1
|
Huang N, Zhu X, Shu Z, Chen S, Wu X, Wang H, Huang X, Hu X, Sun J, Chen P, Graf R, Bai J, Wang B, Li L. Association between elevated serum REG Iα levels and eGFR decline in patients with chronic kidney disease: a cross-sectional study in eastern China. BMJ Open 2025; 15:e086874. [PMID: 39947821 PMCID: PMC11831304 DOI: 10.1136/bmjopen-2024-086874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 01/08/2025] [Indexed: 02/19/2025] Open
Abstract
OBJECTIVES This study aimed to investigate the relationship between serum regenerating protein Iα (REG Iα) levels and estimated glomerular filtration rate (eGFR) and to evaluate the diagnostic efficiency of REG Iα in chronic kidney disease (CKD). DESIGN This is a cross-sectional study. SETTING The study was conducted in eastern China between August 2022 and August 2023. PARTICIPANTS A total of 880 participants aged over 18 years were enrolled, with 220 non-CKD participants (111 males, 50.45%) and 660 patients with CKD (366 males, 55.45%). CKD was diagnosed based on the Kidney Disease: Improving Global Outcomes (KDIGO) 2012 guidelines. Exclusion criteria included participation in other trials, acute kidney injury, end-stage kidney disease undergoing renal replacement therapy, pregnancy, active infections, gastrointestinal or pancreatic inflammation, history of gastrointestinal or pancreatic resections, cancer and mental disorders. RESULTS Serum REG Iα was significantly higher in the CKD group (125.54 (60.28-303.39) ng/mL) compared with those in the non-CKD group (24.62 (14.09-37.32) ng/mL, p<0.001). Positive correlations were observed between serum REG Iα and serum creatinine, cystatin C (Cys-C), and kidney injury molecule 1 (KIM-1), while a negative correlation was identified with eGFR. After adjusting for sex, diabetes, hypertension and fasting blood glucose, the multivariate regression analysis demonstrated a significant association between serum REG Iα and eGFR (OR=1.737 (1.263-2.388), p=0.001). Furthermore, serum REG Iα levels increased progressively with declining kidney function categorised by eGFR (p<0.001). In CKD screening, serum REG Iα demonstrated strong diagnostic performance, with an area under the receiver operating characteristic curves (AUC) of 0.860 (0.813-0.899), providing a sensitivity of 71.63%, a specificity of 86.89%, a positive predictive value of 94.30% and a negative predictive value of 46.85%. Additionally, serum REG Iα exhibited an AUC of 0.769 (0.712-0.819) for identifying high- and very-high-risk CKD based on KDIGO risk stratification. Its sensitivity significantly outperformed serum Cys-C and KIM-1 (82.80% vs 75.16% and 36.94%, respectively). CONCLUSIONS This study provided compelling evidence that serum REG Iα levels were notably elevated in patients with CKD and closely associated with kidney function. REG Iα may serve as a promising biomarker for CKD detection and risk stratification. CLINICAL TRIAL REGISTRATION The study was approved by the Ethics Committee of Zhongda Hospital (approval number: 2022ZDSYLL204-P01) and conducted in compliance with the Helsinki Declaration. The clinical trial was registered under ChiCTR2300072247.
Collapse
Affiliation(s)
- Nan Huang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| | - Xiangyun Zhu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| | - Zhiyi Shu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| | - Sheng Chen
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| | - Xiaodong Wu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| | - Hui Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| | - Xi Huang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiuxiu Hu
- Department of Pathology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jinfang Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Pingsheng Chen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Rolf Graf
- Department of Visceral and Transplantation Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Jianling Bai
- Department of Biostatistics, School of Public Heath, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bin Wang
- Department of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Yu L, Wu Q, Jiang S, Liu J, Liu J, Chen G. Controversial Roles of Regenerating Family Proteins in Tissue Repair and Tumor Development. Biomedicines 2024; 13:24. [PMID: 39857608 PMCID: PMC11762848 DOI: 10.3390/biomedicines13010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Over the past 40 years since the discovery of regenerating family proteins (Reg proteins), numerous studies have highlighted their biological functions in promoting cell proliferation and resisting cell apoptosis, particularly in the regeneration and repair of pancreatic islets and exocrine glands. Successively, short peptides derived from Reg3δ and Reg3α have been employed in clinical trials, showing favorable therapeutic effects in patients with type I and type II diabetes. However, continued reports have been limited, presumably attributed to the potential side effects. Methods: This review summarizes extensive research on Reg proteins over the past decade, combined with our own related studies, proposing that Reg proteins exhibit dimorphic effects. Results: The activity of Reg proteins is not as simplistic as previously perceived but shows auto-immunogenicity depending on different pathophysiological microenvironments. The immunogenicity of Reg proteins could recruit immune cells leading to an anti-tumor effect. Such functional diversity is correlated with their structural characteristics: the N-terminal region contributes to autoantigenicity, while the C-type lectin fragment near the C-terminal determines the trophic action. It should be noted that B-cell masking antigens might also reside within the C-type lectin domain. Conclusions: Reg proteins have dual functional roles under various physiological and pathological conditions. These theoretical foundations facilitate the subsequent development of diagnostic reagents and therapeutic drugs targeting Reg proteins.
Collapse
Affiliation(s)
- Luting Yu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Qingyun Wu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Shenglong Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Jia Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Junli Liu
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Guoguang Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| |
Collapse
|
3
|
Lee GH, Kim H, Moon HW, Yun YM, Park M, Lee S, Hur M. Diagnostic and Prognostic Utilities of Pancreatic Stone Protein in Patients with Suspected Sepsis. Diagnostics (Basel) 2024; 14:2076. [PMID: 39335755 PMCID: PMC11430866 DOI: 10.3390/diagnostics14182076] [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: 08/02/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Pancreatic stone protein (PSP) is an emerging biomarker of sepsis that is secreted from pancreas sensing remote organ damages. We explored the diagnostic and prognostic utilities of PSP in patients with suspected sepsis. Methods: In a total of 285 patients (suspected sepsis, n = 148; sepsis, n = 137), we compared PSP with procalcitonin (PCT) and sequential organ failure assessment (SOFA) score. Sepsis diagnoses were explored using receiver operating characteristic curve analyses with area under the curves (AUCs). Clinical outcomes (in-hospital mortality, 30-day mortality, and kidney replacement therapy [KRT]) were explored using the Kaplan-Meier method and a multivariate analysis with hazard ratio (HR). Results: PCT and PSP were comparable for sepsis diagnosis (AUC = 0.71-0.72, p < 0.001). The sepsis proportion was significantly higher when both biomarkers increased than when either one or both biomarkers did not increase (89.0% vs. 21.3-47.7%, p < 0.001). Each biomarker quartile (Q1-Q4) differed significantly according to their SOFA score (all p < 0.001). Compared with Q1, the Q2-Q4 groups showed worse clinical outcomes (p = 0.002-0.041). Both biomarkers added to the SOFA score showed higher HRs than the SOFA score alone (3.3-9.6 vs. 2.8-4.2, p < 0.001-0.011), with nearly 2.5-fold higher HR (9.6 vs. 4.2) for predicting KRT. Conclusions: Although PCT and PSP did not independently predict clinical outcomes in the multivariate analysis, PSP demonstrated diagnostic and prognostic utilities in patients with suspected sepsis, especially for predicting kidney dysfunction. PSP, alone or in combination with PCT, would be a valuable tool that can be added to clinical assessments.
Collapse
Affiliation(s)
- Gun-Hyuk Lee
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| | - Hanah Kim
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| | - Hee-Won Moon
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| | - Yeo-Min Yun
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| | - Mikyoung Park
- Department of Laboratory Medicine, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Seungho Lee
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan 49201, Republic of Korea
| | - Mina Hur
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| |
Collapse
|
4
|
Zhu S, Waeckel-Énée E, Oshima M, Moser A, Bessard MA, Gdoura A, Roger K, Mode N, Lipecka J, Yilmaz A, Bertocci B, Diana J, Saintpierre B, Guerrera IC, Scharfmann R, Francesconi S, Mauvais FX, van Endert P. Islet cell stress induced by insulin-degrading enzyme deficiency promotes regeneration and protection from autoimmune diabetes. iScience 2024; 27:109929. [PMID: 38799566 PMCID: PMC11126816 DOI: 10.1016/j.isci.2024.109929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/08/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Tuning of protein homeostasis through mobilization of the unfolded protein response (UPR) is key to the capacity of pancreatic beta cells to cope with variable demand for insulin. Here, we asked how insulin-degrading enzyme (IDE) affects beta cell adaptation to metabolic and immune stress. C57BL/6 and autoimmune non-obese diabetic (NOD) mice lacking IDE were exposed to proteotoxic, metabolic, and immune stress. IDE deficiency induced a low-level UPR with islet hypertrophy at the steady state, rapamycin-sensitive beta cell proliferation enhanced by proteotoxic stress, and beta cell decompensation upon high-fat feeding. IDE deficiency also enhanced the UPR triggered by proteotoxic stress in human EndoC-βH1 cells. In Ide-/- NOD mice, islet inflammation specifically induced regenerating islet-derived protein 2, a protein attenuating autoimmune inflammation. These findings establish a role of IDE in islet cell protein homeostasis, demonstrate how its absence induces metabolic decompensation despite beta cell proliferation, and UPR-independent islet regeneration in the presence of inflammation.
