1
|
Mercurio I, D’Abrosca G, della Valle M, Malgieri G, Fattorusso R, Isernia C, Russo L, Di Gaetano S, Pedone EM, Pirone L, Del Gatto A, Zaccaro L, Alberga D, Saviano M, Mangiatordi GF. Molecular interactions between a diphenyl scaffold and PED/PEA15: Implications for type II diabetes therapeutics targeting PED/PEA15 - Phospholipase D1 interaction. Comput Struct Biotechnol J 2024; 23:2001-2010. [PMID: 38770160 PMCID: PMC11103223 DOI: 10.1016/j.csbj.2024.04.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
In a recent study, we have identified BPH03 as a promising scaffold for the development of compounds aimed at modulating the interaction between PED/PEA15 (Phosphoprotein Enriched in Diabetes/Phosphoprotein Enriched in Astrocytes 15) and PLD1 (phospholipase D1), with potential applications in type II diabetes therapy. PED/PEA15 is known to be overexpressed in certain forms of diabetes, where it binds to PLD1, thereby reducing insulin-stimulated glucose transport. The inhibition of this interaction reestablishes basal glucose transport, indicating PED as a potential target of ligands capable to recover glucose tolerance and insulin sensitivity. In this study, we employ computational methods to provide a detailed description of BPH03 interaction with PED, evidencing the presence of a hidden druggable pocket within its PLD1 binding surface. We also elucidate the conformational changes that occur during PED interaction with BPH03. Moreover, we report new NMR data supporting the in-silico findings and indicating that BPH03 disrupts the PED/PLD1 interface displacing PLD1 from its interaction with PED. Our study represents a significant advancement toward the development of potential therapeutics for the treatment of type II diabetes.
Collapse
Affiliation(s)
- Ivan Mercurio
- Institute of Crystallography, CNR, Via Amendola 122/o, 70126 Bari, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Gianluca D’Abrosca
- Department of Clinical and Experimental Medicine, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy
- Institute of Crystallography, CNR, Via Vivaldi 43, 81100, Caserta, Italy
| | - Maria della Valle
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Gaetano Malgieri
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Roberto Fattorusso
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Carla Isernia
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Sonia Di Gaetano
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino 111, 80131 Naples, Italy
| | - Emilia Maria Pedone
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino 111, 80131 Naples, Italy
| | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino 111, 80131 Naples, Italy
| | - Annarita Del Gatto
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino 111, 80131 Naples, Italy
| | - Laura Zaccaro
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino 111, 80131 Naples, Italy
| | - Domenico Alberga
- Institute of Crystallography, CNR, Via Amendola 122/o, 70126 Bari, Italy
| | - Michele Saviano
- Institute of Crystallography, CNR, Via Vivaldi 43, 81100, Caserta, Italy
| | | |
Collapse
|
2
|
Jia D, Li S, Jiang M, Lv Z, Wang H, Zheng Z. Facile Reactive Oxygen Species-Scavenging Supramolecular Hydrogel to Promote Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15752-15760. [PMID: 38507518 DOI: 10.1021/acsami.3c17667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Chronic wound healing impairment is a significant complication in diabetes. Hydrogels that maintain wound moisture and enable sustained drug release have become prominent for enhancing chronic wound care. Particularly, hydrogels that respond to reactive oxygen species (ROS) are sought-after for their dual capacity to mitigate ROS and facilitate controlled drug delivery at the wound site. We have strategically designed an ROS-responsive and scavenging supramolecular hydrogel composed of the simple hexapeptide Glu-Phe-Met-Phe-Met-Glu (EFM). This hydrogelator, composed solely of canonical amino acids without additional ROS-sensitive motifs, forms a hydrogel rapidly upon sonication. Interaction with ROS leads to the oxidation of Met residues to methionine sulfoxide, triggering hydrogel disassembly and consequent payload release. Cellular assays have verified their biocompatibility and efficacy in promoting cell proliferation and migration. In vivo investigations underscore the potential of this straightforward hydrogel as an ROS-scavenger and drug delivery vehicle, enhancing wound healing in diabetic mice. The simplicity and effectiveness of this hydrogel suggest its broader biomedical applications in the future.
Collapse
Affiliation(s)
- Deying Jia
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Shuangshuang Li
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Mengmeng Jiang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zongyu Lv
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Haipeng Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhen Zheng
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
3
|
Jiang R, Zhang H, Liu Q, Yang X, He L, Yuan L, Cheng D. De Novo Design of Near-Infrared Fluorescent Agents Activated by Peroxynitrite and Glutathione-Responsive Imaging for Diabetic Liver Disease. Adv Healthc Mater 2024; 13:e2302466. [PMID: 37840532 DOI: 10.1002/adhm.202302466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Diabetes and its complications, such as diabetes liver disease, is a major problem puzzling people's health. The detection of redox states in its pathological process can effectively help us gain a deeper understanding of the disease. The pair of oxidation-reduction substances peroxynitrite (ONOO- ) and glutathione (GSH) is considered to be closely related to their occurrence and development. Thus, direct visualization of ONOO- and GSH in diabetes liver disease is critical to evaluate the disease at the molecular level. Herein, two activatable agents NTCF-ONOO- and NTCF-GSH are prepared for selectively detecting ONOO- and GSH through protection and deprotection strategies based on hydroxyl and amino groups of near-infrared fluorophore. Fluorescence imaging of exogenous and endogenous ONOO- and GSH changes in living cells and in vivo is observed. The ONOO- and GSH level in the diabetes liver disease cellular model are visualized and the possible redox imbalance mechanism related to the oxidized (NAD+ ) and reduced (NADH) nicotinamide adenine dinucleotides is explored in this process. Moreover, these probes can sensitively recognize ONOO- and GSH in the process of oxidative stress resulting from streptozotocin and streptozotocin/acetaminophen-induced complex diabetic liver disease in vivo. In addition, they can be applied for monitoring the clinical serum sample related with diabetic patients.
Collapse
Affiliation(s)
- Renfeng Jiang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Hongshuai Zhang
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Qian Liu
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Xuefeng Yang
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Longwei He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Dan Cheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
4
|
Viggiano D. Mechanisms of Diabetic Nephropathy Not Mediated by Hyperglycemia. J Clin Med 2023; 12:6848. [PMID: 37959313 PMCID: PMC10650633 DOI: 10.3390/jcm12216848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Diabetes mellitus (DM) is characterized by the appearance of progressive kidney damage, which may progress to end-stage kidney disease. The control of hyperglycemia is usually not sufficient to halt this progression. The kidney damage is quantitatively and qualitatively different in the two forms of diabetes; the typical nodular fibrosis (Kimmelstiel Wilson nodules) appears mostly in type 1 DM, whereas glomerulomegaly is primarily present in type 2 obese DM. An analysis of the different metabolites and hormones in type 1 and type 2 DM and their differential pharmacological treatments might be helpful to advance the hypotheses on the different histopathological patterns of the kidneys and their responses to sodium/glucose transporter type 2 inhibitors (SGLT2i).
Collapse
Affiliation(s)
- Davide Viggiano
- Department of Translational Medical Sciences, University of Campania, 80131 Naples, Italy
| |
Collapse
|
5
|
Zhan X, Cheng L, Huo N, Yu L, Liu C, Liu T, Li G, Fu H. Sodium–glucose cotransporter-2 inhibitor alleviated atrial remodeling in STZ-induced diabetic rats by targeting TLR4 pathway. Front Cardiovasc Med 2022; 9:908037. [PMID: 36148071 PMCID: PMC9485554 DOI: 10.3389/fcvm.2022.908037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose The mechanism of sodium–glucose cotransporter-2 inhibitor (SGLT-2i) reducing the incidence of atrial fibrillation remains unclear. We hypothesize that sodium–glucose cotransporter-2 inhibitor alleviated atrial remodeling in STZ-induced diabetic rats by targeting TLR4 pathway. Methods A total of 42 rats were randomly assigned into three groups: control group (CON group); diabetes group (DM group): diabetes mellitus rats were established by 65 mg/kg streptozotocin (STZ) intraperitoneal injection; and diabetes + dapagliflozin group (DM + DAPA group): diabetic rats were given DAPA gavage administration (DAPA 2mg/kg/d for 4 weeks by gavage administration), 14 rats in each group. Epicardial multiple-lead recording and intracardiac electrophysiology studies were performed to investigate the electrical remodeling in the heart and the atrial fibrillation inducibility in each group. Western blot analysis and real-time PCR were used to determine the protein and mRNA expression of toll-like receptor 4 (TLR4), interleukin receptor-associated kinase 1 (IRAK1), tumor necrosis factor receptor-associated factor 6 (TRAF6), nuclear factor-kappa B (NF-κB), and type I collagen (collagen I). Results Compared with rats in CON group, rats in DM group showed marked myocardial fibrosis, ectopic pacing excitement, reduced conduction velocity, decreased cardiac function. TLR4/IRAK1/TRAF6/NF-κB, collagen I proteins expressions and incidence of atrial fibrillation (27.3%) were increased in DM group. Parts of these changes were reversed by treatment of DAPA. Incidence of atrial fibrillation was decreased in DM + DAPA group (2.8%). Conclusions SGLT-2i dapagliflozin may prevent diabetic rats' atrial remodeling and reduce the inducibility of atrial fibrillation partly by targeting TLR4/IRAK1/TRAF6/NF-κB inflammatory pathway.
Collapse
|
6
|
Abstract
Diabetes has become one of the most prevalent endocrine and metabolic diseases that threaten human health, and it is accompanied by serious complications. Therefore, it is vital and pressing to develop novel strategies or tools for prewarning and therapy of diabetes and its complications. Fluorescent probes have been widely applied in the detection of diabetes due to the fact of their attractive advantages. In this report, we comprehensively summarize the recent progress and development of fluorescent probes in detecting the changes in the various biomolecules in diabetes and its complications. We also discuss the design of fluorescent probes for monitoring diabetes in detail. We expect this review will provide new ideas for the development of fluorescent probes suitable for the prewarning and therapy of diabetes in future clinical transformation and application.
Collapse
|
7
|
Farina B, Pirone L, D’Abrosca G, Della Valle M, Russo L, Isernia C, Sassano M, Del Gatto A, Di Gaetano S, Zaccaro L, Malgieri G, Pedone EM, Fattorusso R. Screening a Molecular Fragment Library to Modulate the PED/PEA15-Phospholipase D1 Interaction in Cellular Lysate Environments. ACS Chem Biol 2021; 16:2798-2807. [PMID: 34825823 DOI: 10.1021/acschembio.1c00688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The overexpression of PED/PEA15, the phosphoprotein enriched in diabetes/phosphoprotein enriched in the astrocytes 15 protein (here referred simply to as PED), observed in some forms of type II diabetes, reduces the transport of insulin-stimulated glucose by binding to the phospholipase D1 (PLD1). The inhibition of the PED/PLD1 interaction was shown to restore basal glucose transport, indicating PED as a pharmacological target for the development of drugs capable of improving insulin sensitivity and glucose tolerance. We here report the identification and selection of PED ligands by means of NMR screening of a library of small organic molecules, NMR characterization of the PED/PLD1 interaction in lysates of cells expressing PLD1, and modulation of such interactions using BPH03, the best selected ligand. Overall, we complement the available literature data by providing detailed information on the structural determinants of the PED/PLD1 interaction in a cellular lysate environment and indicate BPH03 as a precious scaffold for the development of novel compounds that are able to modulate such interactions with possible therapeutic applications in type II diabetes.
