 75251, France
| | - Abdel‐Majid Khatib
- INSERM, U 716, Equipe AVENIR, Institut de Génétique Moléculaire, Paris 75010, France
- Université Paris 7, Paris 75251, France
| |
Collapse
|
43
|
Strating JRPM, Bouw G, Hafmans TGM, Martens GJM. Disparate effects of p24alpha and p24delta on secretory protein transport and processing. PLoS One 2007; 2:e704. [PMID: 17684551 PMCID: PMC1933603 DOI: 10.1371/journal.pone.0000704] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Accepted: 07/02/2007] [Indexed: 11/20/2022] Open
Abstract
Background The p24 family is thought to be somehow involved in endoplasmic reticulum (ER)-to-Golgi protein transport. A subset of the p24 proteins (p24α3, -β1, -γ3 and -δ2) is upregulated when Xenopus laevis intermediate pituitary melanotrope cells are physiologically activated to produce vast amounts of their major secretory cargo, the prohormone proopiomelanocortin (POMC). Methodology/Principal Findings Here we find that transgene expression of p24α3 or p24δ2 specifically in the Xenopus melanotrope cells in both cases causes an effective displacement of the endogenous p24 proteins, resulting in severely distorted p24 systems and disparate melanotrope cell phenotypes. Transgene expression of p24α3 greatly reduces POMC transport and leads to accumulation of the prohormone in large, ER-localized electron-dense structures, whereas p24δ2-transgenesis does not influence the overall ultrastructure of the cells nor POMC transport and cleavage, but affects the Golgi-based processes of POMC glycomaturation and sulfation. Conclusions/Significance Transgenic expression of two distinct p24 family members has disparate effects on secretory pathway functioning, illustrating the specificity and non-redundancy of our transgenic approach. We conclude that members of the p24 family furnish subcompartments of the secretory pathway with specific sets of machinery cargo to provide the proper microenvironments for efficient and correct secretory protein transport and processing.
Collapse
Affiliation(s)
- Jeroen R. P. M. Strating
- Department of Molecular Animal Physiology, Nijmegen Centre for Molecular Life Sciences, Institute for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Gerrit Bouw
- Department of Molecular Animal Physiology, Nijmegen Centre for Molecular Life Sciences, Institute for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Theo G. M. Hafmans
- Department of Molecular Animal Physiology, Nijmegen Centre for Molecular Life Sciences, Institute for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Gerard J. M. Martens
- Department of Molecular Animal Physiology, Nijmegen Centre for Molecular Life Sciences, Institute for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
44
|
Bouatia-Naji N, Vatin V, Lecoeur C, Heude B, Proença C, Veslot J, Jouret B, Tichet J, Charpentier G, Marre M, Balkau B, Froguel P, Meyre D. Secretory granule neuroendocrine protein 1 (SGNE1) genetic variation and glucose intolerance in severe childhood and adult obesity. BMC MEDICAL GENETICS 2007; 8:44. [PMID: 17617923 PMCID: PMC1936990 DOI: 10.1186/1471-2350-8-44] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Accepted: 07/07/2007] [Indexed: 01/22/2023]
Abstract
Background 7B2 is a regulator/activator of the prohormone convertase 2 which is involved in the processing of numerous neuropeptides, including insulin, glucagon and pro-opiomelanocortin. We have previously described a suggestive genetic linkage peak with childhood obesity on chr15q12-q14, where the 7B2 encoding gene, SGNE1 is located. The aim of this study is to analyze associations of SGNE1 genetic variation with obesity and metabolism related quantitative traits. Methods We screened SGNE1 for genetic variants in obese children and genotyped 12 frequent single nucleotide polymorphisms (SNPs). Case control analyses were performed in 1,229 obese (534 children and 695 adults), 1,535 individuals with type 2 diabetes and 1,363 controls, all French Caucasians. We also studied 4,922 participants from the D.E.S.I.R prospective population-based cohort. Results We did not find any association between SGNE1 SNPs and childhood or adult obesity. However, the 5' region SNP -1,701A>G associated with higher area under glucose curve after oral glucose tolerance test (p = 0.0005), higher HOMA-IR (p = 0.005) and lower insulinogenic index (p = 0.0003) in obese children. Similar trends were found in obese adults. SNP -1,701A>G did not associate with risk of T2D but tends to associate with incidence of type 2 diabetes (HR = 0.75 95%CI [0.55–1.01]; p = 0.06) in the prospective cohort. Conclusion SGNE1 genetic variation does not contribute to obesity and common forms of T2D but may worsen glucose intolerance and insulin resistance, especially in the background of severe and early onset obesity. Further molecular studies are required to understand the molecular bases involved in this process.