Collapse
Affiliation(s)
- Shuaishuai Zhu
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | | | - Masaya Oshima
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Anna Moser
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Marie-Andrée Bessard
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Abdelaziz Gdoura
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Kevin Roger
- Université Paris Cité, INSERM, CNRS, Structure Fédérative de Recherche Necker, Proteomics Platform, F-75015 Paris, France
| | - Nina Mode
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Joanna Lipecka
- Université Paris Cité, INSERM, CNRS, Structure Fédérative de Recherche Necker, Proteomics Platform, F-75015 Paris, France
| | - Ayse Yilmaz
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Barbara Bertocci
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Julien Diana
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
| | | | - Ida Chiara Guerrera
- Université Paris Cité, INSERM, CNRS, Structure Fédérative de Recherche Necker, Proteomics Platform, F-75015 Paris, France
| | - Raphael Scharfmann
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Stefania Francesconi
- Genome Dynamics Unit, Institut Pasteur, Centre National de la Recherche Scientifique, UMR3525, F-75015 Paris, France
| | - François-Xavier Mauvais
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
- Service de Physiologie – Explorations Fonctionnelles Pédiatriques, AP-HP, Hôpital Universitaire Robert Debré, F-75019 Paris, France
| | - Peter van Endert
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, F-75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| |
Collapse
|
5
|
Zhu S, Waeckel-Énée E, Moser A, Bessard MA, Roger K, Lipecka J, Yilmaz A, Bertocci B, Diana J, Saintpierre B, Guerrera IC, Francesconi S, Mauvais FX, van Endert P. Pancreatic islet cell stress induced by insulin-degrading enzyme deficiency promotes islet regeneration and protection from autoimmune diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549693. [PMID: 37503145 PMCID: PMC10370150 DOI: 10.1101/2023.07.19.549693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Appropriate tuning of protein homeostasis through mobilization of the unfolded protein response (UPR) is key to the capacity of pancreatic beta cells to cope with highly variable demand for insulin synthesis. An efficient UPR ensures a sufficient beta cell mass and secretory output but can also affect beta cell resilience to autoimmune aggression. The factors regulating protein homeostasis in the face of metabolic and immune challenges are insufficiently understood. We examined beta cell adaptation to stress in mice deficient for insulin-degrading enzyme (IDE), a ubiquitous protease with high affinity for insulin and genetic association with type 2 diabetes. IDE deficiency induced a low-level UPR in both C57BL/6 and autoimmune non-obese diabetic (NOD) mice, associated with rapamycin-sensitive beta cell proliferation strongly enhanced by proteotoxic stress. Moreover, in NOD mice, IDE deficiency protected from spontaneous diabetes and triggered an additional independent pathway, conditional on the presence of islet inflammation but inhibited by proteotoxic stress, highlighted by strong upregulation of regenerating islet-derived protein 2, a protein attenuating autoimmune inflammation. Our findings establish a key role of IDE in islet cell protein homeostasis, identify a link between low-level UPR and proliferation, and reveal an UPR-independent anti-inflammatory islet cell response uncovered in the absence of IDE of potential interest in autoimmune diabetes.
Collapse
|
6
|
Gonzalez P, Dos Santos A, Darnaud M, Moniaux N, Rapoud D, Lacoste C, Nguyen TS, Moullé VS, Deshayes A, Amouyal G, Amouyal P, Bréchot C, Cruciani-Guglielmacci C, Andréelli F, Magnan C, Faivre J. Antimicrobial protein REG3A regulates glucose homeostasis and insulin resistance in obese diabetic mice. Commun Biol 2023; 6:269. [PMID: 36918710 PMCID: PMC10015038 DOI: 10.1038/s42003-023-04616-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Innate immune mediators of pathogen clearance, including the secreted C-type lectins REG3 of the antimicrobial peptide (AMP) family, are known to be involved in the regulation of tissue repair and homeostasis. Their role in metabolic homeostasis remains unknown. Here we show that an increase in human REG3A improves glucose and lipid homeostasis in nutritional and genetic mouse models of obesity and type 2 diabetes. Mice overexpressing REG3A in the liver show improved glucose homeostasis, which is reflected in better insulin sensitivity in normal weight and obese states. Delivery of recombinant REG3A protein to leptin-deficient ob/ob mice or wild-type mice on a high-fat diet also improves glucose homeostasis. This is accompanied by reduced oxidative protein damage, increased AMPK phosphorylation and insulin-stimulated glucose uptake in skeletal muscle tissue. Oxidative damage in differentiated C2C12 myotubes is greatly attenuated by REG3A, as is the increase in gp130-mediated AMPK activation. In contrast, Akt-mediated insulin action, which is impaired by oxidative stress, is not restored by REG3A. These data highlight the importance of REG3A in controlling oxidative protein damage involved in energy and metabolic pathways during obesity and diabetes, and provide additional insight into the dual function of host-immune defense and metabolic regulation for AMP.
Collapse
Affiliation(s)
- Patrick Gonzalez
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Alexandre Dos Santos
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Marion Darnaud
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Nicolas Moniaux
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Delphine Rapoud
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Claire Lacoste
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Tung-Son Nguyen
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Valentine S Moullé
- Université of Paris, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, 75013, France
| | - Alice Deshayes
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | | | | | | | | | - Fabrizio Andréelli
- Sorbonne Université, INSERM, NutriOmics team, Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, 75013, France
| | - Christophe Magnan
- Université of Paris, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, 75013, France
| | - Jamila Faivre
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France.
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France.
- Assistance Publique-Hôpitaux de Paris (AP-HP). Université Paris Saclay, Medical-University Department (DMU) Biology, Genetics, Pharmacy, Paul-Brousse Hospital, Villejuif, 94800, France.
| |
Collapse
|
7
|
Hill JH, Massaquoi MS, Sweeney EG, Wall ES, Jahl P, Bell R, Kallio K, Derrick D, Murtaugh LC, Parthasarathy R, Remington SJ, Round JL, Guillemin K. BefA, a microbiota-secreted membrane disrupter, disseminates to the pancreas and increases β cell mass. Cell Metab 2022; 34:1779-1791.e9. [PMID: 36240759 PMCID: PMC9633563 DOI: 10.1016/j.cmet.2022.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/26/2022] [Accepted: 08/31/2022] [Indexed: 01/11/2023]
Abstract
Microbiome dysbiosis is a feature of diabetes, but how microbial products influence insulin production is poorly understood. We report the mechanism of BefA, a microbiome-derived protein that increases proliferation of insulin-producing β cells during development in gnotobiotic zebrafish and mice. BefA disseminates systemically by multiple anatomic routes to act directly on pancreatic islets. We detail BefA's atomic structure, containing a lipid-binding SYLF domain, and demonstrate that it permeabilizes synthetic liposomes and bacterial membranes. A BefA mutant impaired in membrane disruption fails to expand β cells, whereas the pore-forming host defense protein, Reg3, stimulates β cell proliferation. Our work demonstrates that membrane permeabilization by microbiome-derived and host defense proteins is necessary and sufficient for β cell expansion during pancreas development, potentially connecting microbiome composition with diabetes risk.
Collapse
Affiliation(s)
- Jennifer Hampton Hill
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | - Elena S Wall
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Philip Jahl
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Department of Physics and Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Rickesha Bell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Karen Kallio
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Daniel Derrick
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - L Charles Murtaugh
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Raghuveer Parthasarathy
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Department of Physics and Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - S James Remington
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA; Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1Z8, Canada.
| |
Collapse
|
8
|
Peters LM, Howard J, Leeb T, Mevissen M, Graf R, Reding Graf T. Identification of regenerating island-derived protein 3E in dogs. Front Vet Sci 2022; 9:1010809. [PMID: 36387376 PMCID: PMC9650133 DOI: 10.3389/fvets.2022.1010809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/12/2022] [Indexed: 11/29/2022] Open
Abstract
Regenerating islet-derived protein (REG) 1A (aka pancreatic stone protein) and REG3A (aka pancreatitis-associated protein) are upregulated in humans with sepsis, pancreatitis, and gastrointestinal diseases, but little is known about this protein family in dogs. Our aim was to identify REG1 and REG3 family members in dogs. REG-family genes were computationally annotated in the canine genome and proteome, with verification of gene expression using publicly available RNA-seq data. The presence of the protein in canine pancreatic tissue and plasma was investigated with Western blot and immunohistochemistry, using anti-human REG1A and REG3A antibodies. Protein identity was confirmed with mass spectrometry. Two members of the REG3 subfamily were found in the canine genome, REG3E1 and REG3E2, both encoding for the same 176 AA protein, subsequently named REG3E. Anti-human REG3A antibodies demonstrated cross-reactivity with the canine REG3E protein in pancreas homogenates. In canine plasma, a protein band of approximately 17 kDa was apparent. Mass spectrometry confirmed this protein to be the product of the two annotated REG3E genes. Strong immunoreactivity to anti-human REG3A antibodies was found in sections of canine pancreas affected with acute pancreatitis, but it was weak in healthy pancreatic tissue. Recombinant canine REG3E protein underwent a selective trypsin digestion as described in other species. No evidence for the presence of a homolog of REG1A in dogs was found in any of the investigations. In conclusion, dogs express REG3E in the pancreas, whose role as biomarker merits further investigations. Homologs to human REG1A are not likely to exist in dogs.