Collapse
Affiliation(s)
- Biancamaria Farina
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | - Luciano Pirone
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | - Gianluca D’Abrosca
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Maria Della Valle
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Luigi Russo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Carla Isernia
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Marica Sassano
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Annarita Del Gatto
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | - Sonia Di Gaetano
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | - Laura Zaccaro
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | - Gaetano Malgieri
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Emilia M. Pedone
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | - Roberto Fattorusso
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania─L. Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| |
Collapse
|
8
|
Verbrugge SAJ, Alhusen JA, Kempin S, Pillon NJ, Rozman J, Wackerhage H, Kleinert M. Genes controlling skeletal muscle glucose uptake and their regulation by endurance and resistance exercise. J Cell Biochem 2021; 123:202-214. [PMID: 34812516 DOI: 10.1002/jcb.30179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/27/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022]
Abstract
Exercise improves the insulin sensitivity of glucose uptake in skeletal muscle. Due to that, exercise has become a cornerstone treatment for type 2 diabetes mellitus (T2DM). The mechanisms by which exercise improves skeletal muscle insulin sensitivity are, however, incompletely understood. We conducted a systematic review to identify all genes whose gain or loss of function alters skeletal muscle glucose uptake. We subsequently cross-referenced these genes with recently generated data sets on exercise-induced gene expression and signaling. Our search revealed 176 muscle glucose-uptake genes, meaning that their genetic manipulation altered glucose uptake in skeletal muscle. Notably, exercise regulates the expression or phosphorylation of more than 50% of the glucose-uptake genes or their protein products. This included many genes that previously have not been associated with exercise-induced insulin sensitivity. Interestingly, endurance and resistance exercise triggered some common but mostly unique changes in expression and phosphorylation of glucose-uptake genes or their protein products. Collectively, our work provides a resource of potentially new molecular effectors that play a role in the incompletely understood regulation of muscle insulin sensitivity by exercise.
Collapse
Affiliation(s)
- Sander A J Verbrugge
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Neuherberg, Germany.,Exercise Biology Group, Department for Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Julia A Alhusen
- Molecular Endocrinology, Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum Munich, Helmholtz Diabetes Center (HMGU), Munich, Germany
| | - Shimon Kempin
- Exercise Biology Group, Department for Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Nicolas J Pillon
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jan Rozman
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Henning Wackerhage
- Exercise Biology Group, Department for Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Maximilian Kleinert
- Muscle Physiology and Metabolism Group, German Institute of Human Nutrition, Potsdam - Rehbrücke, Nuthetal, Germany.,Department of Nutrition, Exercise and Sports, Faculty of Science, Section of Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Verschoor PJ, Greig FH, Rochford JJ, Levate G, Delibegovic M, Thompson D, Leeson-Payne A, Dekeryte R, Banks R, Ramos JW, Nixon GF. Phosphoprotein enriched in astrocytes (PEA)-15 is a novel regulator of adipose tissue expansion. Sci Rep 2021; 11:6949. [PMID: 33772049 PMCID: PMC7997924 DOI: 10.1038/s41598-021-86250-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
Excessive expansion of adipose tissue in obesity typically leads to overflow and accumulation of lipids in other tissues, causing fatty liver disease and atherosclerosis. The intracellular protein, phosphoprotein enriched in astrocytes (PEA)-15 has been linked to metabolic disease but its role in lipid storage has not been examined. To delineate the role of PEA-15 in adipose tissue, we placed PEA-15−/− mice on a high fat diet. These mice developed increased body weight and greater white adipose tissue expansion compared to high fat diet-fed wild type mice. This was due to increased adipocyte cell size in PEA-15−/− mice consistent with greater lipid storage capacity. Surprisingly, PEA-15−/− mice exhibited improvements in whole body insulin sensitivity, lower hepatic weight and decreased serum triglycerides indicating a protective phenotype. To determine effects on atherosclerosis, PEA-15−/− mice were crossed with the ApoE−/− mice on a high fat diet. Strikingly, these mice were protected from atherosclerosis and had less hepatic lipid accumulation despite increased adiposity. Therefore, we reveal for the first time that PEA-15 plays a novel role in regulating the expansion of adipose tissue. Decreasing PEA-15 expression increases the sequestering of lipids in adipose tissue, protecting other tissues in obesity, thereby improving metabolic health.
Collapse
Affiliation(s)
- Pola J Verschoor
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Fiona H Greig
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Justin J Rochford
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.,Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Giovanni Levate
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Dawn Thompson
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | | | - Ruta Dekeryte
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Ruth Banks
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Joe W Ramos
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA
| | - Graeme F Nixon
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
10
|
Wang WX, Jiang WL, Mao GJ, Tan M, Fei J, Li Y, Li CY. Monitoring the Fluctuation of Hydrogen Peroxide in Diabetes and Its Complications with a Novel Near-Infrared Fluorescent Probe. Anal Chem 2021; 93:3301-3307. [DOI: 10.1021/acs.analchem.0c05364] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Wen-Xin Wang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Wen-Li Jiang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Guo-Jiang Mao
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, PR China
| | - Min Tan
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Junjie Fei
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Yongfei Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
- College of Chemical Engineering, Xiangtan University, Xiangtan 411105, PR China
| | - Chun-Yan Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| |
Collapse
|
11
|
Auclair N, Sané AT, Delvin E, Spahis S, Levy E. Phospholipase D as a Potential Modulator of Metabolic Syndrome: Impact of Functional Foods. Antioxid Redox Signal 2021; 34:252-278. [PMID: 32586106 DOI: 10.1089/ars.2020.8081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Cardiometabolic disorders (CMD) are composed of a plethora of metabolic dysfunctions such as dyslipidemia, nonalcoholic fatty liver disease, insulin resistance, and hypertension. The development of these disorders is highly linked to inflammation and oxidative stress (OxS), two metabolic states closely related to physiological and pathological conditions. Given the drastically rising CMD prevalence, the discovery of new therapeutic targets/novel nutritional approaches is of utmost importance. Recent Advances: The tremendous progress in methods/technologies and animal modeling has allowed the clarification of phospholipase D (PLD) critical roles in multiple cellular processes, whether directly or indirectly via phosphatidic acid, the lipid product mediating signaling functions. In view of its multiple features and implications in various diseases, PLD has emerged as a drug target. Critical Issues: Although insulin stimulates PLD activity and, in turn, PLD regulates insulin signaling, the impact of the two important PLD isoforms on the metabolic syndrome components remains vague. Therefore, after outlining PLD1/PLD2 characteristics and functions, their role in inflammation, OxS, and CMD has been analyzed and critically reported in the present exhaustive review. The influence of functional foods and nutrients in the regulation of PLD has also been examined. Future Directions: Available evidence supports the implication of PLD in CMD, but only few studies emphasize its mechanisms of action and specific regulation by nutraceutical compounds. Therefore, additional investigations are first needed to clarify the functional role of nutraceutics and, second, to elucidate whether targeting PLDs with food compounds represents an appropriate therapeutic strategy to treat CMD. Antioxid. Redox Signal. 34, 252-278.
Collapse
Affiliation(s)
- Nickolas Auclair
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology & Physiology and Université de Montréal, Montreal, Quebec, Canada
| | - Alain T Sané
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Edgard Delvin
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Schohraya Spahis
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology & Physiology and Université de Montréal, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Validation of mouse phosphoprotein enriched in astrocyte 15 (mPEA15) expressing transgenic pig as a potential model in diabetes translational research. 3 Biotech 2020; 10:34. [PMID: 31988828 DOI: 10.1007/s13205-019-2021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/18/2019] [Indexed: 10/25/2022] Open
Abstract
The present study aimed to investigate the characteristics of mPEA15 expressing transgenic pig (TG pig) as a potential model for diabetes. Expression analysis confirmed the ubiquitous expression of mPEA15 in TG pigs at F4. Oral glucose tolerance test results showed that restoration of normal glucose levels was significantly delayed in the TG pigs when compared with that in the wild-type pigs (WT pigs). Primary skeletal muscle cells isolated from TG pigs demonstrated reduced glucose uptake and reduced GLUT4 translocation to the plasma membrane in response to insulin treatment. Combined, these results suggest that mPEA15 expressing pigs has a glucose intolerance and insulin resistance which are known to mediate the pathophysiology of type 2 diabetes mellitus. Thus, mPEA15 transgenic pigs would serve as a promising model for diabetes translational research.
Collapse
|
13
|
Baig MH, Kausar MA, Husain FM, Shakil S, Ahmad I, Yadav BS, Saeed M. Interfering PLD1-PED/PEA15 interaction using self-inhibitory peptides: An in silico study to discover novel therapeutic candidates against type 2 diabetes. Saudi J Biol Sci 2019; 26:160-164. [PMID: 30622421 PMCID: PMC6319087 DOI: 10.1016/j.sjbs.2018.08.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 11/25/2022] Open
Abstract
Diabetes type 2 (T2D) is a very complex disorder with a large number of cases reported worldwide. There are several reported molecular targets which are being used towards drug design. In spite of extensive research efforts, there is no sure shot treatment available. One of the major reasons for this failure or restricted success in T2D research is the identification of a major/breakthrough therapeutic target responsible for the progression of T2D. It has been well documented that one of the major causes mediating the insulin resistance is the interaction of PLD1 with PED/PEA15. Herein, we have performed in silico experiments to investigate the interaction between PLD1 with PED/PEA15. Furthermore, this study has explored pertinent molecular interactions involving the self-derived peptides. The peptides identified in this study are found to be capable of restricting the interaction of these two proteins. Accordingly, the study suggests that the “self-derived peptides” could be used as promising therapeutic candidate(s) against T2D.
Collapse
Affiliation(s)
- Mohammad Hassan Baig
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Corresponding author.
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Saudi Arabia
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, Saudi Arabia
| | - Shazi Shakil
- Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Research center for advanced materials science, King Khalid university, Abha, Saudi Arabia
| | - Brijesh S. Yadav
- Department of Bioengineering, University of Information Science and Technology, The Former Yugolav Republic of Macedonia
| | - Mohd Saeed
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Saudi Arabia
| |
Collapse
|
14
|
Vastolo V, Nettore IC, Ciccarelli M, Albano L, Raciti GA, Longo M, Beguinot F, Ungaro P. High-fat diet unveils an enhancer element at the Ped/Pea-15 gene responsible for epigenetic memory in skeletal muscle. Metabolism 2018; 87:70-79. [PMID: 29928894 DOI: 10.1016/j.metabol.2018.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/09/2018] [Accepted: 06/17/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The impact of nutrition on the evolution towards type 2 diabetes has recently received increasing attention because of the effect on chromatin structure and gene expression. PURPOSE Evaluate the effect of high-fat diet on chromatin remodelling and expression of Ped/Pea-15, a gene commonly overexpressed in individuals at risk of type 2 diabetes. METHODS We used mouse and cell models to investigate Ped/Pea-15 transcriptional regulation by high-fat diet and glucose, respectively. Chromatin structure and histone modification marks were assessed by Micrococcal Nuclease Protection and Chromatin Immunoprecipitation assays. RESULTS Sixteen-week exposure of C57BL/6J mice to a high-fat diet impaired glucose tolerance and enhanced Ped/Pea-15 expression in their skeletal muscle tissue. This effect was associated with increased chromatin accessibility at specific regulatory sites at the Ped/Pea-15 gene. In particular, the region at -1900 to -1300 bp from Ped/Pea-15 transcription start site was revealed to feature enhancer activity as demonstrated by its function in the luciferase assay, increased p300 recruitment and H3K4me1 and H3K27Ac levels, all marks of functionally active enhancers. Returning mice to a standard chow diet was accompanied by rapid loss of acetylation of K27 on histone H3 and p300 recruitment at Ped/Pea-15. In contrast, the increased H3K4me1, which accompanied the high-fat diet exposure, remained stable. Incubation of muscle cells in culture medium supplemented with 25 mM glucose (HG) increased Ped/Pea-15 mRNA expression and H3K4me1 at the enhancer region. These effects became measurable upon 72 h of exposure to the HG medium and were not rescued upon returning the cells to the 5 mM glucose-containing medium. Interestingly, after 25 mM and sequential 5 mM glucose treatments, re-exposure of the same cells to HG medium further enhanced Ped/Pea-15 expression and increased H3K4me1 above the levels induced by the initial HG challenge already upon 24 h. CONCLUSION Transient exposure to HFD or HG unveiled the presence of an enhancer element at the Ped/Pea-15 gene. Epigenetic changes imposed at this region by diets, which impair glucose tolerance generate metabolic memory of the nutritional injury and leave Ped/Pea-15 induction in a poised state.