Collapse
Affiliation(s)
| | - Vincent Vatin
- CNRS-8090-Institute of Biology, Pasteur Institute, Lille, France
| | - Cécile Lecoeur
- CNRS-8090-Institute of Biology, Pasteur Institute, Lille, France
| | | | | | - Jacques Veslot
- CNRS-8090-Institute of Biology, Pasteur Institute, Lille, France
| | | | | | | | | | | | - Philippe Froguel
- CNRS-8090-Institute of Biology, Pasteur Institute, Lille, France
- Genomic Medicine, Hammersmith Hospital, Imperial College London, UK
| | - David Meyre
- CNRS-8090-Institute of Biology, Pasteur Institute, Lille, France
| |
Collapse
|
45
|
Mbikay M, Croissandeau G, Sirois F, Anini Y, Mayne J, Seidah NG, Chrétien M. A targeted deletion/insertion in the mouse Pcsk1 locus is associated with homozygous embryo preimplantation lethality, mutant allele preferential transmission and heterozygous female susceptibility to dietary fat. Dev Biol 2007; 306:584-98. [PMID: 17490633 DOI: 10.1016/j.ydbio.2007.03.523] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Revised: 03/02/2007] [Accepted: 03/27/2007] [Indexed: 01/08/2023]
Abstract
Proprotein convertase 1 (PC1) is a neuroendocrine proteinase involved in the proteolytic activation of precursors to hormones and neuropeptides. To determine the physiological importance of PC1, we produced a mutant mouse from embryonic stem cells in which its locus (Pcsk1) had been inactivated by homologous recombination. The inactivating mutation consisted of a 32.7-kb internal deletion and a 1.8 kb insertion of the bacterial neomycin resistance gene (neo) under the mouse phosphoglycerate kinase 1 protein (PGKneo). Intercross of Pcsk1(+/-) mice produced no Pcsk1(-/-) offspring or blastocysts; in addition, more than 80% of the offspring were Pcsk1(+/-). These observations suggested that the mutation caused preimplantation lethality of homozygous embryos and preferential transmission of the mutant allele. Interestingly, RT-PCR analysis on RNA from endocrine tissues from Pcsk1(+/-) mice revealed the presence of aberrant transcripts specifying the N-terminal half of the PC1 propeptide fused to neo gene product. Mass spectrometric profiles of proopiomelanocortin-derived peptides in the anterior pituitary were similar between Pcsk1(+/-) and Pcsk1(+/+) mice, but significantly different between male and female mice of the same genotype. Relative to their wild-type counterparts, female mutant mice exhibited stunted growth under a low fat diet, and catch-up growth under a high-fat diet. The complex phenotype exhibited by this Pcsk1 mutant mouse model may be due to PC1 deficiency aggravated by expression of aberrant gene products from the mutant allele.
Collapse
Affiliation(s)
- Majambu Mbikay
- Ottawa Health Research Institute and The Ottawa Hospital, 725 Parkdale Avenue, Ottawa, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|
46
|
Waha A, Koch A, Hartmann W, Milde U, Felsberg J, Hübner A, Mikeska T, Goodyer CG, Sörensen N, Lindberg I, Wiestler OD, Pietsch T, Waha A. SGNE1/7B2 is epigenetically altered and transcriptionally downregulated in human medulloblastomas. Oncogene 2007; 26:5662-8. [PMID: 17334394 DOI: 10.1038/sj.onc.1210338] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In a genome-wide screen using differential methylation hybridization (DMH), we have identified a CpG island within the 5' region and untranslated first exon of the secretory granule neuroendocrine protein 1 gene (SGNE1/7B2) that showed hypermethylation in medulloblastomas compared to fetal cerebellum. Bisulfite sequencing and combined bisulfite restriction assay were performed to confirm the methylation status of this CpG island in primary medulloblastomas and medulloblastoma cell lines. Hypermethylation was detected in 16/23 (70%) biopsies and 7/8 (87%) medulloblastoma cell lines, but not in non-neoplastic fetal (n=8) cerebellum. Expression of SGNE1 was investigated by semi-quantitative competitive reverse transcription-polymerase chain reaction and found to be significantly downregulated or absent in all, but one primary medulloblastomas and all cell lines compared to fetal cerebellum. After treatment of medulloblastoma cell lines with 5-aza-2'-deoxycytidine, transcription of SGNE1 was restored. No mutation was found in the coding region of SGNE1 by single-strand conformation polymorphism analysis. Reintroduction of SGNE1 into the medulloblastoma cell line D283Med led to a significant growth suppression and reduced colony formation. In summary, we have identified SGNE1 as a novel epigenetically silenced gene in medulloblastomas. Its frequent inactivation, as well as its inhibitory effect on tumor cell proliferation and focus formation strongly argues for a significant role in medulloblastoma development.