Collapse
Affiliation(s)
- Laureen M. Peters
- Department of Clinical Veterinary Medicine, Clinical Diagnostic Laboratory, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- *Correspondence: Laureen M. Peters
| | - Judith Howard
- Department of Clinical Veterinary Medicine, Clinical Diagnostic Laboratory, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tosso Leeb
- Department of Clinical Research and Veterinary Public Health, Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Meike Mevissen
- Division of Veterinary Pharmacology and Toxicology, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Rolf Graf
- Department of Surgery and Transplantation, Pancreas Research Laboratory, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Theresia Reding Graf
- Department of Surgery and Transplantation, Pancreas Research Laboratory, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| |
Collapse
|
9
|
Koopen A, Witjes J, Wortelboer K, Majait S, Prodan A, Levin E, Herrema H, Winkelmeijer M, Aalvink S, Bergman JJGHM, Havik S, Hartmann B, Levels H, Bergh PO, van Son J, Balvers M, Bastos DM, Stroes E, Groen AK, Henricsson M, Kemper EM, Holst J, Strauch CM, Hazen SL, Bäckhed F, De Vos WM, Nieuwdorp M, Rampanelli E. Duodenal Anaerobutyricum soehngenii infusion stimulates GLP-1 production, ameliorates glycaemic control and beneficially shapes the duodenal transcriptome in metabolic syndrome subjects: a randomised double-blind placebo-controlled cross-over study. Gut 2022; 71:1577-1587. [PMID: 34697034 PMCID: PMC9279853 DOI: 10.1136/gutjnl-2020-323297] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/09/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Although gut dysbiosis is increasingly recognised as a pathophysiological component of metabolic syndrome (MetS), the role and mode of action of specific gut microbes in metabolic health remain elusive. Previously, we identified the commensal butyrogenic Anaerobutyricum soehngenii to be associated with improved insulin sensitivity in subjects with MetS. In this proof-of-concept study, we investigated the potential therapeutic effects of A. soehngenii L2-7 on systemic metabolic responses and duodenal transcriptome profiles in individuals with MetS. DESIGN In this randomised double-blind placebo-controlled cross-over study, 12 male subjects with MetS received duodenal infusions of A. soehngenii/ placebo and underwent duodenal biopsies, mixed meal tests (6 hours postinfusion) and 24-hour continuous glucose monitoring. RESULTS A. soehngenii treatment provoked a markedly increased postprandial excursion of the insulinotropic hormone glucagon-like peptide 1 (GLP-1) and an elevation of plasma secondary bile acids, which were positively associated with GLP-1 levels. Moreover, A. soehngenii treatment robustly shaped the duodenal expression of 73 genes, with the highest fold induction in the expression of regenerating islet-protein 1B (REG1B)-encoding gene. Strikingly, duodenal REG1B expression positively correlated with GLP-1 levels and negatively correlated with peripheral glucose variability, which was significantly diminished in the 24 hours following A. soehngenii intake. Mechanistically, Reg1B expression is induced upon sensing butyrate or bacterial peptidoglycan. Importantly, A. soehngenii duodenal administration was safe and well tolerated. CONCLUSIONS A single dose of A. soehngenii improves peripheral glycaemic control within 24 hours; it specifically stimulates intestinal GLP-1 production and REG1B expression. Further studies are needed to delineate the specific pathways involved in REG1B induction and function in insulin sensitivity. TRIAL REGISTRATION NUMBER NTR-NL6630.
Collapse
Affiliation(s)
- Annefleur Koopen
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Julia Witjes
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Koen Wortelboer
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Soumia Majait
- Clinical Pharmacy, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Andrei Prodan
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Evgeni Levin
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Hilde Herrema
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Steven Aalvink
- Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Stephan Havik
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Bolette Hartmann
- Biomedical Sciences, University of Copenhagen Novo Nordisk Foundation Center for Basic Metabolic Research, Kobenhavn, Denmark
| | - Han Levels
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Per-Olof Bergh
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Goteborg, Sweden
| | - Jamie van Son
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Manon Balvers
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | | | - Erik Stroes
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Albert K Groen
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Marcus Henricsson
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Goteborg, Sweden
| | | | - Jens Holst
- Biomedical Sciences, University of Copenhagen Novo Nordisk Foundation Center for Basic Metabolic Research, Kobenhavn, Denmark
| | - Christopher M Strauch
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stanley L Hazen
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fredrik Bäckhed
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Goteborg, Sweden
| | - Willem M De Vos
- Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Elena Rampanelli
- Experimental Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Pancreatic Stone Protein: Review of a New Biomarker in Sepsis. J Clin Med 2022; 11:jcm11041085. [PMID: 35207355 PMCID: PMC8880320 DOI: 10.3390/jcm11041085] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 01/27/2023] Open
Abstract
Sepsis is a life-threatening syndrome characterized by a dysregulated host response to an infection that may evolve rapidly into septic shock and multiple organ failure. Management of sepsis relies on the early recognition and diagnosis of infection and the providing of adequate and prompt antibiotic therapy and organ support. A novel protein biomarker, the pancreatic stone protein (PSP), has recently been studied as a biomarker of sepsis and the available evidence suggests that it has a higher diagnostic performance for the identification of infection than the most used available biomarkers and adds prognostic value. This review summarizes the clinical evidence available for PSP in the diagnosis and prognosis of sepsis.
Collapse
|
11
|
OKAMOTO H, TAKASAWA S. Okamoto model for necrosis and its expansions, CD38-cyclic ADP-ribose signal system for intracellular Ca 2+ mobilization and Reg (Regenerating gene protein)-Reg receptor system for cell regeneration. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:423-461. [PMID: 34629354 PMCID: PMC8553518 DOI: 10.2183/pjab.97.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/22/2021] [Indexed: 05/03/2023]
Abstract
In pancreatic islet cell culture models and animal models, we studied the molecular mechanisms involved in the development of insulin-dependent diabetes. The diabetogenic agents, alloxan and streptozotocin, caused DNA strand breaks, which in turn activated poly(ADP-ribose) polymerase/synthetase (PARP) to deplete NAD+, thereby inhibiting islet β-cell functions such as proinsulin synthesis and ultimately leading to β-cell necrosis. Radical scavengers protected against the formation of DNA strand breaks and inhibition of proinsulin synthesis. Inhibitors of PARP prevented the NAD+ depletion, inhibition of proinsulin synthesis and β-cell death. These findings led to the proposed unifying concept for β-cell damage and its prevention (the Okamoto model). The model met one proof with PARP knockout animals and was further extended by the discovery of cyclic ADP-ribose as the second messenger for Ca2+ mobilization in glucose-induced insulin secretion and by the identification of Reg (Regenerating gene) for β-cell regeneration. Physiological and pathological events found in pancreatic β-cells have been observed in other cells and tissues.
Collapse
Affiliation(s)
- Hiroshi OKAMOTO
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Shin TAKASAWA
- Department of Biochemistry, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
12
|
Ebrahimi AG, Hollister-Lock J, Sullivan BA, Tsuchida R, Bonner-Weir S, Weir GC. Beta cell identity changes with mild hyperglycemia: Implications for function, growth, and vulnerability. Mol Metab 2020; 35:100959. [PMID: 32244186 PMCID: PMC7082551 DOI: 10.1016/j.molmet.2020.02.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE As diabetes develops, marked reductions of insulin secretion are associated with very modest elevations of glucose. We wondered if these glucose changes disrupt beta cell differentiation enough to account for the altered function. METHODS Rats were subjected to 90% partial pancreatectomies and those with only mild glucose elevations 4 weeks or 10 weeks after surgery had major alterations of gene expression in their islets as determined by RNAseq. RESULTS Changes associated with glucose toxicity demonstrated that many of the critical genes responsible for insulin secretion were downregulated while the expression of normally suppressed genes increased. Also, there were marked changes in genes associated with replication, aging, senescence, stress, inflammation, and increased expression of genes controlling both class I and II MHC antigens. CONCLUSIONS These findings suggest that mild glucose elevations in the early stages of diabetes lead to phenotypic changes that adversely affect beta cell function, growth, and vulnerability.
Collapse
Affiliation(s)
- Aref G Ebrahimi
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place, Harvard Medical School, Boston, MA 02215, USA
| | - Jennifer Hollister-Lock
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place, Harvard Medical School, Boston, MA 02215, USA
| | - Brooke A Sullivan
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place, Harvard Medical School, Boston, MA 02215, USA
| | - Ryohei Tsuchida
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place, Harvard Medical School, Boston, MA 02215, USA
| | - Susan Bonner-Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place, Harvard Medical School, Boston, MA 02215, USA
| | - Gordon C Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
13
|
Chen Z, Downing S, Tzanakakis ES. Four Decades After the Discovery of Regenerating Islet-Derived (Reg) Proteins: Current Understanding and Challenges. Front Cell Dev Biol 2019; 7:235. [PMID: 31696115 PMCID: PMC6817481 DOI: 10.3389/fcell.2019.00235] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
Regenerating islet-derived (Reg) proteins have emerged as multifunctional agents with pro-proliferative, anti-apoptotic, differentiation-inducing and bactericidal properties. Over the last 40 years since first discovered, Reg proteins have been implicated in a gamut of maladies including diabetes, various types of cancer of the digestive tract, and Alzheimer disease. Surprisingly though, a consensus is still absent on the regulation of their expression, and molecular underpinning of their function. Here, we provide a critical appraisal of recent findings in the field of Reg protein biology. Specifically, the structural characteristics are reviewed particularly in connection with established or purported functions of different members of the Reg family. Moreover, Reg expression patterns in different tissues both under normal and pathophysiological conditions are summarized. Putative receptors and cascades reported to relay Reg signaling inciting cellular responses are presented aiming at a better appreciation of the biological activities of the distinct Reg moieties. Challenges are also discussed that have hampered thus far the rapid progress in this field such as the use of non-standard nomenclature for Reg molecules among various research groups, the existence of multiple Reg members with significant degree of homology and possibly compensatory modes of action, and the need for common assays with robust readouts of Reg activity. Coordinated research is warranted going forward, given that several research groups have independently linked Reg proteins to diseased states and raised the possibility that these biomolecules can serve as therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, United States
| | - Shawna Downing
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
| | - Emmanuel S Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, United States.,Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
14
|
Downing S, Zhang F, Chen Z, Tzanakakis ES. MicroRNA-7 directly targets Reg1 in pancreatic cells. Am J Physiol Cell Physiol 2019; 317:C366-C374. [PMID: 31166710 DOI: 10.1152/ajpcell.00013.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerating islet-derived (Reg) proteins, which were first discovered in the pancreas, are associated with increased proliferation, prevention of apoptosis, and enhanced differentiation in normal and disease states, but very little is known about the regulation of their expression. We hypothesized that Reg expression is influenced by microRNAs. Bioinformatic analysis predicted Reg1 to be a target of microRNA-7 (miR-7), which influences pancreatic β-cell function. To this end, we investigated the effects of miR-7 on Reg1 expression in pancreatic acinar and islet β-cells. High levels of Reg1 were noted by immunostaining and Western blotting in acinar cells in contrast to islet cells. A reciprocal expression pattern was observed for miR-7. Overexpression of miR-7 resulted in Reg1 mRNA suppression and reduction of secreted Reg1 protein. Conversely, miR-7 knockdown led to increases in Reg1. Targeting of Reg1 by miR-7 was confirmed via luciferase activity assays. In contrast, miR-7 did not directly repress the human ortholog of Reg1 REG1A as well as REG1B indicating species differences in the regulation of Reg expression. This is the first account of microRNA modulation of any Reg member warranting studies to fill gaps in our knowledge of Reg protein biology, particularly in disease contexts.