Collapse
Affiliation(s)
- Viviana Vastolo
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy
| | - Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy
| | - Marco Ciccarelli
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy
| | - Luigi Albano
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy
| | - Gregory Alexander Raciti
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy
| | - Michele Longo
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy
| | - Francesco Beguinot
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy.
| | - Paola Ungaro
- URT Genomics of Diabetes-IEOS, CNR/Department of Translational Medicine, "Federico II" University Medical School of Napoli, via Sergio Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
15
|
Liotti A, Cabaro S, Cimmino I, Ricci S, Procaccini C, Paciello O, Raciti GA, Spinelli R, Iossa S, Matarese G, Miele C, Formisano P, Beguinot F, Oriente F. Prep1 deficiency improves metabolic response in white adipose tissue. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:515-525. [PMID: 29474930 DOI: 10.1016/j.bbalip.2018.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 02/09/2018] [Accepted: 02/17/2018] [Indexed: 12/13/2022]
Abstract
Prep1 is a gene encoding for a homeodomain transcription factor which induces hepatic and muscular insulin resistance. In this study, we show that Prep1 hypomorphic heterozygous (Prep1i/+) mice, expressing low levels of protein, featured a 23% and a 25% reduction of total body lipid content and epididymal fat, respectively. The percentage of the small adipocytes (25-75 μm) was 30% higher in Prep1i/+ animals than in the WT, with a reciprocal difference in the large adipose cells (100-150 and >150 μm). Insulin-stimulated insulin receptor tyrosine and Akt serine phosphorylation markedly increased in Prep1i/+ mice, paralleled by 3-fold higher glucose uptake and a significant increase of proadipogenic genes such as C/EBPα, GLUT4, and FABP4. Moreover, T cells infiltration and TNF-α, IFNγ and leptin expression were reduced in adipose tissue from Prep1i/+ mice, while adiponectin levels were 2-fold higher. Furthermore, Prep1i/+ mature adipocytes released lower amounts of pro-inflammatory cytokines and higher amount of adiponectin compared to WT cells. Incubation of murine liver cell line (NMuLi) with conditioned media (CM) from mature adipocytes of Prep1i/+ mice improved glucose metabolism, while those from WT mice had no effect. Consistent with these data, Prep1 overexpression in 3T3-L1 adipocytes impaired adipogenesis and insulin signaling, and increased proinflammatory cytokine secretion. All these findings suggest that Prep1 silencing reduces inflammatory response and increases insulin sensitivity in adipose tissue. In addition, CM from mature adipocytes of Prep1i/+ mice improve metabolism in hepatic cells.
Collapse
Affiliation(s)
- Antonietta Liotti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Ricci
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Claudio Procaccini
- Laboratory of Immunology, National Council of Research (CNR), Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, Federico II University of Naples, Naples, Italy
| | - Gregory A Raciti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Rosa Spinelli
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Susanna Iossa
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Giuseppe Matarese
- Laboratory of Immunology, National Council of Research (CNR), Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy.
| |
Collapse
|
16
|
Cimmino I, Lorenzo V, Fiory F, Doti N, Ricci S, Cabaro S, Liotti A, Vitagliano L, Longo M, Miele C, Formisano P, Beguinot F, Ruvo M, Oriente F. A peptide antagonist of Prep1-p160 interaction improves ceramide-induced insulin resistance in skeletal muscle cells. Oncotarget 2017; 8:71845-71858. [PMID: 29069751 PMCID: PMC5641094 DOI: 10.18632/oncotarget.18286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022] Open
Abstract
Prep1 is a homeodomain transcription factor belonging to the TALE protein family. Its overexpression affects glucose metabolism in several tissues. In particular, in skeletal muscle tissue the interaction of Prep1 with its cofactor p160 impairs GLUT4 expression and glucose uptake. In this study, we show that ceramides (C2cer), a class of lipids antagonizing insulin signalling, increase the levels of Prep1 and p160 in a dose and time-dependent fashion in L6 cells and induce their association by 80%. We find that C2cer exposure inhibits insulin receptor, IRS1 and Akt phosphorylation and reduces insulin-stimulated glycogen content and glucose uptake by 1.3- and 2.1-fold, respectively. The synthetic Prep1(54-72) peptide, mimicking the Prep1 region involved in the interaction with p160, reduces in vitro Prep1-p160 binding in a dose-dependent way (IC50 = 0.20μM). In C2cer-treated L6 cells, 10μM Prep1(54-72) restores insulin signalling impaired by ceramide treatment. Prep1 overexpressing L6 cells display similar metabolic alterations observed in ceramide-treated L6 cells and the presence of Prep1(54-72) mitigates these events. All these findings suggest that disruption of the Prep1/p160 molecular interaction enhances insulin sensitivity impaired by ceramides in skeletal muscle cells and indicate this complex as an important target for type 2 diabetes.
Collapse
Affiliation(s)
- Ilaria Cimmino
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Virginia Lorenzo
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Francesca Fiory
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Nunzianna Doti
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Serena Ricci
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Antonietta Liotti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Luigi Vitagliano
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Michele Longo
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| |
Collapse
|
17
|
Fiory F, Spinelli R, Raciti GA, Parrillo L, D'esposito V, Formisano P, Miele C, Beguinot F. Targetting PED/PEA-15 for diabetes treatment. Expert Opin Ther Targets 2017; 21:571-581. [PMID: 28395542 DOI: 10.1080/14728222.2017.1317749] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION PED/PEA-15 is an ubiquitously expressed protein, involved in the regulation of proliferation and apoptosis. It is commonly overexpressed in Type 2 Diabetes (T2D) and in different T2D-associated comorbidities, including cancer and certain neurodegenerative disorders. Areas covered: In mice, Ped/Pea-15 overexpression impairs glucose tolerance and, in combination with high fat diets, further promotes insulin resistance and T2D. It also controls β-cell mass, altering caspase-3 activation and the expression of pro- and antiapoptotic genes. These changes are mediated by PED/PEA-15-PLD1 binding. Overexpression of PLD1 D4 domain specifically blocks Ped/Pea-15-PLD1 interaction, reverting the effect of Ped/Pea-15 in vivo. D4α, a D4 N-terminal peptide, is able to displace Ped/Pea-15-PLD1 binding, but features greater stability in vivo compared to the entire D4 peptide. Here, we review early mechanistic studies on PED/PEA-15 relevance in apoptosis before focusing on its role in cancer and T2D. Finally, we describe potential therapeutic opportunities for T2D based on PED/PEA-15 targeting. Expert opinion: T2D is a major problem for public health and economy. Thus, the identification of new molecules with pharmacological activity for T2D represents an urgent need. Further studies with D4α will help to identify smaller pharmacologically active peptides and innovative molecules of potential pharmacological interest for T2D treatment.
Collapse
Affiliation(s)
- Francesca Fiory
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Rosa Spinelli
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Gregory Alexander Raciti
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Luca Parrillo
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Vittoria D'esposito
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Pietro Formisano
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Claudia Miele
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| | - Francesco Beguinot
- a National Council of Research , URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore" , Naples , Italy.,b Department of Translational Medical Sciences , University of Naples "Federico II" , Naples , Italy
| |
Collapse
|
18
|
Ascione F, Vasaturo A, Caserta S, D'Esposito V, Formisano P, Guido S. Comparison between fibroblast wound healing and cell random migration assays in vitro. Exp Cell Res 2016; 347:123-132. [PMID: 27475838 DOI: 10.1016/j.yexcr.2016.07.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 11/29/2022]
Abstract
Cell migration plays a key role in many biological processes, including cancer growth and invasion, embryogenesis, angiogenesis, inflammatory response, and tissue repair. In this work, we compare two well-established experimental approaches for the investigation of cell motility in vitro: the cell random migration (CRM) and the wound healing (WH) assay. In the former, extensive tracking of individual live cells trajectories by time-lapse microscopy and elaborate data processing are used to calculate two intrinsic motility parameters of the cell population under investigation, i.e. the diffusion coefficient and the persistence time. In the WH assay, a scratch is made in a confluent cell monolayer and the closure time of the exposed area is taken as an easy-to-measure, empirical estimate of cell migration. To compare WH and CRM we applied the two assays to investigate the motility of skin fibroblasts isolated from wild type and transgenic mice (TgPED) overexpressing the protein PED/PEA-15, which is highly expressed in patients with type 2 diabetes. Our main result is that the cell motility parameters derived from CRM can be also estimated from a time-resolved analysis of the WH assay, thus showing that the latter is also amenable to a quantitative analysis for the characterization of cell migration. To our knowledge this is the first quantitative comparison of these two widely used techniques.
Collapse
Affiliation(s)
- Flora Ascione
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale (DICMAPI), Università di Napoli Federico II, P.le Tecchio, 80, 80125 Napoli, Italy
| | - Angela Vasaturo
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale (DICMAPI), Università di Napoli Federico II, P.le Tecchio, 80, 80125 Napoli, Italy
| | - Sergio Caserta
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale (DICMAPI), Università di Napoli Federico II, P.le Tecchio, 80, 80125 Napoli, Italy; CEINGE Biotecnologie Avanzate, Via Sergio Pansini, 5, 80131 Naples, Italy; Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), UdR INSTM Napoli Federico II, P.le Tecchio, 80, 80125 Napoli, Italy.
| | - Vittoria D'Esposito
- Dipartimento di Scienze Mediche Traslazionali (DISMET), Università di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali (DISMET), Università di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy; Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Via Pansini 5, 80131 Napoli, Italy
| | - Stefano Guido
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale (DICMAPI), Università di Napoli Federico II, P.le Tecchio, 80, 80125 Napoli, Italy; CEINGE Biotecnologie Avanzate, Via Sergio Pansini, 5, 80131 Naples, Italy; Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), UdR INSTM Napoli Federico II, P.le Tecchio, 80, 80125 Napoli, Italy
| |
Collapse
|
19
|
Perruolo G, Viggiano D, Fiory F, Cassese A, Nigro C, Liotti A, Miele C, Beguinot F, Formisano P. Parkinson-like phenotype in insulin-resistant PED/PEA-15 transgenic mice. Sci Rep 2016; 6:29967. [PMID: 27426254 PMCID: PMC4947959 DOI: 10.1038/srep29967] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 06/28/2016] [Indexed: 12/30/2022] Open
Abstract
Neurological abnormalities, such as Parkinson-like disorders (PlD), are often co-morbidities of Type 2 Diabetic (T2D) patients, although the epidemiological link between these two disorders remains controversial. The PED/PEA-15 protein represents a possible candidate linking T2D and PD, because it is increased in subjects with T2D and is highly expressed in the brain. To test this hypothesis, we have analyzed the neurological and neurochemical phenotype of transgenic mice overexpressing PED/PEA-15 (tgPED). These mice develop impaired glucose tolerance and insulin resistance, accompanied by neurological features resembling PlD: feet clasping, slow and delayed locomotor movements in different behavioral tests in absence of clear cognitive deficits, ataxia or anxiety. Morphological analysis of the brains showed selective modifications of metabolic activity in the striatal region. In the same region, we have observed 26% decrease of dopamine fibers, confirmed by immunohistochemistry and Western Blot for tyrosine hydroxylase. Moreover, they also showed 48% reduction of dopamine levels in the striatum. Thus the tgPED mice may represent a genetic animal model of neurological disease linked to T2D.