Collapse
Affiliation(s)
- A Waha
- Department of Neuropathology, University of Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jenks BG, Kidane AH, Scheenen WJJM, Roubos EW. Plasticity in the melanotrope neuroendocrine interface of Xenopus laevis. Neuroendocrinology 2007; 85:177-85. [PMID: 17389778 DOI: 10.1159/000101434] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 02/22/2007] [Indexed: 11/19/2022]
Abstract
Melanotrope cells of the amphibian pituitary pars intermedia produce alpha-melanophore-stimulating hormone (alpha-MSH), a peptide which causes skin darkening during adaptation to a dark background. The secretory activity of the melanotrope of the South African clawed toad Xenopus laevis is regulated by multiple factors, both classical neurotransmitters and neuropeptides from the brain. This review concerns the plasticity displayed in this intermediate lobe neuroendocrine interface during physiological adaptation to the environment. The plasticity includes dramatic morphological plasticity in both pre- and post-synaptic elements of the interface. Inhibitory neurons in the suprachiasmatic nucleus, designated suprachiasmatic melanotrope-inhibiting neurons (SMINs), possess more and larger synapses on the melanotrope cells in white than in black-background adapted animals; in the latter animals the melanotropes are larger and produce more proopiomelanocortin (POMC), the precursor of alpha-MSH. On a white background, pre-synaptic SMIN plasticity is reflected by a higher expression of inhibitory neuropeptide Y (NPY) and is closely associated with postsynaptic melanotrope plasticity, namely a higher expression of the NPY Y1 receptor. Interestingly, melanotrope cells in such animals also display higher expression of the receptors for thyrotropin-releasing hormone (TRH) and urocortin 1, two neuropeptides that stimulate alpha-MSH secretion. Possibly, in white-adapted animals melanotropes are sensitized to neuropeptide stimulation so that, when the toad moves to a black background, they can immediately initiate alpha-MSH secretion to achieve rapid adaptation to the new background condition. The melanotrope cell also produces brain-derived neurotrophic factor (BDNF), which is co-sequestered with alpha-MSH in secretory granules within the cells. The neurotrophin seems to control melanotrope cell plasticity in an autocrine way and we speculate that it may also control presynaptic SMIN plasticity.
Collapse
Affiliation(s)
- Bruce G Jenks
- Department of Cellular Animal Physiology, Radboud University Nijmegen, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
48
|
Tsuji A, Kikuchi Y, Sato Y, Koide S, Yuasa K, Nagahama M, Matsuda Y. A proteomic approach reveals transient association of reticulocalbin-3, a novel member of the CREC family, with the precursor of subtilisin-like proprotein convertase, PACE4. Biochem J 2006; 396:51-9. [PMID: 16433634 PMCID: PMC1449992 DOI: 10.1042/bj20051524] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
SPCs (subtilisin-like proprotein convertases) are a family of seven structurally related serine endoproteases that are involved in the proteolytic activation of proproteins. In an effort to examine the substrate protein for PACE4 (paired basic amino-acid-cleaving enzyme-4), an SPC, a potent protein inhibitor of PACE4, an alpha1-antitrypsin RVRR (Arg-Val-Arg-Arg) variant, was expressed in GH4C1 cells. Ectopic expression of the RVRR variant caused accumulation of the 48 kDa protein in cells. Sequence analysis indicates that the 48 kDa protein is a putative Ca2+-binding protein, RCN-3 (reticulocalbin-3), which had previously been predicted by bioinformatic analysis of cDNA from the human hypothalamus. RCN-3 is a member of the CREC (Cab45/reticulocalbin/ERC45/calumenin) family of multiple EF-hand Ca2+-binding proteins localized to the secretory pathway. The most interesting feature of the RCN-3 sequence is the presence of five Arg-Xaa-Xaa-Arg motifs, which represents the target sequence of SPCs. Biosynthetic studies showed that RCN-3 is transiently associated with proPACE4, but not with mature PACE4. Inhibition of PACE4 maturation by a Ca2+ ionophore resulted in accumulation of the proPACE4-RCN-3 complex in cells. Furthermore, autoactivation and secretion of PACE4 was increased upon co-expression with RCN-3. Our findings suggest that selective and transient association of RCN-3 with the precursor of PACE4 plays an important role in the biosynthesis of PACE4.