Collapse
Affiliation(s)
- Shawna Downing
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts
| | - Fan Zhang
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts
| | - Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts
| | - Emmanuel S Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts.,Clinical and Translational Science Institute, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
15
|
Lo CW, Kryvalap Y, Sheu TJ, Chang CH, Czyzyk J. Cellular proliferation in mouse and human pancreatic islets is regulated by serpin B13 inhibition and downstream targeting of E-cadherin by cathepsin L. Diabetologia 2019; 62:822-834. [PMID: 30824970 DOI: 10.1007/s00125-019-4834-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS We previously reported that exposure to antibodies neutralising serpin B13, a protease inhibitor expressed in exocrine pancreatic ducts, promotes beta cell proliferation, underscoring the importance of a functional relationship between exocrine and endocrine pancreas. The aim of the present study was to identify the molecular events that link inhibition of serpin B13 to islet cell proliferation. METHODS We used an in vitro culture system consisting of isolated pancreatic islets, an extract of pancreatic ductal epithelium and a monoclonal antibody (mAb) to serpin B13 or IgG isotype control. In vivo studies involved treatment of mice with these mAbs. RESULTS The catalytic activity of cathepsin L (CatL), a cysteine protease target of serpin B13, was augmented in the pancreas of mice injected with serpin B13 mAb. Furthermore, the addition of serpin B13 mAb to the islets, together with the pancreatic ductal epithelium lysate, caused CatL-dependent cleavage of E-cadherin and concomitant upregulation of REG genes, ultimately leading to beta cell proliferation. Direct blockade of E-cadherin with mAb also markedly enhanced REG gene induction, while chemical inhibition of β-catenin, a binding target of E-cadherin, prevented the serpin B13 mAb-induced upregulation of REG genes. CONCLUSIONS/INTERPRETATION Our work implicates the CatL-E-cadherin-REG pathway in the regulation of islet cell proliferation in response to signals generated in exocrine pancreatic tissue and demonstrates that protease activity may promote adaptive changes in the islets. DATA AVAILABILITY Microarray data that support the findings of this study have been deposited in Gene Expression Omnibus (GEO) with the accession no. GSE125151.
Collapse
Affiliation(s)
- Chi-Wen Lo
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY, USA
| | - Yury Kryvalap
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave SE, Minneapolis, MN, 55455, USA
| | - Tzong-Jen Sheu
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Ching-Ho Chang
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Jan Czyzyk
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
16
|
Huan C, Stanek A, Mueller C, Ou P, Dong S, Zhang J, Abdel-Naby R, Gruessner R. Loss of Reg proteins' protection of islet β cells in chronic pancreatitis: A potential mechanism for the pathogenesis of type 3c diabetes. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2019; 5:21-28. [DOI: 10.1016/j.coemr.2019.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2025]
|
17
|
Aida K, Kobayashi T, Takeshita A, Jimbo E, Nishida Y, Yagihashi S, Hosoi M, Fukui T, Sugawara A, Takasawa S. Crucial role of Reg I from acinar-like cell cluster touching with islets (ATLANTIS) on mitogenesis of beta cells in EMC virus-induced diabetic mice. Biochem Biophys Res Commun 2018; 503:963-969. [PMID: 29935186 DOI: 10.1016/j.bbrc.2018.06.103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
Abstract
Recently, we reported the presence of distinct cell clusters named acinar-like cell clusters touching Langerhans islets with thin interstitial surrounding (ATLANTIS) in human pancreas. A morphological study in humans demonstrated that ATLANTIS and islet cell clusters are found together in the microenvironment enclosed by a common basement membrane, and ATLANTIS releases vesicles containing Regenerating gene protein (REG Iα) to islet cell clusters. We examined 1) the presence or absence of ATLANTIS in homozygous Reg I (mouse homologue of human REG Iα) deficient (Reg I-/-) and wild-type mice, and 2) the possible role of ATLANTIS in the regeneration of beta cell clusters after encephalomyocarditis (EMC) virus (D-variant) infection in Reg I-/- and wild-type mice. ATLANTIS was found in both wild-type and Reg I-/- mice. In both groups, mean blood glucose increased transiently to greater than 14.0 mmol/L at 5 days after EMC virus infection and recovered to baseline at 12 days. At 12 days after EMC virus infection, lower BrdU labeling indices were observed in islet beta cells of Reg I-/- mice compared to wild-type mice. Beta cell volume 12 days after EMC virus infection in Reg I-/- mice did not differ from that of wild-type mice. These results suggest that Reg I, which is released from ATLANTIS to islet beta cell clusters, has a crucial role in beta cell regeneration in EMC virus-induced diabetes. The presence of mechanism(s) other than that mediated by Reg I in beta cell restoration after destruction by EMC virus was also suggested.
Collapse
Affiliation(s)
- Kaoru Aida
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Tetsuro Kobayashi
- Division of Immunology and Molecular Medicine, Okinaka Memorial Institute for Medical Research, Tokyo, Japan; Department of Endocrinology and Metabolism, Toranomon Hospital, Tokyo, Japan.
| | - Akira Takeshita
- Department of Endocrinology and Metabolism, Toranomon Hospital, Tokyo, Japan
| | - Erika Jimbo
- Division of Immunology and Molecular Medicine, Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Yoriko Nishida
- Department of Nursing, Interdisciplinary Graduate School of Medicine and Engineering, Department of Medical Sciences Nursing Science (Basic and Clinical Nursing Sciences), University of Yamanashi, Yamanashi, Japan
| | - Soroku Yagihashi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Mitsuko Hosoi
- Division of Immunology and Molecular Medicine, Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Tomoyasu Fukui
- Division of Diabetes, Metabolism and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shin Takasawa
- Department of Biochemistry (Biochemistry and Molecular Biology), Nara Medical University, Kashihara, Japan
| |
Collapse
|
18
|
Significance of Interleukin-6/STAT Pathway for the Gene Expression of REG Iα, a New Autoantigen in Sjögren's Syndrome Patients, in Salivary Duct Epithelial Cells. Clin Rev Allergy Immunol 2018; 52:351-363. [PMID: 27339601 DOI: 10.1007/s12016-016-8570-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The regenerating gene, Reg, was originally isolated from a rat regenerating islet complementary DNA (cDNA) library, and its human homologue was named REG Iα. Recently, we reported that REG Iα messenger RNA (mRNA), as well as its product, was overexpressed in ductal epithelial cells in the salivary glands of Sjögren's syndrome patients. Furthermore, autoantibodies against REG Iα were found in the sera of Sjögren's syndrome patients, and the patients who were positive for the anti-REG Iα antibody showed significantly lower saliva secretion than antibody-negative patients. We found the mechanism of REG Iα induction in salivary ductal epithelial cells. Reporter plasmid containing REG Iα promoter (-1190/+26) upstream of a luciferase gene was introduced into human NS-SV-DC and rat A5 salivary ductal cells. The cells were treated with several cytokines (interleukin (IL)-6, IL-8, etc.), upregulated in Sjögren's syndrome salivary ducts, and the transcriptional activity was measured. IL-6 stimulation significantly enhanced the REG Iα promoter activity in both cells. Deletion analysis revealed that the -141∼-117 region of the REG Iα gene was responsible for the promoter activation by IL-6, which contains a consensus sequence for signal transducer and activator of transcription (STAT) binding. The introduction of small interfering RNA for human STAT3 abolished IL-6-induced REG Iα transcription. These results indicated that IL-6 stimulation induced REG Iα transcription through STAT3 activation and binding to the REG Iα promoter in salivary ductal cells. This dependence of REG Iα induction upon IL-6/STAT in salivary duct epithelial cells may play an important role in the pathogenesis/progression of Sjögren's syndrome.
Collapse
|
19
|
Duran-Ortiz S, Brittain AL, Kopchick JJ. The impact of growth hormone on proteomic profiles: a review of mouse and adult human studies. Clin Proteomics 2017; 14:24. [PMID: 28670222 PMCID: PMC5492507 DOI: 10.1186/s12014-017-9160-2] [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: 02/06/2017] [Accepted: 06/20/2017] [Indexed: 12/17/2022] Open
Abstract
Growth hormone (GH) is a protein that is known to stimulate postnatal growth, counter regulate insulin's action and induce expression of insulin-like growth factor-1. GH exerts anabolic or catabolic effects depending upon on the targeted tissue. For instance, GH increases skeletal muscle and decreases adipose tissue mass. Our laboratory has spent the past two decades studying these effects, including the effects of GH excess and depletion, on the proteome of several mouse and human tissues. This review first discusses proteomic techniques that are commonly used for these types of studies. We then examine the proteomic differences found in mice with excess circulating GH (bGH mice) or mice with disruption of the GH receptor gene (GHR-/-). We also describe the effects of increased and decreased GH action on the proteome of adult patients with either acromegaly, GH deficiency or patients after short-term GH treatment. Finally, we explain how these proteomic studies resulted in the discovery of potential biomarkers for GH action, particularly those related with the effects of GH on aging, glucose metabolism and body composition.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA
| | - Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
20
|
Ebrahimi A, Jung MH, Dreyfuss JM, Pan H, Sgroi D, Bonner-Weir S, Weir GC. Evidence of stress in β cells obtained with laser capture microdissection from pancreases of brain dead donors. Islets 2017; 9:19-29. [PMID: 28252345 PMCID: PMC5345752 DOI: 10.1080/19382014.2017.1283083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Isolated islets used for transplantation are known to be stressed, which can result from the circumstances of death, in particular brain death, the preservation of the pancreas with its warm and cold ischemia, from the trauma of the isolation process, and the complex events that occur during tissue culture. The current study focused upon the events that occur before the islet isolation procedure. Pancreases were obtained from brain dead donors (n = 7) with mean age 50 (11) and normal pancreatic tissue obtained at surgery done for pancreatic neoplasms (n = 7), mean age 69 (9). Frozen sections were subjected to laser capture microdissection (LCM) to obtain β-cell rich islet tissue, from which extracted RNA was analyzed with microarrays. Gene expression of the 2 groups was evaluated with differential expression analysis for genes and pathways. Marked changes were found in pathways concerned with endoplasmic reticulum stress with its unfolded protein response (UPR), apoptotic pathways and components of inflammation. In addition, there were changes in genes important for islet cell identity. These findings advance our understanding of why islets are stressed before transplantation, which may lead to strategies to reduce this stress and lead to better clinical outcomes.