Collapse
Affiliation(s)
- Giuseppe Perruolo
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Davide Viggiano
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy.,Dept Medicine and Health Sciences, Univ. Molise, Italy
| | - Francesca Fiory
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Angela Cassese
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy
| | - Cecilia Nigro
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Antonietta Liotti
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Claudia Miele
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Francesco Beguinot
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Pietro Formisano
- URT- Genomica del Diabete, Istituto di Endocrinologia ed Oncologia Sperimentale (IEOS-CNR), Naples, Italy.,Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Naples, Italy
| |
Collapse
|
20
|
Exler RE, Guo X, Chan D, Livne-Bar I, Vicic N, Flanagan JG, Sivak JM. Biomechanical insult switches PEA-15 activity to uncouple its anti-apoptotic function and promote erk mediated tissue remodeling. Exp Cell Res 2016; 340:283-94. [DOI: 10.1016/j.yexcr.2015.11.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/27/2015] [Accepted: 11/21/2015] [Indexed: 11/15/2022]
|
21
|
Raciti GA, Longo M, Parrillo L, Ciccarelli M, Mirra P, Ungaro P, Formisano P, Miele C, Béguinot F. Understanding type 2 diabetes: from genetics to epigenetics. Acta Diabetol 2015; 52:821-7. [PMID: 25841587 DOI: 10.1007/s00592-015-0741-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/14/2015] [Indexed: 12/18/2022]
Abstract
The known genetic variability (common DNA polymorphisms) does not account either for the current epidemics of type 2 diabetes or for the family transmission of this disorder. However, clinical, epidemiological and, more recently, experimental evidence indicates that environmental factors have an extraordinary impact on the natural history of type 2 diabetes. Some of these environmental hits are often shared in family groups and proved to be capable to induce epigenetic changes which alter the function of genes affecting major diabetes traits. Thus, epigenetic mechanisms may explain the environmental origin as well as the familial aggregation of type 2 diabetes much easier than common polymorphisms. In the murine model, exposure of parents to environmental hits known to cause epigenetic changes reprograms insulin sensitivity as well as beta-cell function in the progeny, indicating that certain epigenetic changes can be transgenerationally transmitted. Studies from different laboratories revealed that, in humans, lifestyle intervention modulates the epigenome and reverts environmentally induced epigenetic modifications at specific target genes. Finally, specific human epigenotypes have been identified which predict adiposity and type 2 diabetes with much greater power than any polymorphism so far identified. These epigenotypes can be recognized in easily accessible white cells from peripheral blood, indicating that, in the future, epigenetic profiling may enable effective type 2 diabetes prediction. This review discusses recent evidence from the literature supporting the immediate need for further investigation to uncover the power of epigenetics in the prediction, prevention and treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Gregory Alexander Raciti
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Michele Longo
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Luca Parrillo
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Marco Ciccarelli
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Paola Mirra
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Paola Ungaro
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Claudia Miele
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Francesco Béguinot
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy.
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
22
|
On the Quest of Cellular Functions of PEA-15 and the Therapeutic Opportunities. Pharmaceuticals (Basel) 2015; 8:455-73. [PMID: 26263999 PMCID: PMC4588177 DOI: 10.3390/ph8030455] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/18/2015] [Accepted: 07/24/2015] [Indexed: 02/03/2023] Open
Abstract
Phosphoprotein enriched in astrocytes, 15 KDa (PEA-15), a ubiquitously expressed small protein in all mammals, is known for decades for its potent interactions with various protein partners along distinct biological pathways. Most notable interacting partners of PEA-15 include extracellular signal-regulated kinase 1 and 2 (ERK1/2) in the mitogen activated protein kinase (MAPK) pathway, the Fas-associated death domain (FADD) protein involving in the formation of the death-inducing signaling complex (DISC), and the phospholipase D1 (PLD1) affecting the insulin sensitivity. However, the actual cellular functions of PEA-15 are still mysterious, and the question why this protein is expressed in almost all cell and tissue types remains unanswered. Here we synthesize the most recent structural, biological, and clinical studies on PEA-15 with emphases on its anti-apoptotic, anti-proliferative, and anti-inflammative properties, and propose a converged protective role of PEA-15 that maintains the balance of death and survival in different cell types. Under conditions that this delicate balance is unsustainable, PEA-15 may become pathological and lead to various diseases, including cancers and diabetes. Targeting PEA-15 interactions, or the use of PEA-15 protein as therapeutics, may provide a wider window of opportunities to treat these diseases.
Collapse
|
23
|
Fiory F, Parrillo L, Raciti GA, Zatterale F, Nigro C, Mirra P, Falco R, Ulianich L, Di Jeso B, Formisano P, Miele C, Beguinot F. PED/PEA-15 inhibits hydrogen peroxide-induced apoptosis in Ins-1E pancreatic beta-cells via PLD-1. PLoS One 2014; 9:e113655. [PMID: 25489735 PMCID: PMC4260953 DOI: 10.1371/journal.pone.0113655] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 10/30/2014] [Indexed: 12/20/2022] Open
Abstract
The small scaffold protein PED/PEA-15 is involved in several different physiologic and pathologic processes, such as cell proliferation and survival, diabetes and cancer. PED/PEA-15 exerts an anti-apoptotic function due to its ability to interfere with both extrinsic and intrinsic apoptotic pathways in different cell types. Recent evidence shows that mice overexpressing PED/PEA-15 present larger pancreatic islets and increased beta-cells mass. In the present work we investigated PED/PEA-15 role in hydrogen peroxide-induced apoptosis in Ins-1E beta-cells. In pancreatic islets isolated from TgPED/PEA-15 mice hydrogen peroxide-induced DNA fragmentation was lower compared to WT islets. TUNEL analysis showed that PED/PEA-15 overexpression increases the viability of Ins-1E beta-cells and enhances their resistance to apoptosis induced by hydrogen peroxide exposure. The activity of caspase-3 and the cleavage of PARP-1 were markedly reduced in Ins-1E cells overexpressing PED/PEA-15 (Ins-1EPED/PEA-15). In parallel, we observed a decrease of the mRNA levels of pro-apoptotic genes Bcl-xS and Bad. In contrast, the expression of the anti-apoptotic gene Bcl-xL was enhanced. Accordingly, DNA fragmentation was higher in control cells compared to Ins-1EPED/PEA-15 cells. Interestingly, the preincubation with propranolol, an inhibitor of the pathway of PLD-1, a known interactor of PED/PEA-15, responsible for its deleterious effects on glucose tolerance, abolishes the antiapoptotic effects of PED/PEA-15 overexpression in Ins-1E beta-cells. The same results have been obtained by inhibiting PED/PEA-15 interaction with PLD-1 in Ins-1EPED/PEA-15. These results show that PED/PEA-15 overexpression is sufficient to block hydrogen peroxide-induced apoptosis in Ins-1E cells through a PLD-1 mediated mechanism.
Collapse
Affiliation(s)
- Francesca Fiory
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Luca Parrillo
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Gregory Alexander Raciti
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Federica Zatterale
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Cecilia Nigro
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Paola Mirra
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Roberta Falco
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Luca Ulianich
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Bruno Di Jeso
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali, Università del Salento, Lecce, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Claudia Miele
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
- * E-mail: (CM); (FB)
| | - Francesco Beguinot
- Dipartimento di Scienze Mediche e Traslazionali dell'Università di Napoli “Federico II”, Naples, Italy
- URT dell'Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
- * E-mail: (CM); (FB)
| |
Collapse
|
24
|
Greig FH, Nixon GF. Phosphoprotein enriched in astrocytes (PEA)-15: a potential therapeutic target in multiple disease states. Pharmacol Ther 2014; 143:265-74. [PMID: 24657708 PMCID: PMC4127788 DOI: 10.1016/j.pharmthera.2014.03.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Phosphoprotein enriched in astrocytes-15 (PEA-15) is a cytoplasmic protein that sits at an important junction in intracellular signalling and can regulate diverse cellular processes, such as proliferation and apoptosis, dependent upon stimulation. Regulation of these processes occurs by virtue of the unique interaction of PEA-15 with other signalling proteins. PEA-15 acts as a cytoplasmic tether for the mitogen-activated protein kinases, extracellular signal-regulated kinase 1/2 (ERK1/2) preventing nuclear localisation. In order to release ERK1/2, PEA-15 requires to be phosphorylated via several potential pathways. PEA-15 (and its phosphorylation state) therefore regulates many ERK1/2-dependent processes, including proliferation, via regulating ERK1/2 nuclear translocation. In addition, PEA-15 contains a death effector domain (DED) which allows interaction with other DED-containing proteins. PEA-15 can bind the DED-containing apoptotic adaptor molecule, Fas-associated death domain protein (FADD) which is also dependent on the phosphorylation status of PEA-15. PEA-15 binding of FADD can inhibit apoptosis as bound FADD cannot participate in the assembly of apoptotic signalling complexes. Through these protein–protein interactions, PEA-15-regulated cellular effects have now been investigated in a number of disease-related studies. Changes in PEA-15 expression and regulation have been observed in diabetes mellitus, cancer, neurological disorders and the cardiovascular system. These changes have been suggested to contribute to the pathology related to each of these disease states. As such, new therapeutic targets based around PEA-15 and its associated interactions are now being uncovered and could provide novel avenues for treatment strategies in multiple diseases.
Collapse
Affiliation(s)
- Fiona H Greig
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Graeme F Nixon
- School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
25
|
Oriente F, Cabaro S, Liotti A, Longo M, Parrillo L, Pagano TB, Raciti GA, Penkov D, Paciello O, Miele C, Formisano P, Blasi F, Beguinot F. PREP1 deficiency downregulates hepatic lipogenesis and attenuates steatohepatitis in mice. Diabetologia 2013; 56:2713-22. [PMID: 24052111 DOI: 10.1007/s00125-013-3053-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/26/2013] [Indexed: 01/07/2023]
Abstract
AIMS/HYPOTHESIS The aim of this study was to investigate the function of Prep1 (also known as Pknox1) in hepatic lipogenesis. METHODS The hepatic lipogenesis pathway was evaluated by real-time RT-PCR and Western blot. Biochemical variables were assessed using a clinical chemistry analyser. RESULTS Serum triacylglycerols and liver expression of fatty acid synthase (FAS) were significantly decreased in Prep1 hypomorphic heterozygous (Prep1 (i/+) ) mice compared with their non-hypomorphic littermates. Upstream FAS expression, phosphorylation of protein kinase C (PKC)ζ, liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) increased in Prep1 (i/+) mice, while protein and mRNA levels of the lipid phosphatase inhibitor of PKCζ, SH2-containing inositol 5'-phosphatase 2 (SHIP2), was more than 60% reduced. Consistent with these findings, HepG2 cells transfected with Prep1 cDNA exhibited increased triacylglycerol accumulation and FAS expression, with strongly reduced PKCζ, LKB1, AMPK and ACC phosphorylation. Further experiments revealed the presence of both Prep1 and its major partner Pbx1 at the Ship2 (also known as Inppl1) promoter. PBX-regulating protein 1 (PREP1) and pre-B cell leukaemia transcription factor 1 (PBX1) enhanced Ship2 transcription. The PREP1HR mutant, which is unable to bind PBX1, exhibited no effect on Ship2 function, indicating transcriptional activation of Ship2 by the PREP1/PBX1 complex. Treatment with a methionine- and choline-deficient diet (MCDD) induced steatosis in both Prep1 (i/+) and non-hypomorphic control mice. However, alanine aminotransferase increase, intracellular triacylglycerol content and histological evidence of liver steatosis, inflammation and necrosis were significantly less evident in Prep1 (i/+) mice, indicating that Prep1 silencing protects mice from MCDD-induced steatohepatitis. CONCLUSIONS/INTERPRETATION Our results indicate that Prep1 silencing reduces lipotoxicity by increasing PKCζ/LKB1/AMPK/ACC signalling, while levels of PREP1 expression may determine the risk of steatohepatitis and its progression.