Collapse
Affiliation(s)
- Akihiko Tsuji
- Department of Biological Science and Technology, Faculty of Engineering, University of Tokushima, 2-1 Minamijosanjima, Tokushima, 770-8506, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Collin RWJ, Martens GJM. The coding sequence of amyloid-beta precursor protein APP contains a neural-specific promoter element. Brain Res 2006; 1087:41-51. [PMID: 16626649 DOI: 10.1016/j.brainres.2006.02.101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2005] [Revised: 01/23/2006] [Accepted: 02/26/2006] [Indexed: 10/24/2022]
Abstract
The amyloid-beta precursor protein APP is generally accepted to be involved in the pathology of Alzheimer's disease. Since its physiological role is still unclear, we decided to study the function of APP via stable transgenesis in the amphibian Xenopus laevis. However, the application of constructs encoding (mutant) APP fused to the C-terminus of the green fluorescent protein GFP (GFP-APP), and harboring a tissue-specific or an inducible gene promoter did not result in transgene expression of APP in neuronal and neuroendocrine cells. Surprisingly, a construct encoding either Xenopus or human APP fused to the N-terminus of GFP (APP-GFP) gave fluorescence throughout the whole brain of the tadpole, despite the fact that a proopiomelanocortin gene promoter was used to target transgene expression specifically to the intermediate pituitary cells. Detailed analysis with deletion mutants revealed the presence of a neural-specific, transcriptionally active DNA element within the 3'-end of the APP-coding sequence that gave rise to an aberrant transcript and protein in the APP-GFP transgenic animals. The DNA element appears to prevent proper APP transgene expression in Xenopus neuronal and neuroendocrine cells. Thus, the coding sequences of Xenopus and human APP contain a neural-specific promoter element, the physiological significance of which is at present unclear.
Collapse
Affiliation(s)
- Rob W J Collin
- Department of Molecular Animal Physiology, Nijmegen Center for Molecular Life Sciences, NCMLS, and Institute for Neuroscience, Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | | |
Collapse
|
50
|
van Rosmalen JWG, Martens GJM. Cell type-specific transgene expression of the prion protein in Xenopus intermediate pituitary cells. FEBS J 2006; 273:847-62. [PMID: 16441670 DOI: 10.1111/j.1742-4658.2006.05118.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The cellular form of prion protein (PrPC) is anchored to the plasma membrane of the cell and expressed in most tissues, but predominantly in the brain, including in the pituitary gland. Thus far, the biosynthesis of PrPC has been studied only in cultured (transfected) tumour cell lines and not in primary cells. Here, we investigated the intracellular fate of PrPCin vivo by using the neuroendocrine intermediate pituitary melanotrope cells of the South-African claw-toed frog Xenopus laevis as a model system. These cells are involved in background adaptation of the animal and produce high levels of its major secretory cargo proopiomelanocortin (POMC) when the animal is black-adapted. The technique of stable Xenopus transgenesis in combination with the POMC gene promoter was used as a tool to express Xenopus PrPC amino-terminally tagged with the green fluorescent protein (GFP-PrPC) specifically in the melanotrope cells. The GFP-PrPC fusion protein was expressed from stage-25 tadpoles onwards to juvenile frogs, the expression was induced on a black background and the fusion protein was subcellularly located mainly in the Golgi apparatus and at the plasma membrane. Pulse-chase metabolic cell labelling studies revealed that GFP-PrPC was initially synthesized as a 45-kDa protein that was subsequently stepwise glycosylated to 48-, 51-, and eventually 55-kDa forms. Furthermore, we revealed that the mature 55-kDa GFP-PrPC protein was sulfated, anchored to the plasma membrane and cleaved to a 33-kDa product. Despite the high levels of transgene expression, the subcellular structures as well as POMC synthesis and processing, and the secretion of POMC-derived products remained unaffected in the transgenic melanotrope cells. Hence, we studied PrPC in a neuroendocrine cell and in a well-defined physiological context.
Collapse
Affiliation(s)
- Jos W G van Rosmalen
- Department of Molecular Animal Physiology, Nijmegen Center for Molecular Life Sciences and Institute for Neuroscience, Radboud University, Nijmegen, the Netherlands
| | | |
Collapse
|