Collapse
Affiliation(s)
- Aref Ebrahimi
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Min-Ho Jung
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan M. Dreyfuss
- Bioinformatics Core, Joslin Diabetes Center, Research Division, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hui Pan
- Bioinformatics Core, Joslin Diabetes Center, Research Division, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dennis Sgroi
- Massachusetts General Hospital, Department of Molecular Pathology, Harvard Medical School, Boston, MA, USA
| | | | - Gordon C. Weir
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- CONTACT Gordon C. Weir, MD Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| |
Collapse
|
21
|
Kitayama Y, Fukui H, Hara K, Eda H, Kodani M, Yang M, Sun C, Yamagishi H, Tomita T, Oshima T, Watari J, Takasawa S, Miwa H. Role of regenerating gene I in claudin expression and barrier function in the small intestine. Transl Res 2016; 173:92-100. [PMID: 27055226 DOI: 10.1016/j.trsl.2016.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/17/2016] [Accepted: 03/12/2016] [Indexed: 01/01/2023]
Abstract
We have recently shown that loss of the regenerating gene (Reg) I causes susceptibility to nonsteroidal anti-inflammatory drug-induced gastrointestinal damage. However, the mechanism by which Reg I plays a protective role against this pathophysiological condition is unclear. Here, we investigated whether Reg I plays roles in the induction of tight junction proteins and mucosal barrier function in the small intestine. The small-intestinal permeability was evaluated in Reg I-deficient mice by FITC-dextran and transepithelial electrical resistance (TEER) assay. The effect of REG Iα on TEER, claudins expression, and intracellular signaling was examined using Caco2 cells in vitro. Small-intestinal expression of claudins 3 and 4 was investigated in Reg I-deficient mice in vivo. REG I deficiency significantly decreased the expression of claudin 3 in the small-intestinal epithelium. When mice were treated with indomethacin, the serum level of FITC-dextran in Reg I knockout mice was significantly higher than that in wild-type (WT) mice. The level of small-intestinal TEER was significantly decreased in Reg I knockout mice compared with WT mice under normal condition. REG Iα stimulation significantly enhanced the level of TEER in Caco2 cells. Treatment with REG Iα enhanced the expression of claudins 3 and 4 and promoted Sp1, Akt, and ERK phosphorylation in Caco2 cells, whereas these effects were attenuated by treatment with anti-REG Iα antibody. Reg I may play a role in the maintenance of mucosal barrier function by inducing tight junction proteins such as claudins 3 and 4.
Collapse
Affiliation(s)
- Yoshitaka Kitayama
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hirokazu Fukui
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.
| | - Ken Hara
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hirotsugu Eda
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Mio Kodani
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Mo Yang
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Sun
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Hidetsugu Yamagishi
- Department of Surgical and Molecular Pathology, Dokkyo University School of Medicine, Tochigi, Japan
| | - Toshihiko Tomita
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tadayuki Oshima
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Jiro Watari
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | - Hiroto Miwa
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
22
|
Xia F, Cao H, Du J, Liu X, Liu Y, Xiang M. Reg3g overexpression promotes β cell regeneration and induces immune tolerance in nonobese-diabetic mouse model. J Leukoc Biol 2015; 99:1131-40. [PMID: 26667474 DOI: 10.1189/jlb.3a0815-371rrr] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/11/2015] [Indexed: 12/21/2022] Open
Abstract
The regenerating islet-derived gene was first isolated in regenerated pancreas tissues, greatly contributing to β cell regeneration. It is an anti-inflammatory in response to cellular stress. This encouraged us to investigate the exact role of a novel member of Reg family, regenerating islet-derived gene γ, in type 1 diabetes of nonobese-diabetic mice. For this, Reg3g gene was overexpressed in pancreatic islets, and conferred beneficial effects on β cell regeneration through activating the Janus kinase 2/signal transducer and activator of transcription 3/nuclear factor κB signaling pathway. Lentiviral vector-encoding regenerating islet-derived gene γ treatment also decreased lymphocyte infiltrates of the intra-islet and peri-islet by inducing both differentiation of regulatory T cell and immature dendritic cells of tolerogenic properties, which attenuated autoimmunity. This treatment further contributed to rebalanced levels of type 1/2 helper T cell cytokines and elevated α1-antitrypsin levels in the serum. These results were not observed in phosphate-buffered saline-treated mice or in lentivirus-control mice. We have shown, for the first time, to our knowledge, that regenerating islet-derived gene γ promotes β cell regeneration and preserves β cells from autoimmunity damage by increasing regulatory T cell differentiation and inducing tolerated dendritic cells. This regenerating islet-derived gene γ infusion could probably be developed into an optimal gene therapy for the prevention and reversal of type 1 diabetes.
Collapse
Affiliation(s)
- Fei Xia
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Hui Cao
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Jiao Du
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Xiulan Liu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Yang Liu
- Synergy Innovation Center of Biological Peptide Antidiabetics of Hubei Province, School of Life Science, Wuchang University of Technology, Wuhan, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| |
Collapse
|
23
|
Nikoopour E, Bellemore SM, Singh B. IL-22, cell regeneration and autoimmunity. Cytokine 2015; 74:35-42. [PMID: 25467639 DOI: 10.1016/j.cyto.2014.09.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/18/2014] [Accepted: 09/20/2014] [Indexed: 12/16/2022]
Abstract
IL-22 as a cytokine is described with opposing pro-inflammatory and anti-inflammatory functions. Cell regeneration, tissue remodelling and balance between commensal bacteria in the gut and host immune system are considered as anti-inflammatory features of IL-22, whereas production of IL-22 from Th17 cells links this cytokine to pro-inflammatory pathways. Th17 cells and group 3 innate lymphoid cells (ILC3) are two major producers of IL-22 and both cell types express ROR-γt and Aryl hydrocarbon receptor (AhR) transcription factors. Typically, the immune system cells are the main producers of IL-22. However, targets of this cytokine are mostly non-hematopoietic cells such as hepatocytes, keratinocytes, and epithelial cells of lung and intestine. Association of IL-22 with other cytokines or transcription factors in different cell types might explain its contrasting role in health and disease. In this review we discuss the regulation of IL-22 production by AhR- and IL-23-driven pathways. A clear understanding of the biology of IL-22 will provide new opportunities for its application to improve human health involving many debilitating conditions.
Collapse
Affiliation(s)
- Enayat Nikoopour
- Centre for Human Immunology, Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Stacey M Bellemore
- Centre for Human Immunology, Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Bhagirath Singh
- Centre for Human Immunology, Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
24
|
Sun C, Fukui H, Hara K, Kitayama Y, Eda H, Yang M, Yamagishi H, Tomita T, Oshima T, Watari J, Takasawa S, Chiba T, Miwa H. Expression of Reg family genes in the gastrointestinal tract of mice treated with indomethacin. Am J Physiol Gastrointest Liver Physiol 2015; 308:G736-44. [PMID: 25747353 DOI: 10.1152/ajpgi.00362.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/19/2015] [Indexed: 01/31/2023]
Abstract
Regenerating gene (Reg) family proteins, which are classified into four types, commonly act as trophic and/or antiapoptotic factors in gastrointestinal (GI) diseases. However, it remains unclear how these proteins coordinate their similar roles under such pathophysiological conditions. Here, we investigated the interrelationships of Reg family gene expression with mucosal cell proliferation and apoptosis in nonsteroidal anti-inflammatory drug (NSAID)-induced GI injury. GI injury was induced by subcutaneous injection of indomethacin into Reg I knockout (KO) and wild-type (WT) mice, and its severity was scored histopathologically. Temporal changes in the expression of Reg family genes, mucosal proliferation, and apoptosis were evaluated throughout the GI tract by real-time RT-PCR, Ki-67 immunoreactivity, and TUNEL assay, respectively. Reg I, Reg III family, and Reg IV were predominantly expressed in the upper, middle, and lower GI mucosa, respectively. Expression of Reg I and Reg III family genes was upregulated in specific portions of the GI tract after indomethacin treatment. Ki-67-positive epithelial cells were significantly decreased in the gastric and small-intestinal mucosa of Reg I KO mice under normal conditions. After treatment with indomethacin, the number of TUNEL-positive cells was significantly greater throughout the GI mucosa in Reg I KO mice than in WT mice. Expression of Reg I was independent of that of other Reg family genes in, not only normal GI tissues, but also indomethacin-induced GI lesions. Members of the Reg gene family show distinct profiles of expression in the GI tract, and Reg I independently plays a role in protecting the GI mucosa against NSAID-induced injury.