Collapse
Affiliation(s)
- Francesco Oriente
- Department of Translational Medical Sciences, 'Federico II' University of Naples and Institute of Experimental Endocrinology and Oncology, National Council of Research, Via Pansini 5, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Farina B, Doti N, Pirone L, Malgieri G, Pedone EM, Ruvo M, Fattorusso R. Molecular basis of the PED/PEA15 interaction with the C-terminal fragment of phospholipase D1 revealed by NMR spectroscopy. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:1572-80. [PMID: 23608947 DOI: 10.1016/j.bbapap.2013.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 04/02/2013] [Accepted: 04/13/2013] [Indexed: 10/26/2022]
Abstract
PED/PEA15 is a small protein involved in many protein-protein interactions that modulates the function of a number of key cellular effectors involved in major cell functions, including apoptosis, proliferation and glucose metabolism. In particular, PED/PEA15 interacts with the phospholipase D (PLD) isoforms 1 and 2 increasing protein kinase C-α isoform activity and affects both insulin-stimulated glucose transport and glucose-stimulated insulin secretion. The C-terminal portion (residues 712-1074) of PLD1, named D4, is still able to interact with PED/PEA15. In this study we characterized, by means of NMR spectroscopy, the molecular interaction of PED/PEA15 with D4α, a smaller region of D4, encompassing residues 712-818, shown to have the same affinity for PED/PEA15 and to induce the same effects as D4 in PED/PEA15-overexpressing cells. Chemical shift perturbation (CSP) studies allowed to define D4α binding site of PED/PEA15 and to identify a smaller region likely affected by an allosteric effect. Moreover, ELISA-like experiments showed that three 20-mer overlapping synthetic peptides, covering the 762-801 region of D4α, strongly inhibit PED/PEA15-D4α interaction through their binding to PED/PEA15 with KDs in low micromolar range. Finally, molecular details of the interaction of PED/PEA15 with one of the three peptides have been revealed by CSP and saturation transfer difference (STD) analyses.
Collapse
|
27
|
Cassese A, Raciti GA, Fiory F, Nigro C, Ulianich L, Castanò I, D’Esposito V, Terracciano D, Pastore L, Formisano P, Beguinot F, Miele C. Adenoviral gene transfer of PLD1-D4 enhances insulin sensitivity in mice by disrupting phospholipase D1 interaction with PED/PEA-15. PLoS One 2013; 8:e60555. [PMID: 23585839 PMCID: PMC3621763 DOI: 10.1371/journal.pone.0060555] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 02/27/2013] [Indexed: 01/10/2023] Open
Abstract
Over-expression of phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (PED/PEA-15) causes insulin resistance by interacting with the D4 domain of phospholipase D1 (PLD1). Indeed, the disruption of this association restores insulin sensitivity in cultured cells over-expressing PED/PEA-15. Whether the displacement of PLD1 from PED/PEA-15 improves insulin sensitivity in vivo has not been explored yet. In this work we show that treatment with a recombinant adenoviral vector containing the human D4 cDNA (Ad-D4) restores normal glucose homeostasis in transgenic mice overexpressing PED/PEA-15 (Tg ped/pea-15) by improving both insulin sensitivity and secretion. In skeletal muscle of these mice, D4 over-expression inhibited PED/PEA-15-PLD1 interaction, decreased Protein Kinase C alpha activation and restored insulin induced Protein Kinase C zeta activation, leading to amelioration of insulin-dependent glucose uptake. Interestingly, Ad-D4 administration improved insulin sensitivity also in high-fat diet treated obese C57Bl/6 mice. We conclude that PED/PEA-15-PLD1 interaction may represent a novel target for interventions aiming at improving glucose tolerance.
Collapse
Affiliation(s)
- Angela Cassese
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Gregory A. Raciti
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Francesca Fiory
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Cecilia Nigro
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Luca Ulianich
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Ilenia Castanò
- Dipartimento di Biochimica e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Vittoria D’Esposito
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Daniela Terracciano
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Lucio Pastore
- Dipartimento di Biochimica e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Francesco Beguinot
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
- * E-mail: (FB); (CM)
| | - Claudia Miele
- Dipartimento di Scienze Mediche e Traslazionali, Università di Napoli “Federico II” and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
- * E-mail: (FB); (CM)
| |
Collapse
|
28
|
Funke V, Lehmann-Koch J, Bickeböller M, Benner A, Tagscherer KE, Grund K, Pfeifer M, Herpel E, Schirmacher P, Chang-Claude J, Brenner H, Hoffmeister M, Roth W. The PEA-15/PED protein regulates cellular survival and invasiveness in colorectal carcinomas. Cancer Lett 2013; 335:431-40. [PMID: 23481023 DOI: 10.1016/j.canlet.2013.02.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 02/20/2013] [Accepted: 02/27/2013] [Indexed: 11/16/2022]
Abstract
The PEA-15/PED (phosphoprotein enriched in astrocytes 15kD/phosphoprotein enriched in diabetes) protein is a multifunctional phosphoprotein involved in various signaling pathways which determine survival, proliferation, and migration of cancer cells. Here, we investigated the expression and cellular functions of PEA-15 in colorectal carcinoma (CRC). PEA-15 is expressed in the majority of human CRC, predominantly in well differentiated tumor areas. A tissue microarray analysis of 1262 human CRC specimens from the DACHS study showed that PEA-15 expression is significantly associated with a low pT stadium as defined by limited invasion into the bowel wall. Moreover, patients with PEA-15-positive CRC exhibited a significantly longer tumor-specific survival time. To investigate the functional relevance of PEA-15 expression on a cellular level, we over-expressed PEA-15 in several CRC cell lines. Increased expression of PEA-15 resulted in a strong inhibition of clonogenicity, proliferation, and invasiveness of CRC cells. These effects were associated with a PEA-15-dependent down-regulation of integrin αvβ5 as well as with elevated levels of the phosphorylated MAP kinase ERK1/2. Moreover, expression of PEA-15 resulted in significant protection from cell death induced by cytotoxic drugs (5-FU, cisplatin), by the death ligand TRAIL, or by serum withdrawal. In conclusion, the PEA-15 protein regulates invasiveness, proliferation, and apoptosis resistance in CRC cells. PEA-15 might play an important role in chemoresistance, progression and metastasis in CRC.
Collapse
Affiliation(s)
- Verena Funke
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ungaro P, Mirra P, Oriente F, Nigro C, Ciccarelli M, Vastolo V, Longo M, Perruolo G, Spinelli R, Formisano P, Miele C, Beguinot F. Peroxisome proliferator-activated receptor-γ activation enhances insulin-stimulated glucose disposal by reducing ped/pea-15 gene expression in skeletal muscle cells: evidence for involvement of activator protein-1. J Biol Chem 2012; 287:42951-61. [PMID: 23105093 DOI: 10.1074/jbc.m112.406637] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The gene network responsible for inflammation-induced insulin resistance remains enigmatic. In this study, we show that, in L6 cells, rosiglitazone- as well as pioglitazone-dependent activation of peroxisome proliferator-activated receptor-γ (PPARγ) represses transcription of the ped/pea-15 gene, whose increased activity impairs glucose tolerance in mice and humans. Rosiglitazone enhanced insulin-induced glucose uptake in L6 cells expressing the endogenous ped/pea-15 gene but not in cells expressing ped/pea-15 under the control of an exogenous promoter. The ability of PPARγ to affect ped/pea-15 expression was also lost in cells and in C57BL/6J transgenic mice expressing ped/pea-15 under the control of an exogenous promoter, suggesting that ped/pea-15 repression may contribute to rosiglitazone action on glucose disposal. Indeed, high fat diet mice showed insulin resistance and increased ped/pea-15 levels, although these effects were reduced by rosiglitazone treatment. Both supershift and ChIP assays revealed the presence of the AP-1 component c-JUN at the PED/PEA-15 promoter upon 12-O-tetradecanoylphorbol-13-acetate stimulation of the cells. In these experiments, rosiglitazone treatment reduced c-JUN presence at the PED/PEA-15 promoter. This effect was not associated with a decrease in c-JUN expression. In addition, c-jun silencing in L6 cells lowered ped/pea-15 expression and caused nonresponsiveness to rosiglitazone, although c-jun overexpression enhanced the binding to the ped/pea-15 promoter and blocked the rosiglitazone effect. These results indicate that PPARγ regulates ped/pea-15 transcription by inhibiting c-JUN binding at the ped/pea-15 promoter. Thus, ped/pea-15 is downstream of a major PPARγ-regulated inflammatory network. Repression of ped/pea-15 transcription might contribute to the PPARγ regulation of muscle sensitivity to insulin.
Collapse
Affiliation(s)
- Paola Ungaro
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università di Napoli "Federico II", Consiglio Nazionale delle Ricerche, 80131 Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Twomey EC, Cordasco DF, Wei Y. Profound conformational changes of PED/PEA-15 in ERK2 complex revealed by NMR backbone dynamics. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012; 1824:1382-93. [PMID: 22820249 DOI: 10.1016/j.bbapap.2012.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 05/30/2012] [Accepted: 07/05/2012] [Indexed: 01/09/2023]
Abstract
PED/PEA-15 is a small, non-catalytic, DED containing protein that is widely expressed in different tissues and highly conserved among mammals. PED/PEA-15 has been found to interact with several protein targets in various pathways, including FADD and procaspase-8 (apoptosis), ERK1/2 (cell cycle entry), and PLD1/2 (diabetes). In this research, we have studied the PED/PEA-15 in a complex with ERK2, a MAP kinase, using NMR spectroscopic techniques. MAP Kinase signaling pathways are involved in the regulation of many cellular functions, including cell proliferation, differentiation, apoptosis and survival. ERK1/2 are activated by a variety of external stimuli, including growth factors, hormones and neurotransmitters. Inactivated ERK2 is primarily found in the cytosol. Once the ERK/MAPK cascade is initiated, ERK2 is phosphorylated and stimulated, allowing it to redistribute in the cell nucleus and act as a transcription factor. Previous studies have shown that PED/PEA-15 complexes with ERK2 in the cytoplasm and prevents redistribution into the nucleus. Although the NMR structure and dynamics of PED/PEA-15 in the free form have been documented recently, no detailed structural and dynamic information for the ERK2-bound form is available. Here we report NMR chemical shift perturbation and backbone dynamic studies at the fast ps-ns timescale of PED/PEA-15, in its free form and in the complex with ERK2. These analyses characterize motions and conformational changes involved in ERK2 recognition and binding that orchestrate the reorganization of the DED and immobilization of the C-terminal tail. A new induced fit binding model for PED/PEA-15 is proposed.