Collapse
Affiliation(s)
- Chao Sun
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Hirokazu Fukui
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan;
| | - Ken Hara
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yoshitaka Kitayama
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hirotsugu Eda
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Mo Yang
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Hidetsugu Yamagishi
- Department of Surgical and Molecular Pathology, Dokkyo University School of Medicine, Tochigi, Japan
| | - Toshihiko Tomita
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tadayuki Oshima
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Jiro Watari
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroto Miwa
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
25
|
Yamauchi A, Itaya-Hironaka A, Sakuramoto-Tsuchida S, Takeda M, Yoshimoto K, Miyaoka T, Fujimura T, Tsujinaka H, Tsuchida C, Ota H, Takasawa S. Synergistic activations of REG I α and REG I β promoters by IL-6 and Glucocorticoids through JAK/STAT pathway in human pancreatic β cells. J Diabetes Res 2015; 2015:173058. [PMID: 25767811 PMCID: PMC4342170 DOI: 10.1155/2015/173058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/26/2015] [Indexed: 12/31/2022] Open
Abstract
Reg (Regenerating gene) gene was originally isolated from rat regenerating islets and its encoding protein was revealed as an autocrine/paracrine growth factor for β cells. Rat Reg gene is activated in inflammatory conditions for β cell regeneration. In human, although five functional REG family genes (REG Iα, REG Iβ, REG III, HIP/PAP, and REG IV) were isolated, their expressions in β cells under inflammatory conditions remained unclear. In this study, we found that combined addition of IL-6 and dexamethasone (Dx) induced REG Iα and REG Iβ expression in human 1.1B4 β cells. Promoter assay revealed that a signal transducer and activator of transcription- (STAT-) binding site in each promoter of REG Iα (TGCCGGGAA) and REG Iβ (TGCCAGGAA) was essential for the IL-6+Dx-induced promoter activation. A Janus kinase 2 (JAK2) inhibitor significantly inhibited the IL-6+Dx-induced REG Iα and REG Iβ transcription. Electrophoretic mobility shift assay and chromatin immunoprecipitation revealed that IL-6+Dx stimulation increased STAT3 binding to the REG Iα promoter. Furthermore, small interfering RNA-mediated targeting of STAT3 blocked the IL-6+Dx-induced expression of REG Iα and REG Iβ. These results indicate that the expression of REG Iα and REG Iβ should be upregulated in human β cells under inflammatory conditions through the JAK/STAT pathway.
Collapse
Affiliation(s)
- Akiyo Yamauchi
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | | | | | - Maiko Takeda
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Kiyomi Yoshimoto
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Tomoko Miyaoka
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Takanori Fujimura
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiroki Tsujinaka
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Chikatsugu Tsuchida
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiroyo Ota
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
26
|
Uppal SS, Naveed AK, Baig S, Chaudhry B. Expression of REG Iα gene in type 2 diabetics in Pakistan. Diabetol Metab Syndr 2015; 7:96. [PMID: 26568772 PMCID: PMC4643495 DOI: 10.1186/s13098-015-0092-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 11/02/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The escalating rate of diabetes' has prompted researchers around the world to explore for early markers. A deficit of functional β-cell mass plays a central role in the pathophysiology of type 2 diabetes. The REG (Regenerating) gene, encoding a 166 amino acid REG protein was discovered in rats and humans which is released in response to β-cells damage and play a role in their regeneration. The objective of this study was to characterize serum levels of REG Iα proteins in type 2 diabetic patients as indicator of β-cell apoptosis as well as regeneration. METHODS Unrelated type 2 diabetic patients (n = 55) of different age groups and disease duration were recruited from the Medical OPD of PNS Shifa Hospital. Age and sex matched non diabetic controls (n = 20) without family history of diabetes were selected from the same setting. Demographical details were recorded on a structured questionnaire. Biochemical parameters like FBG, HbA1c, TC and TG levels were measured. Serum levels of REG Iα protein were determined by ELISA. RESULTS Levels of REG Iα protein were found significantly raised in type 2 diabetic patients compared to controls (p < 001). Patients with short duration of the disease had higher levels of REG Iα as compared to patients with longer duration of the disease. Although the patients were on anti hyperglycemic agents, a positive correlation was found between REG Iα serum levels, FBG and HbA1c levels. Patients with higher BMI had higher levels of serum REG Iα levels. Serum TC, TG and Hb levels showed no correlation. CONCLUSION REG Iα may be used as a marker/predictor of type 2 diabetes especially in the early stages of the disease.
Collapse
Affiliation(s)
- Sadaf Saleem Uppal
- />Department of Biochemistry and Molecular Biology, Army Medical College, Rawalpindi and National University of Science and Technology, Islamabad, Pakistan
| | - Abdul Khaliq Naveed
- />Department of Biochemistry, Islamic International Medical College, Riphah International University, Islamabad, Pakistan
| | - Saeeda Baig
- />Department of Biochemistry, Ziauddin University, Karachi, Pakistan
| | - Bushra Chaudhry
- />Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
27
|
Fu W, Farache J, Clardy SM, Hattori K, Mander P, Lee K, Rioja I, Weissleder R, Prinjha RK, Benoist C, Mathis D. Epigenetic modulation of type-1 diabetes via a dual effect on pancreatic macrophages and β cells. eLife 2014; 3:e04631. [PMID: 25407682 PMCID: PMC4270084 DOI: 10.7554/elife.04631] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022] Open
Abstract
Epigenetic modifiers are an emerging class of anti-tumor drugs, potent in multiple cancer contexts. Their effect on spontaneously developing autoimmune diseases has been little explored. We report that a short treatment with I-BET151, a small-molecule inhibitor of a family of bromodomain-containing transcriptional regulators, irreversibly suppressed development of type-1 diabetes in NOD mice. The inhibitor could prevent or clear insulitis, but had minimal influence on the transcriptomes of infiltrating and circulating T cells. Rather, it induced pancreatic macrophages to adopt an anti-inflammatory phenotype, impacting the NF-κB pathway in particular. I-BET151 also elicited regeneration of islet β-cells, inducing proliferation and expression of genes encoding transcription factors key to β-cell differentiation/function. The effect on β cells did not require T cell infiltration of the islets. Thus, treatment with I-BET151 achieves a 'combination therapy' currently advocated by many diabetes investigators, operating by a novel mechanism that coincidentally dampens islet inflammation and enhances β-cell regeneration.
Collapse
Affiliation(s)
- Wenxian Fu
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| | - Julia Farache
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| | - Susan M Clardy
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Kimie Hattori
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| | - Palwinder Mander
- Epinova DPU, Immuno-Inflammation Therapy Area, Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | - Kevin Lee
- Epinova DPU, Immuno-Inflammation Therapy Area, Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | - Inmaculada Rioja
- Epinova DPU, Immuno-Inflammation Therapy Area, Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Rab K Prinjha
- Epinova DPU, Immuno-Inflammation Therapy Area, Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
28
|
Aida K, Saitoh S, Nishida Y, Yokota S, Ohno S, Mao X, Akiyama D, Tanaka S, Awata T, Shimada A, Oikawa Y, Shimura H, Furuya F, Takizawa S, Ichijo M, Ichijo S, Itakura J, Fujii H, Hashiguchi A, Takasawa S, Endo T, Kobayashi T. Distinct cell clusters touching islet cells induce islet cell replication in association with over-expression of Regenerating Gene (REG) protein in fulminant type 1 diabetes. PLoS One 2014; 9:e95110. [PMID: 24759849 PMCID: PMC3997392 DOI: 10.1371/journal.pone.0095110] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/23/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Pancreatic islet endocrine cell-supporting architectures, including islet encapsulating basement membranes (BMs), extracellular matrix (ECM), and possible cell clusters, are unclear. PROCEDURES The architectures around islet cell clusters, including BMs, ECM, and pancreatic acinar-like cell clusters, were studied in the non-diabetic state and in the inflamed milieu of fulminant type 1 diabetes in humans. RESULT Immunohistochemical and electron microscopy analyses demonstrated that human islet cell clusters and acinar-like cell clusters adhere directly to each other with desmosomal structures and coated-pit-like structures between the two cell clusters. The two cell-clusters are encapsulated by a continuous capsule composed of common BMs/ECM. The acinar-like cell clusters have vesicles containing regenerating (REG) Iα protein. The vesicles containing REG Iα protein are directly secreted to islet cells. In the inflamed milieu of fulminant type 1 diabetes, the acinar-like cell clusters over-expressed REG Iα protein. Islet endocrine cells, including beta-cells and non-beta cells, which were packed with the acinar-like cell clusters, show self-replication with a markedly increased number of Ki67-positive cells. CONCLUSION The acinar-like cell clusters touching islet endocrine cells are distinct, because the cell clusters are packed with pancreatic islet clusters and surrounded by common BMs/ECM. Furthermore, the acinar-like cell clusters express REG Iα protein and secrete directly to neighboring islet endocrine cells in the non-diabetic state, and the cell clusters over-express REG Iα in the inflamed milieu of fulminant type 1 diabetes with marked self-replication of islet cells.
Collapse
Affiliation(s)
- Kaoru Aida
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Sei Saitoh
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Yoriko Nishida
- Department of Nursing, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Sadanori Yokota
- Section of Functional Morphology, Faculty of Pharmaceutical Sciences, Nagasaki International University, Saseho, Nagasaki, Japan
| | - Shinichi Ohno
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Xiayang Mao
- Department of Computer Science, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Daiichiro Akiyama
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Shoichiro Tanaka
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Takuya Awata
- Division of Endocrinology and Diabetes, Department of Medicine, Saitama Medical School, Moroyama, Saitama, Japan
| | - Akira Shimada
- Department of Internal Medicine, Saiseikai Central Hospital, Tokyo, Japan
| | - Youichi Oikawa
- Department of Internal Medicine, Saiseikai Central Hospital, Tokyo, Japan
| | - Hiroki Shimura
- Department of Laboratory Medicine, Fukushima Medical University, Fukushima, Fukushima, Japan
| | - Fumihiko Furuya
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Soichi Takizawa
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Masashi Ichijo
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Sayaka Ichijo
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Jun Itakura
- Department of Surgery I, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hideki Fujii
- Department of Surgery I, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Akinori Hashiguchi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara, Wakayama, Japan
| | - Toyoshi Endo
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Tetsuro Kobayashi
- Department of Internal Medicine III, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
- * E-mail:
| |
Collapse
|
29
|
Calderari S, Irminger JC, Giroix MH, Ehses JA, Gangnerau MN, Coulaud J, Rickenbach K, Gauguier D, Halban P, Serradas P, Homo-Delarche F. Regenerating 1 and 3b gene expression in the pancreas of type 2 diabetic Goto-Kakizaki (GK) rats. PLoS One 2014; 9:e90045. [PMID: 24587207 PMCID: PMC3936001 DOI: 10.1371/journal.pone.0090045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023] Open
Abstract
Regenerating (REG) proteins are associated with islet development, β-cell damage, diabetes and pancreatitis. Particularly, REG-1 and REG-3-beta are involved in cell growth/survival and/or inflammation and the Reg1 promoter contains interleukin-6 (IL-6)-responsive elements. We showed by transcriptome analysis that islets of Goto-Kakizaki (GK) rats, a model of spontaneous type 2 diabetes, overexpress Reg1, 3α, 3β and 3γ, vs Wistar islets. Goto-Kakizaki rat islets also exhibit increased cytokine/chemokine expression/release, particularly IL-6. Here we analyzed Reg1 and Reg3β expression and REG-1 immuno-localization in the GK rat pancreas in relationship with inflammation. Isolated pancreatic islets and acinar tissue from male adult Wistar and diabetic GK rats were used for quantitative RT-PCR analysis. REG-1 immunohistochemistry was performed on paraffin sections with a monoclonal anti-rat REG-1 antibody. Islet cytokine/chemokine release was measured after 48 h-culture. Islet macrophage-positive area was quantified on cryostat sections using anti-CD68 and major histocompatibility complex (MHC) class II antibodies. Pancreatic exocrine-to-endocrine Reg1 and Reg3β mRNA ratios were markedly increased in Wistar vs GK rats. Conversely, both genes were upregulated in isolated GK rat islets. These findings were unexpected, because Reg genes are expressed in the pancreatic acinar tissue. However, we observed REG-1 protein labeling in acinar peri-ductal tissue close to islets and around large, often disorganized, GK rat islets, which may retain acinar cells due to their irregular shape. These large islets also showed peri-islet macrophage infiltration and increased release of various cytokines/chemokines, particularly IL-6. Thus, IL-6 might potentially trigger acinar REG-1 expression and secretion in the vicinity of large diabetic GK rat islets. This increased acinar REG-1 expression might reflect an adaptive though unsuccessful response to deleterious microenvironment.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chemokines/blood
- Chemokines/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Regulation
- Islets of Langerhans/metabolism
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lithostathine/genetics
- Lithostathine/metabolism
- Macrophages/metabolism
- Male
- Pancreatitis-Associated Proteins
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Sophie Calderari
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Jean-Claude Irminger
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Marie-Hélène Giroix
- Equipe associée au Centre National de la Recherche Scientifique (CNRS) 4413-Unité de Biologie Fonctionnelle et Adaptative (BFA), Team 1 (Biologie et Pathologie du Pancréas Endocrine (B2PE)), Université Paris-Diderot Sorbonne-Paris-Cité, Paris, France
| | - Jan A. Ehses
- Department of Surgery, Faculty of Medicine, University of British Columbia and Child and Family Research Institute, Vancouver, BC, Canada
| | - Marie-Noëlle Gangnerau
- Equipe associée au Centre National de la Recherche Scientifique (CNRS) 4413-Unité de Biologie Fonctionnelle et Adaptative (BFA), Team 1 (Biologie et Pathologie du Pancréas Endocrine (B2PE)), Université Paris-Diderot Sorbonne-Paris-Cité, Paris, France
| | - Josiane Coulaud
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Katharina Rickenbach
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Dominique Gauguier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Philippe Halban
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Patricia Serradas
- INSERM UMRS 872, Team 9, CRC, Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Françoise Homo-Delarche
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
- * E-mail:
| |
Collapse
|
30
|
Coffey R, Nam H, Knutson MD. Microarray analysis of rat pancreas reveals altered expression of Alox15 and regenerating islet-derived genes in response to iron deficiency and overload. PLoS One 2014; 9:e86019. [PMID: 24465846 PMCID: PMC3897611 DOI: 10.1371/journal.pone.0086019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/04/2013] [Indexed: 12/27/2022] Open
Abstract
It is well known that iron overload can result in pancreatic iron deposition, beta-cell destruction, and diabetes in humans. Recent studies in animals have extended the link between iron status and pancreatic function by showing that iron depletion confers protection against beta-cell dysfunction and diabetes. The aim of the present study was to identify genes in the pancreas that are differentially expressed in response to iron deficiency or overload. Weanling male Sprague-Dawley rats (n = 6/group) were fed iron-deficient, iron-adequate, or iron-overloaded diets for 3 weeks to alter their iron status. Total RNA was isolated from the pancreases and pooled within each group for microarray analyses in which gene expression levels were compared to those in iron-adequate controls. In iron-deficient pancreas, a total of 66 genes were found to be differentially regulated (10 up, 56 down), whereas in iron-overloaded pancreas, 164 genes were affected (82 up, 82 down). The most up-regulated transcript in iron-deficient pancreas was arachidonate 15-lipoxygenase (Alox15), which has been implicated in the development of diabetes. In iron-overloaded pancreas, the most upregulated transcripts were Reg1a, Reg3a, and Reg3b belonging to the regenerating islet-derived gene (Reg) family. Reg expression has been observed in response to pancreatic stress and is thought to facilitate pancreatic regeneration. Subsequent qRT-PCR validation indicated that Alox15 mRNA levels were 4 times higher in iron-deficient than in iron-adequate pancreas and that Reg1a, Reg3a, and Reg3b mRNA levels were 17–36 times higher in iron-overloaded pancreas. The elevated Alox15 mRNA levels in iron-deficient pancreas were associated with 8-fold higher levels of Alox15 protein as indicated by Western blotting. Overall, these data raise the possibility that Reg expression may serve as a biomarker for iron-related pancreatic stress, and that iron deficiency may adversely affect the risk of developing diabetes through up-regulation of Alox15.
Collapse
Affiliation(s)
- Richard Coffey
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, United States of America
| | - Hyeyoung Nam
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, United States of America
| | - Mitchell D. Knutson
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
31
|
Nandan MO, Ghaleb AM, Liu Y, Bialkowska AB, McConnell BB, Shroyer KR, Robine S, Yang VW. Inducible intestine-specific deletion of Krüppel-like factor 5 is characterized by a regenerative response in adult mouse colon. Dev Biol 2014; 387:191-202. [PMID: 24440658 DOI: 10.1016/j.ydbio.2014.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 01/01/2014] [Accepted: 01/03/2014] [Indexed: 01/29/2023]
Abstract
Krüppel-like factor 5 (KLF5) is a pro-proliferative transcriptional regulator primarily expressed in the intestinal crypt epithelial cells. Constitutive intestine-specific deletion of Klf5 is neonatal lethal suggesting a crucial role for KLF5 in intestinal development and homeostasis. We have previously shown Klf5 to play an active role regulating intestinal tumorigenesis. Here we examine the effect of inducible intestine-specific deletion of Klf5 in adult mice. Klf5 is lost from the intestine beginning at day 3 after the start of a 5-day treatment with the inducer tamoxifen. Although the mice have no significant weight loss or lethality, the colonic tissue shows signs of epithelial distress starting at day 3 following induction. Accompanying the morphological changes is a significant loss of proliferative crypt epithelial cells as revealed by BrdU or Ki67 staining at days 3 and 5 after start of tamoxifen. We also observed a loss of goblet cells from the colon and Paneth cells from the small intestine upon induced deletion of Klf5. In addition, loss of Klf5 from the colonic epithelium is accompanied by a regenerative response that coincides with an expansion in the zone of Sox9 expression along the crypt axis. At day 11, both proliferation and Sox9 expression return to baseline levels. Microarray and quantitative PCR analyses reveal an up-regulation of several regeneration-associated genes (Reg1A, Reg3G and Reg3B) and down-regulation of many Klf5 targets (Ki-67, cyclin B, Cdc2 and cyclin D1). Sox9 and Reg1A protein levels are also increased upon Klf5 loss. Lentiviral-mediated knockdown of KLF5 and exogenous expression of KLF5 in colorectal cancer cell lines confirm that Sox9 expression is negatively regulated by KLF5. Furthermore, ChIP assays reveal a direct association of KLF5 with both the Sox9 and Reg1A promoters. We have shown that disruption of epithelial homeostasis due to Klf5 loss from the adult colon is followed by a regenerative response led by Sox9 and the Reg family of proteins. Our study demonstrates that adult mouse colonic tissue undergoes acute physiological changes to accommodate the loss of Klf5 withstanding epithelial damage further signifying importance of Klf5 in colonic homeostasis.
Collapse
Affiliation(s)
- Mandayam O Nandan
- Department of Medicine, Stony Brook University School of Medicine, HSC-T16 Room 020, Stony Brook, NY 11794, USA.
| | - Amr M Ghaleb
- Department of Medicine, Stony Brook University School of Medicine, HSC-T16 Room 020, Stony Brook, NY 11794, USA.
| | - Yang Liu
- Department of Medicine, Stony Brook University School of Medicine, HSC-T16 Room 020, Stony Brook, NY 11794, USA.
| | - Agnieszka B Bialkowska
- Department of Medicine, Stony Brook University School of Medicine, HSC-T16 Room 020, Stony Brook, NY 11794, USA.
| | - Beth B McConnell
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA.