Collapse
Affiliation(s)
- Edward C Twomey
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, NJ 07079, USA
| | | | | |
Collapse
|
31
|
Buonomo R, Giacco F, Vasaturo A, Caserta S, Guido S, Pagliara V, Garbi C, Mansueto G, Cassese A, Perruolo G, Oriente F, Miele C, Beguinot F, Formisano P. PED/PEA-15 controls fibroblast motility and wound closure by ERK1/2-dependent mechanisms. J Cell Physiol 2012; 227:2106-16. [PMID: 21780113 PMCID: PMC3306794 DOI: 10.1002/jcp.22944] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell migration is dependent on the control of signaling events that play significant roles in creating contractile force and in contributing to wound closure. We evaluated wound closure in fibroblasts from mice overexpressing (TgPED) or lacking ped/pea-15 (KO), a gene overexpressed in patients with type 2 diabetes. Cultured skin fibroblasts isolated from TgPED mice showed a significant reduction in the ability to recolonize wounded area during scratch assay, compared to control fibroblasts. This difference was observed both in the absence and in the presence of mytomicin C, an inhibitor of mitosis. In time-lapse experiments, TgPED fibroblasts displayed about twofold lower velocity and diffusion coefficient, as compared to controls. These changes were accompanied by reduced spreading and decreased formation of stress fibers and focal adhesion plaques. At the molecular level, TgPED fibroblasts displayed decreased RhoA activation and increased abundance of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2). Inhibition of ERK1/2 activity by PD98059 restored RhoA activation, cytoskeleton organization and cell motility, and almost completely rescued wound closure of TgPED fibroblasts. Interestingly, skin fibroblasts isolated from KO mice displayed an increased wound closure ability. In vivo, healing of dorsal wounds was delayed in TgPED and accelerated in KO mice. Thus, PED/PEA-15 may affect fibroblast motility by a mechanism, at least in part, mediated by ERK1/2. J. Cell. Physiol. 227: 2106–2116, 2012. © 2011 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roberta Buonomo
- Department of Cellular and Molecular Biology and Pathology, Federico II University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Panariello F, Perruolo G, Cassese A, Giacco F, Botta G, Barbagallo APM, Muscettola G, Beguinot F, Formisano P, de Bartolomeis A. Clozapine impairs insulin action by up-regulating Akt phosphorylation and Ped/Pea-15 protein abundance. J Cell Physiol 2012; 227:1485-92. [PMID: 21618539 PMCID: PMC3306790 DOI: 10.1002/jcp.22864] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Clinical and experimental evidence indicates that atypical antipsychotics impair glucose metabolism. We investigated whether clozapine may directly affect insulin action by analyzing insulin signaling in vitro and in vivo. Clozapine reduced insulin-stimulated glucose uptake in PC12 and in L6 cells, representative models of neuron and skeletal muscle, respectively. Consistently, clozapine reduced insulin effect on insulin receptor (IR) by 40% and on IR substrate-1 (IRS1) tyrosine phosphorylation by 60%. Insulin-stimulated Akt phosphorylation was also reduced by about 40%. Moreover, insulin-dependent phosphorylation of protein kinase C-ζ (PKC-ζ) was completely blunted in clozapine-treated cells. Interestingly, clozapine treatment was accompanied by an insulin-independent increase of Akt phosphorylation, with no change of IR, IRS1, and PKC-ζ basal phosphorylation. The cellular abundance of Ped/Pea-15, an Akt substrate and inducer of insulin resistance, was also increased following clozapine exposure, both in the absence and in the presence of cyclohexymide, a protein synthesis inhibitor. Similar as in cellular models, in the caudate-putamen and in the tibialis muscle of clozapine-treated C57/BL/KsJ mice, Akt phosphorylation and Ped/Pea-15 protein levels were increased and PKC-ζ phosphorylation was decreased. Thus, in these experimental models, clozapine deranged Akt function and up-regulated Ped/Pea-15, thereby inhibiting insulin stimulation of PKC-ζ and of glucose uptake.
Collapse
Affiliation(s)
- Fabio Panariello
- Dipartimento di Neuroscienze, Sezione di Psichiatria, Laboratorio di Psichiatria Molecolare, University of Napoli Federico II, Napoli, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Santulli G, Lombardi A, Sorriento D, Anastasio A, Del Giudice C, Formisano P, Béguinot F, Trimarco B, Miele C, Iaccarino G. Age-related impairment in insulin release: the essential role of β(2)-adrenergic receptor. Diabetes 2012; 61:692-701. [PMID: 22315324 PMCID: PMC3282797 DOI: 10.2337/db11-1027] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In this study, we investigated the significance of β(2)-adrenergic receptor (β(2)AR) in age-related impaired insulin secretion and glucose homeostasis. We characterized the metabolic phenotype of β(2)AR-null C57Bl/6N mice (β(2)AR(-/-)) by performing in vivo and ex vivo experiments. In vitro assays in cultured INS-1E β-cells were carried out in order to clarify the mechanism by which β(2)AR deficiency affects glucose metabolism. Adult β(2)AR(-/-) mice featured glucose intolerance, and pancreatic islets isolated from these animals displayed impaired glucose-induced insulin release, accompanied by reduced expression of peroxisome proliferator-activated receptor (PPAR)γ, pancreatic duodenal homeobox-1 (PDX-1), and GLUT2. Adenovirus-mediated gene transfer of human β(2)AR rescued these defects. Consistent effects were evoked in vitro both upon β(2)AR knockdown and pharmacologic treatment. Interestingly, with aging, wild-type (β(2)AR(+/+)) littermates developed impaired insulin secretion and glucose tolerance. Moreover, islets from 20-month-old β(2)AR(+/+) mice exhibited reduced density of β(2)AR compared with those from younger animals, paralleled by decreased levels of PPARγ, PDX-1, and GLUT2. Overexpression of β(2)AR in aged mice rescued glucose intolerance and insulin release both in vivo and ex vivo, restoring PPARγ/PDX-1/GLUT2 levels. Our data indicate that reduced β(2)AR expression contributes to the age-related decline of glucose tolerance in mice.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Clinical Medicine, Cardiovascular & Immunologic Sciences, “Federico II” University of Naples, Naples, Italy
- Columbia-Presbyterian Medical Center, College of Physicians & Surgeons, Columbia University, New York, New York
| | - Angela Lombardi
- Columbia University Medical Center, Columbia University, New York, New York
- Department of Cellular and Molecular Biology and Pathology and Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore,” “Federico II” University of Naples, Naples, Italy
| | - Daniela Sorriento
- Department of Clinical Medicine, Cardiovascular & Immunologic Sciences, “Federico II” University of Naples, Naples, Italy
| | - Antonio Anastasio
- Department of Clinical Medicine, Cardiovascular & Immunologic Sciences, “Federico II” University of Naples, Naples, Italy
| | - Carmine Del Giudice
- Department of Clinical Medicine, Cardiovascular & Immunologic Sciences, “Federico II” University of Naples, Naples, Italy
| | - Pietro Formisano
- Department of Cellular and Molecular Biology and Pathology and Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore,” “Federico II” University of Naples, Naples, Italy
| | - Francesco Béguinot
- Department of Cellular and Molecular Biology and Pathology and Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore,” “Federico II” University of Naples, Naples, Italy
| | - Bruno Trimarco
- Department of Clinical Medicine, Cardiovascular & Immunologic Sciences, “Federico II” University of Naples, Naples, Italy
| | - Claudia Miele
- Department of Cellular and Molecular Biology and Pathology and Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore,” “Federico II” University of Naples, Naples, Italy
- Corresponding authors: Guido Iaccarino, , and Claudia Miele,
| | - Guido Iaccarino
- School of Medicine, University of Salerno, Salerno, Italy
- Corresponding authors: Guido Iaccarino, , and Claudia Miele,
| |
Collapse
|
34
|
Iovino S, Oriente F, Botta G, Cabaro S, Iovane V, Paciello O, Viggiano D, Perruolo G, Formisano P, Beguinot F. PED/PEA-15 induces autophagy and mediates TGF-beta1 effect on muscle cell differentiation. Cell Death Differ 2012; 19:1127-38. [PMID: 22281705 PMCID: PMC3374077 DOI: 10.1038/cdd.2011.201] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
TGF-beta1 has been shown to induce autophagy in certain cells but whether and how this action is exerted in muscle and whether this activity relates to TGF-beta1 control of muscle cell differentiation remains unknown. Here, we show that expression of the autophagy-promoting protein phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (PED/PEA-15) progressively declines during L6 and C2C12 skeletal muscle cell differentiation. PED/PEA-15 underwent rapid induction upon TGF-beta1 exposure of L6 and C2C12 myoblasts, accompanied by impaired differentiation into mature myotubes. TGF-beta1 also induced autophagy in the L6 and C2C12 cells through a PP2A/FoxO1-mediated mechanism. Both the TGF-beta1 effect on differentiation and that on autophagy were blocked by specific PED/PEA-15 ShRNAs. Myoblasts stably overexpressing PED/PEA-15 did not differentiate and showed markedly enhanced autophagy. In these same cells, the autophagy inhibitor 3-methyladenine rescued TGF-beta1 effect on both autophagy and myogenesis, indicating that PED/PEA-15 mediates TGF-beta1 effects in muscle. Muscles from transgenic mice overexpressing PED/PEA-15 featured a significant number of atrophic fibers, accompanied by increased light chain 3 (LC3)II to LC3I ratio and reduced PP2A/FoxO1 phosphorylation. Interestingly, these mice showed significantly impaired locomotor activity compared with their non-transgenic littermates. TGF-beta1 causes transcriptional upregulation of the autophagy-promoting gene PED/PEA-15, which in turn is capable to induce atrophic responses in skeletal muscle in vivo.
Collapse
Affiliation(s)
- S Iovino
- Department of Cellular and Molecular Biology and Pathology, University of Naples Federico II, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Choi SG, Ruf-Zamojski F, Pincas H, Roysam B, Sealfon SC. Characterization of a MAPK scaffolding protein logic gate in gonadotropes. Mol Endocrinol 2011; 25:1027-39. [PMID: 21436256 DOI: 10.1210/me.2010-0387] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In the pituitary gonadotropes, both protein kinase C (PKC) and MAPK/ERK signaling cascades are activated by GnRH. Phosphoprotein-enriched in astrocytes 15 (PEA-15) is a cytosolic ERK scaffolding protein, which is expressed in LβT2 gonadotrope cells. Pharmacological inhibition of PKC and small interfering RNA-mediated silencing of Gαq/11 revealed that GnRH induces accumulation of phosphorylated PEA-15 in a PKC-dependent manner. To investigate the potential role of PEA-15 in GnRH signaling, we examined the regulation of ERK subcellular localization and the activation of ribosomal S6 kinase, a substrate of ERK. Results obtained by cellular fractionation/Western blot analysis and immunohistochemistry revealed that GnRH-induced accumulation of phosphorylated ERK in the nucleus was attenuated when PEA-15 expression was reduced. Conversely, in the absence of GnRH stimulation, PEA-15 anchors ERK in the cytosol. Our data suggest that GnRH-induced nuclear translocation of ERK requires its release from PEA-15, which occurs upon PEA-15 phosphorylation by PKC. Additional gene-silencing experiments in GnRH-stimulated cells demonstrated that ribosomal S6 kinase activation was dependent on both PEA-15 and PKC. Furthermore, small interfering RNA-mediated knockdown of PEA-15 caused a reduction in GnRH-stimulated expression of early response genes Egr2 and c-Jun, as well as gonadotropin FSHβ-subunit gene expression. PEA-15 knockdown increased LHβ and common α-glycoprotein subunit mRNAs, suggesting a possible role in differential regulation of gonadotropin subunit gene expression. We propose that PEA-15 represents a novel point of convergence of the PKC and MAPK/ERK pathways under GnRH stimulation. PKC, ERK, and PEA-15 form an AND logic gate that shapes the response of the gonadotrope cell to GnRH.
Collapse
Affiliation(s)
- Soon Gang Choi
- Center for Translational Systems Biology and Department of Neurology, Mount Sinai School of Medicine, Annenberg 14-94, Box 1137, One Gustave L. Levy Place, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
36
|
Oriente F, Iovino S, Cabaro S, Cassese A, Longobardi E, Miele C, Ungaro P, Formisano P, Blasi F, Beguinot F. Prep1 controls insulin glucoregulatory function in liver by transcriptional targeting of SHP1 tyrosine phosphatase. Diabetes 2011; 60:138-47. [PMID: 20864515 PMCID: PMC3012165 DOI: 10.2337/db10-0860] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE We investigated the function of the Prep1 gene in insulin-dependent glucose homeostasis in liver. RESEARCH DESIGN AND METHODS Prep1 action on insulin glucoregulatory function has been analyzed in liver of Prep1-hypomorphic mice (Prep1(i/i)), which express 2-3% of Prep1 mRNA. RESULTS Based on euglycemic hyperinsulinemic clamp studies and measurement of glycogen content, livers from Prep1(i/i) mice feature increased sensitivity to insulin. Tyrosine phosphorylation of both insulin receptor (IR) and insulin receptor substrate (IRS)1/2 was significantly enhanced in Prep1(i/i) livers accompanied by a specific downregulation of the SYP and SHP1 tyrosine phosphatases. Prep1 overexpression in HepG2 liver cells upregulated SYP and SHP1 and inhibited insulin-induced IR and IRS1/2 phosphorylation and was accompanied by reduced glycogen content. Consistently, overexpression of the Prep1 partner Pbx1, but not of p160MBP, mimicked Prep1 effects on tyrosine phosphorylations, glycogen content, and on SYP and SHP1 expression. In Prep1 overexpressing cells, antisense silencing of SHP1, but not that of SYP, rescued insulin-dependent IR phosphorylation and glycogen accumulation. Both Prep1 and Pbx1 bind SHP1 promoter at a site located between nucleotides -2,113 and -1,778. This fragment features enhancer activity and induces luciferase function by 7-, 6-, and 30-fold, respectively, in response to Prep1, Pbx1, or both. CONCLUSIONS SHP1, a known silencer of insulin signal, is a transcriptional target of Prep1. In liver, transcriptional activation of SHP1 gene by Prep1 attenuates insulin signal transduction and reduces glucose storage.
Collapse
Affiliation(s)
- Francesco Oriente
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Salvatore Iovino
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Serena Cabaro
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Angela Cassese
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Elena Longobardi
- Istituto FIRC di Oncologia Molecolare (Fondazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology), Milano, Italy
- Università Vita Salute San Raffaele, Milano, Italy
| | - Claudia Miele
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Paola Ungaro
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Pietro Formisano
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Francesco Blasi
- Istituto FIRC di Oncologia Molecolare (Fondazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology), Milano, Italy
- Università Vita Salute San Raffaele, Milano, Italy
| | - Francesco Beguinot
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Università degli Studi di Napoli Federico II, Naples, Italy
- Corresponding author: Francesco Beguinot,
| |
Collapse
|
37
|
Farina B, Pirone L, Russo L, Viparelli F, Doti N, Pedone C, Pedone EM, Fattorusso R. NMR backbone dynamics studies of human PED/PEA-15 outline protein functional sites. FEBS J 2010; 277:4229-40. [PMID: 20825483 DOI: 10.1111/j.1742-4658.2010.07812.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PED/PEA-15 (phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes) is a ubiquitously expressed protein and a key regulator of cell growth and glucose metabolism. PED/PEA-15 mediates both homotypic and heterotypic interactions and is constituted by an N-terminal canonical death effector domain and a C-terminal tail. In the present study, the backbone dynamics of PED/PEA-15 via (15)N R(1) and R(2) and steady-state [(1)H]-(15)N NOE measurements is reported. The dynamic parameters were analyzed using both Lipari-Szabo model-free formalism and a reduced spectral density mapping approach. The results obtained define a polar and charged surface of the death effector domain characterized by internal motions in the micro- to millisecond timescale, which is crucial for the multiple heterotypic functional protein-protein interactions in which PED/PEA-15 is involved. The present study contributes to a better understanding of the molecular basis of the PED/PEA-15 functional interactions and provides a more detailed surface for the design and development of PED/PEA-15 binders.
Collapse
|
38
|
Doti N, Cassese A, Marasco D, Paturzo F, Sabatella M, Viparelli F, Dathan N, Monti SM, Miele C, Formisano P, Beguinot F, Ruvo M. Residues 762-801 of PLD1 mediate the interaction with PED/PEA15. MOLECULAR BIOSYSTEMS 2010; 6:2039-48. [PMID: 20714510 DOI: 10.1039/c005272h] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The interaction of Phospholipase D1 (PLD1) by its C-terminal domain D4 with PED/PEA15 has been indicated as a target for type 2 diabetes. PED/PEA15 is overexpressed in several tissues of individuals affected by type 2 diabetes and its overexpression in intact cells and in transgenic animal models impairs insulin regulation of glucose transport by a mechanism mediated by the interaction with D4 and the consequent increase of protein kinase C-alpha activity. Expression of D4 or administration of a peptide mimicking the PED/PEA15 region involved in this interaction to cells stably overexpressing PED/PEA15 reduces its interaction with PLD1, thereby lowering PKC-alpha activation and restoring normal glucose transport mediated by PKC-zeta. By using D4 deletion mutants, we have restricted the PLD1 region involved in PED/PEA15 interaction to an N-terminal fragment named D4alpha (residues 712-818). This region binds PED/PEA15 with the same efficacy as D4 (K(D) approximately 0.7 microM) and, when transfected in different PED/PEA15-overexpressing cells, it is able to reduce PKC-alpha activity and to restore the sensitivity of PKC-zeta to insulin stimulation, independently of the PI3K/Akt signalling. We also show that the effective disruption of the PED/PEA15-PLD1 interaction can restore the normal ERK1/2 signalling. Finally, using a set of overlapping peptides that cover the D4alpha region, we have further restricted the shortest PED/PEA15-binding site to a segment encompassing residues 762-801, suggesting that a quite limited binding interface mostly contributes to the interaction and can thus be a selective target for the design of effective antagonists.
Collapse
Affiliation(s)
- Nunzianna Doti
- Istituto di Biostrutture e Bioimmagini, CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ungaro P, Teperino R, Mirra P, Longo M, Ciccarelli M, Raciti GA, Nigro C, Miele C, Formisano P, Beguinot F. Hepatocyte nuclear factor (HNF)-4alpha-driven epigenetic silencing of the human PED gene. Diabetologia 2010; 53:1482-92. [PMID: 20396999 DOI: 10.1007/s00125-010-1732-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 02/25/2010] [Indexed: 01/03/2023]
Abstract
AIMS/HYPOTHESIS Overexpression of PED (also known as PEA15) determines insulin resistance and impaired insulin secretion and may contribute to progression toward type 2 diabetes. Recently, we found that the transcription factor hepatocyte nuclear factor (HNF)-4alpha binds to PED promoter and represses its transcription. However, the molecular details responsible for regulation of PED gene remain unclear. METHODS Here we used gain and loss of function approaches to investigate the hypothesis that HNF-4alpha controls chromatin remodelling at the PED promoter in human cell lines. RESULTS HNF-4alpha production and binding induce chromatin remodelling at the -250 to 50 region of PED, indicating that remodelling is limited to two nucleosomes located at the proximal promoter. Chromatin immunoprecipitation assays also revealed concomitant HNF-4alpha-induced deacetylation of histone H3 at Lys9 and Lys14, and increased dimethylation of histone H3 at Lys9. The latter was followed by reduction of histone H3 Lys4 dimethylation. HNF-4alpha was also shown to target the histone deacetylase complex associated with silencing mediator of retinoic acid and thyroid hormone receptor, both at the PED promoter, and at GRB14 and USP21 regulatory regions, leading to a reduction of mRNA levels. Moreover, HNF-4alpha silencing and PED overexpression were accompanied by a significant reduction of hepatic glycogen content. CONCLUSIONS/INTERPRETATION These results show that HNF-4alpha serves as a scaffold protein for histone deacetylase activities, thereby inhibiting liver expression of genes including PED. Dysregulation of these mechanisms may lead to upregulation of the PED gene in type 2 diabetes.
Collapse
Affiliation(s)
- P Ungaro
- Dipartimento di Biologia e Patologia Cellulare e Molecolare L. Califano & Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Università di Napoli Federico II, Via Sergio Pansini, 5, Naples, 80131, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Watanabe Y, Yamasaki F, Kajiwara Y, Saito T, Nishimoto T, Bartholomeusz C, Ueno NT, Sugiyama K, Kurisu K. Expression of phosphoprotein enriched in astrocytes 15 kDa (PEA-15) in astrocytic tumors: a novel approach of correlating malignancy grade and prognosis. J Neurooncol 2010; 100:449-57. [DOI: 10.1007/s11060-010-0201-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
|
41
|
Oriente F, Iovino S, Cassese A, Romano C, Miele C, Troncone G, Balletta M, Perfetti A, Santulli G, Iaccarino G, Valentino R, Beguinot F, Formisano P. Overproduction of phosphoprotein enriched in diabetes (PED) induces mesangial expansion and upregulates protein kinase C-beta activity and TGF-beta1 expression. Diabetologia 2009; 52:2642-52. [PMID: 19789852 DOI: 10.1007/s00125-009-1528-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 08/05/2009] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS Overproduction of phosphoprotein enriched in diabetes (PED, also known as phosphoprotein enriched in astrocytes-15 [PEA-15]) is a common feature of type 2 diabetes and impairs insulin action in cultured cells and in mice. Nevertheless, the potential role of PED in diabetic complications is still unknown. METHODS We studied the effect of PED overproduction and depletion on kidney function in animal and cellular models. RESULTS Transgenic mice overexpressing PED (PEDTg) featured age-dependent increases of plasma creatinine levels and urinary volume, accompanied by expansion of the mesangial area, compared with wild-type littermates. Serum and kidney levels of TGF-beta1 were also higher in 6- and 9-month-old PEDTg. Overexpression of PED in human kidney 2 cells significantly increased TGF-beta1 levels, SMAD family members (SMAD)2/3 phosphorylation and fibronectin production. Opposite results were obtained following genetic silencing of PED in human kidney 2 cells by antisense oligonucleotides. Inhibition of phospholipase D and protein kinase C-beta by 2-butanol and LY373196 respectively reduced TGF-beta1, SMAD2/3 phosphorylation and fibronectin production. Moreover, inhibition of TGF-beta1 receptor activity and SMAD2/3 production by SB431542 and antisense oligonucleotides respectively reduced fibronectin secretion by about 50%. TGF-beta1 circulating levels were significantly reduced in Ped knockout mice and positively correlated with PED content in peripheral blood leucocytes of type 2 diabetic patients. CONCLUSIONS/INTERPRETATION These data indicate that PED regulates fibronectin production via phospholipase D/protein kinase C-beta and TGF-beta1/SMAD pathways in kidney cells. Raised PED levels may therefore contribute to the abnormal accumulation of extracellular matrix and renal dysfunction in diabetes.
Collapse
MESH Headings
- Actins/genetics
- Animals
- Astrocytes/metabolism
- Blood Pressure
- DNA Primers
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Nephropathies/epidemiology
- Fatty Acids, Nonesterified/blood
- Fibronectins/genetics
- Gene Expression Regulation
- Heart Rate
- Humans
- Insulin/blood
- Kidney/physiology
- Kidney Failure, Chronic/etiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Phenotype
- Phosphoproteins/biosynthesis
- Phosphoproteins/genetics
- Protein Kinase C/genetics
- Protein Kinase C beta
- Reverse Transcriptase Polymerase Chain Reaction
- Smad2 Protein/genetics
- Transforming Growth Factor beta1/genetics
- Up-Regulation
Collapse
Affiliation(s)
- F Oriente
- Department of Cellular and Molecular Biology and Pathology, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Negative regulators of insulin signaling revealed in a genome-wide functional screen. PLoS One 2009; 4:e6871. [PMID: 19727444 PMCID: PMC2731165 DOI: 10.1371/journal.pone.0006871] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 08/03/2009] [Indexed: 12/30/2022] Open
Abstract
Background Type 2 diabetes develops due to a combination of insulin resistance and β-cell failure and current therapeutics aim at both of these underlying causes. Several negative regulators of insulin signaling are known and are the subject of drug discovery efforts. We sought to identify novel contributors to insulin resistance and hence potentially novel targets for therapeutic intervention. Methodology An arrayed cDNA library encoding 18,441 human transcripts was screened for inhibitors of insulin signaling and revealed known inhibitors and numerous potential novel regulators. The novel hits included proteins of various functional classes such as kinases, phosphatases, transcription factors, and GTPase associated proteins. A series of secondary assays confirmed the relevance of the primary screen hits to insulin signaling and provided further insight into their modes of action. Conclusion/Significance Among the novel hits was PALD (KIAA1274, paladin), a previously uncharacterized protein that when overexpressed led to inhibition of insulin's ability to down regulate a FOXO1A-driven reporter gene, reduced upstream insulin-stimulated AKT phosphorylation, and decreased insulin receptor (IR) abundance. Conversely, knockdown of PALD gene expression resulted in increased IR abundance, enhanced insulin-stimulated AKT phosphorylation, and an improvement in insulin's ability to suppress FOXO1A-driven reporter gene activity. The present data demonstrate that the application of arrayed genome-wide screening technologies to insulin signaling is fruitful and is likely to reveal novel drug targets for insulin resistance and the metabolic syndrome.
Collapse
|
43
|
Fiory F, Formisano P, Perruolo G, Beguinot F. Frontiers: PED/PEA-15, a multifunctional protein controlling cell survival and glucose metabolism. Am J Physiol Endocrinol Metab 2009; 297:E592-601. [PMID: 19531639 DOI: 10.1152/ajpendo.00228.2009] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PED/PEA-15 is a 15-kDa ubiquitously expressed protein implicated in a number of fundamental cellular functions, including apoptosis, proliferation, and glucose metabolism. PED/PEA-15 lacks enzymatic function and serves mainly as a molecular adaptor. PED/PEA-15 is an endogenous substrate for protein kinase C (PKC), calcium/calmodulin-dependent protein kinase II (CAM kinase II), and Akt. In particular, PKC phosphorylates PED/PEA-15 at Ser(104) and CAM kinase II or Akt at Ser(116), modifying its stability. Evidence obtained over the past 10 years has indicated that PED/PEA-15 regulates cell survival by interfering with both intrinsic and extrinsic apoptotic pathways. In addition, it may also control cell proliferation by interfering with ERK1/2-mediated pathways. Indeed, PED/PEA-15 has been identified as an ERK1/2 interactor, which modifies its subcellular localization and targeting to a specific subset of substrates. Increased PED/PEA-15 levels may affect tumorigenesis and cancer progression as well as sensitivity to anticancer agents. Moreover, PED/PEA-15 affects astrocyte motility and increases susceptibility to skin carcinogenesis in vivo. PED/PEA-15 expression is regulated at the transcriptional and the posttranslational levels. Increased PED/PEA-15 expression has been identified in individuals with type 2 diabetes early during the natural history of the disease. Evidence generated over the past 10 years indicated that this defect contributes to altering glucose tolerance by impairing insulin action and insulin secretion and might play a role in the development of diabetes-associated neurological disorders. Strategies are being devised to target key signaling events in PED/PEA-15 action aimed at improving glucose tolerance and at facilitating cancer cell death.
Collapse
Affiliation(s)
- Francesca Fiory
- Dept. of Cellular and Molecular Biology and Pathology, Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Federico II Univ. of Naples, Naples, Italy
| | | | | | | |
Collapse
|
44
|
Sandomenico A, Monti SM, Sabatella M, De Capua A, Tornatore L, Doti N, Viparelli F, Dathan NA, Pedone C, Ruvo M, Marasco D. Protein-Protein Interactions: A Simple Strategy to Identify Binding Sites and Peptide Antagonists. Chem Biol Drug Des 2009; 73:483-93. [DOI: 10.1111/j.1747-0285.2009.00805.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Viparelli F, Doti N, Monti S, Marasco D, Dathan N, Pedone C, Miele C, Formisano P, Beguinot F, Ruvo M. Peptide Antagonists of the PED-hPLD1 Binding. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 611:445-6. [DOI: 10.1007/978-0-387-73657-0_192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
46
|
Ungaro P, Teperino R, Mirra P, Cassese A, Fiory F, Perruolo G, Miele C, Laakso M, Formisano P, Beguinot F. Molecular cloning and characterization of the human PED/PEA-15 gene promoter reveal antagonistic regulation by hepatocyte nuclear factor 4alpha and chicken ovalbumin upstream promoter transcription factor II. J Biol Chem 2008; 283:30970-9. [PMID: 18765665 PMCID: PMC2662169 DOI: 10.1074/jbc.m803895200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 08/27/2008] [Indexed: 11/06/2022] Open
Abstract
Overexpression of the ped/pea-15 gene in mice impairs glucose tolerance and leads to diabetes in conjunction with high fat diet treatment. PED/PEA-15 is also overexpressed in type 2 diabetics as well as in euglycemic offspring from these subjects. The cause(s) of this abnormality remains unclear. In the present work we have cloned and localized the promoter region of the human PED/PEA-15 gene within the first 230 bp of the 5(R)-flanking region. A cis-acting regulatory element located between -320 and -335 bps upstream the PED/PEA-15 gene transcriptional start site (+1) is recognized by both the hepatocyte nuclear factor 4alpha (HNF-4alpha) and the chicken ovalbumin upstream promoter transcription factor II (COUP-TFII), two members of the steroid/thyroid superfamily of transcription factors, both of which are involved in the control of lipid and glucose homeostasis. HNF-4alpha represses PED/PEA-15 expression in HeLa cells, whereas COUP-TFII activates its expression. In hepatocytes, the activation of PED/PEA-15 gene transcription is paralleled by the establishment of a partially dedifferentiated phenotype accompanied by a reduction in mRNA levels encoded by genes normally expressed during liver development. Cotransfection of HeLa cells with a reporter construct containing the PED/PEA-15 response element and various combinations of HNF-4alpha and COUP-TFII expression vectors indicated that COUP-TFII antagonizes the repression of the PED/PEA-15 gene by HNF-4alpha. Thus, at least in part, transcription of the PED/PEA-15 gene in vivo is dependent upon the intracellular balance of these positive and negative regulatory factors. Abnormalities in HNF-4alpha and COUP-TFII balance might have important consequences on glucose tolerance in humans.
Collapse
Affiliation(s)
- Paola Ungaro
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università di Napoli Federico II, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Prep1 deficiency induces protection from diabetes and increased insulin sensitivity through a p160-mediated mechanism. Mol Cell Biol 2008; 28:5634-45. [PMID: 18644868 DOI: 10.1128/mcb.00117-08] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We have examined glucose homeostasis in mice hypomorphic for the homeotic transcription factor gene Prep1. Prep1-hypomorphic (Prep1(i/i)) mice exhibit an absolute reduction in circulating insulin levels but normal glucose tolerance. In addition, these mice exhibit protection from streptozotocin-induced diabetes and enhanced insulin sensitivity with improved glucose uptake and insulin-dependent glucose disposal by skeletal muscle. This muscle phenotype does not depend on reduced expression of the known Prep1 transcription partner, Pbx1. Instead, in Prep1(i/i) muscle, we find normal Pbx1 but reduced levels of the recently identified novel Prep1 interactor p160. Consistent with this reduction, we find a muscle-selective increase in mRNA and protein levels of PGC-1alpha, accompanied by enhanced expression of the GLUT4 transporter, responsible for insulin-stimulated glucose uptake in muscle. Indeed, using L6 skeletal muscle cells, we induced the opposite effects by overexpressing Prep1 or p160, but not Pbx1. In vivo skeletal muscle delivery of p160 cDNA in Prep1(i/i) mice also reverses the molecular phenotype. Finally, we show that Prep1 controls the stability of the p160 protein. We conclude that Prep1 controls insulin sensitivity through the p160-GLUT4 pathway.
Collapse
|
49
|
Viparelli F, Cassese A, Doti N, Paturzo F, Marasco D, Dathan NA, Monti SM, Basile G, Ungaro P, Sabatella M, Miele C, Teperino R, Consiglio E, Pedone C, Beguinot F, Formisano P, Ruvo M. Targeting of PED/PEA-15 molecular interaction with phospholipase D1 enhances insulin sensitivity in skeletal muscle cells. J Biol Chem 2008; 283:21769-78. [PMID: 18541525 DOI: 10.1074/jbc.m803771200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (PED/PEA-15) is overexpressed in several tissues of individuals affected by type 2 diabetes. In intact cells and in transgenic animal models, PED/PEA-15 overexpression impairs insulin regulation of glucose transport, and this is mediated by its interaction with the C-terminal D4 domain of phospholipase D1 (PLD1) and the consequent increase of protein kinase C-alpha activity. Here we show that interfering with the interaction of PED/PEA-15 with PLD1 in L6 skeletal muscle cells overexpressing PED/PEA-15 (L6(PED/PEA-15)) restores insulin sensitivity. Surface plasmon resonance and ELISA-like assays show that PED/PEA-15 binds in vitro the D4 domain with high affinity (K(D) = 0.37 +/- 0.13 mum), and a PED/PEA-15 peptide, spanning residues 1-24, PED-(1-24), is able to compete with the PED/PEA-15-D4 recognition. When loaded into L6(PED/PEA-15) cells and in myocytes derived from PED/PEA-15-overexpressing transgenic mice, PED-(1-24) abrogates the PED/PEA-15-PLD1 interaction and reduces protein kinase C-alpha activity to levels similar to controls. Importantly, the peptide restores insulin-stimulated glucose uptake by approximately 70%. Similar results are obtained by expression of D4 in L6(PED/PEA-15). All these findings suggest that disruption of the PED/PEA-15-PLD1 molecular interaction enhances insulin sensitivity in skeletal muscle cells and indicate that PED/PEA-15 as an important target for type 2 diabetes.
Collapse
Affiliation(s)
- Francesca Viparelli
- Istituto di Biostrutture e Bioimmagini and Istituto di Endocrinologia e Oncologia Sperimentale Gaetano Salvatore, Consiglio Nazionale delle Ricerche, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Expression and purification of the D4 region of PLD1 and characterization of its interaction with PED-PEA15. Protein Expr Purif 2008; 59:302-8. [DOI: 10.1016/j.pep.2008.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 02/22/2008] [Accepted: 02/22/2008] [Indexed: 11/20/2022]
|