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University School of Medicine, BST-9, Stony Brook Medicine, Stony Brook, NY 11794-8691, USA.
| | - Sylvie Robine
- Department of Morphogenesis and Intracellular Signalling, Institut Curie-CNRS, Paris, France.
| | - Vincent W Yang
- Department of Medicine, Stony Brook University School of Medicine, HSC-T16 Room 020, Stony Brook, NY 11794, USA.
| |
Collapse
|
32
|
Goosse K, Bouckenooghe T, Sisino G, Aurientis S, Remacle C, Reusens B. Increased susceptibility to streptozotocin and impeded regeneration capacity of beta-cells in adult offspring of malnourished rats. Acta Physiol (Oxf) 2014; 210:99-109. [PMID: 23701924 DOI: 10.1111/apha.12121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/22/2013] [Accepted: 05/16/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Epidemiological studies related poor maternal nutrition and subsequent growth retardation in the progeny to the development of diabetes later in life. Low-protein diet during gestation altered the beta-cell development of the rat progeny by decreasing beta-cell proliferation and increasing their sensitivity to nitric oxide and cytokines in the foetus. This disturbed maternal environment had long-lasting consequences because the higher beta-cell vulnerability was maintained at adulthood. AIM The aim of this study was to determine whether early malnutrition influences the vulnerability and the regeneration capacity of beta-cells after streptozotocin (STZ) damage at adulthood. METHODS Gestating rats were fed either a control or a low-protein diet until weaning. Adult female offspring received injections of Freund's adjuvant weekly for 5 weeks followed 24 h later by STZ. Half of the cohort was killed at d34, whereas the other half was maintained until d48 to analyse the regeneration capacity of the beta-cells. RESULTS Although control and low-protein rats had equivalent pancreatic insulin content and beta-cell volume density at d34, hyperglycaemia appeared earlier and was more dramatic in low-protein rats than in control rats. STZ treatment increased beta-cell proliferation similarly in both groups. At d48, apoptotic rate was higher in the low-protein group. Regeneration appeared in control, but not in the low-protein rats, where beta-cell aggregates/surface area and Reg1-positive area were decreased compared to control. CONCLUSION Maternal malnutrition programmes a more vulnerable endocrine pancreas in the progeny which is unable to regenerate after injury, therefore predisposing it to develop glucose intolerance and diabetes later in life.
Collapse
Affiliation(s)
- K. Goosse
- Laboratory of Cell Biology; Université catholique de Louvain; Louvain-la-Neuve Belgium
| | - T. Bouckenooghe
- Laboratory of Cell Biology; Université catholique de Louvain; Louvain-la-Neuve Belgium
- EA 4489 “Environnement périnatal et croissance”; Faculté de Médecine; H Warembourg; Lille France
| | - G. Sisino
- EA 4489 “Environnement périnatal et croissance”; Faculté de Médecine; H Warembourg; Lille France
| | - S. Aurientis
- EA 4489 “Environnement périnatal et croissance”; Faculté de Médecine; H Warembourg; Lille France
| | - C. Remacle
- Laboratory of Cell Biology; Université catholique de Louvain; Louvain-la-Neuve Belgium
| | - B. Reusens
- Laboratory of Cell Biology; Université catholique de Louvain; Louvain-la-Neuve Belgium
| |
Collapse
|
33
|
Abstract
INTRODUCTION Major trauma still represents one of the leading causes of death in the first four decades of life. Septic complications represent the predominant causes of late death (45% of overall mortality) in polytrauma patients. The ability of clinicians to early differentiate between systemic inflammatory response syndrome (SIRS) and sepsis is demonstrated to improve clinical outcome and mortality. The identification of an "ideal" biomarker able to early recognize incoming septic complications in trauma patients is still a challenge for researchers. AIM To evaluate the existing evidence regarding the role of biomarkers to predict or facilitate early diagnosis of sepsis in trauma patients, trying to compile some recommendations for the clinical setting. METHODS An Internet-based search of the MEDLINE, EMBASE and Cochrane Library databases was performed using the search terms: "Biomarkers", "Sepsis" and "Trauma" in various combinations. The methodological quality of the included studies was assessed using the Quality Assessment of Diagnostic Accuracy Studies Checklist (QUADAS). After data extraction, the level of evidence available for each bio-marker was rated and presented using the "best-evidence synthesis" method, in line with the US Agency for Healthcare Research and Quality. RESULTS Thirty studies were eligible for the final analysis: 13 case-control studies and 17 cohort studies. The "strong evidence" available demonstrated the potential use of procalcitonin as an early indicator of post-traumatic septic complications and reported the inability of c-reactive protein (CRP) to specifically identify infective complications. Moderate, conflicting and limited evidence are available for the other 31 biomarkers. CONCLUSION Several biomarkers have been evaluated for predicting or making early diagnosis of sepsis in trauma patients. Current evidence does not support the use of a single biomarker in diagnosing sepsis. However, procalcitonin trend was found to be useful in early identification of post-traumatic septic course and its use is suggested (Recommendation Grade: B) in clinical practice.
Collapse
|
34
|
Nakagawa K, Takasawa S, Nata K, Yamauchi A, Itaya-Hironaka A, Ota H, Yoshimoto K, Sakuramoto-Tsuchida S, Miyaoka T, Takeda M, Unno M, Okamoto H. Prevention of Reg I-induced β-cell apoptosis by IL-6/dexamethasone through activation of HGF gene regulation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2988-2995. [DOI: 10.1016/j.bbamcr.2013.08.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 08/05/2013] [Accepted: 08/07/2013] [Indexed: 12/12/2022]
|
35
|
Expression of Ins1 and Ins2 genes in mouse fetal liver. Cell Tissue Res 2013; 355:303-14. [PMID: 24258027 DOI: 10.1007/s00441-013-1741-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 10/09/2013] [Indexed: 10/26/2022]
Abstract
A possible cure for diabetes is explored by using non-pancreatic cells such as fetal hepatocytes. The expression of insulin and transcription factors for insulin is investigated in mouse fetal liver. We detected mRNAs for insulin I (Ins1) and insulin II (Ins2) and proinsulin- and mature insulin-positive cells in mouse fetal liver by reverse transcription plus the polymerase chain reaction and immunohistochemistry. Glucagon, somatostatin and pancreatic polypeptide were not expressed throughout development. Mouse Ins2 and Ins1 promoters were transiently activated in mouse fetal hepatocytes of embryonic days 13.5 and 16.5, respectively. Pancreatic and duodenal homeobox 1 (Pdx1) mRNA was not expressed during development of the liver. In contrast, mRNAs and proteins of neurogenic differentiation (NeuroD)/β cell E-box transactivator 2 (Beta2) and v-maf musculoaponeurotic fibrosarcoma oncogene homolog (MafA) were almost simultaneously expressed with insulin genes in the liver. Ins2 and Ins1 promoters were activated in hepatoma cells by the transfection of the expression vector for NeuroD/Beta2 alone and by the combination of NeuroD/Beta2 and MafA, respectively. These results indicate that the expression of NeuroD/Beta2 and MafA is linked temporally with the transcription of Ins2 and Ins1 genes in mouse fetal liver and suggest the potential usage of fetal hepatocytes to make insulin-producing β cells by introducing transcription factors.
Collapse
|
36
|
Abstract
The regenerating gene (Reg) family is a group of small molecules that includes four members found in various species, although only three are found in human tissues. Their expression is stimulated by certain growth factors or cytokines. The Reg family plays different roles in proliferation, migration, and anti-apoptosis through activating different signaling pathways. Their dysexpression is closely associated with a number of human conditions and diseases such as inflammation and cancer, especially in the human digestive system. Clinically, upregulation of Reg proteins is usually demonstrated in histological sections and sera from cancer patients. Therefore, Reg proteins can predict the progression and prognosis of cancers, especially those of the digestive tract, and can also act as diagnostic markers and therapeutic targets.
Collapse
|
37
|
Pancreatic β cell proliferation by intermittent hypoxia via up-regulation of Reg family genes and HGF gene. Life Sci 2013; 93:664-72. [DOI: 10.1016/j.lfs.2013.09.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 07/30/2013] [Accepted: 09/05/2013] [Indexed: 11/19/2022]
|
38
|
Cox AR, Beamish CA, Carter DE, Arany EJ, Hill DJ. Cellular mechanisms underlying failed beta cell regeneration in offspring of protein-restricted pregnant mice. Exp Biol Med (Maywood) 2013; 238:1147-59. [PMID: 23986224 DOI: 10.1177/1535370213493715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Low birth weight and poor foetal growth following low protein (LP) exposure are associated with altered islet development and glucose intolerance in adulthood. Additionally, LP-fed offspring fail to regenerate their β-cells following depletion with streptozotocin (STZ) in contrast to control-fed offspring that restore β-cell mass. Our objective was to identify signalling pathways and cellular functions that may be critically altered in LP offspring rendering them susceptible to developing long-term glucose intolerance and decreased β-cell plasticity. Pregnant Balb/c mice were fed a control (C; 20% protein) or an isocaloric LP (8% protein) diet throughout gestation and C diet thereafter. Female offspring were injected intraperitoneally with 35 mg/kg STZ or vehicle on days 1 to 5 for each dietary treatment. At 30 days of age, total RNA was extracted from pancreatic tissue for microarray analysis using the Affymetrix GeneChip Mouse Genome 430 2.0. Gene and protein expression were quantified from isolated islets. Finally, β-cell proliferation was determined in vitro following REG1α treatment. The microarray data and GO enrichment analysis indicated that foetal protein restriction alters the early expression of genes necessary for many cell functions, such as oxidative phosphorylation and free radical scavenging. Expression of Reg1 was upregulated following STZ, whereas protein content was decreased in LP + STZ islets. Furthermore, REG1α failed to stimulate β-cell proliferation in vitro in LP + STZ islets. Therefore, early nutritional insults may programme the Reg1 pathway resulting in a limited ability to increase β-cell mass during metabolic stress. In conclusion, this study implicates the Reg1 pathway in β-cell regeneration and describes altered programming of gene expression in LP offspring, which underlies later development of cell dysfunction and glucose intolerance in adulthood.
Collapse
Affiliation(s)
- Aaron R Cox
- Lawson Health Research Institute, St. Joseph's Health Care, London, Ontario, Canada, N6A 4V2
| | | | | | | | | |
Collapse
|
39
|
Jia J, Liu X, Chen Y, Zheng X, Tu L, Huang X, Wang X. Establishment of a pancreatic β cell proliferation model in vitro and a platform for diabetes drug screening. Cytotechnology 2013; 66:687-97. [PMID: 23979319 DOI: 10.1007/s10616-013-9622-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/19/2013] [Indexed: 01/13/2023] Open
Abstract
Diabetes, a disease resulting from loss of functional β cells, is globally an increasingly important condition. Based on the islet-differentiation ability of ductal epithelial cells and stimulating β cell proliferation ability of the Reg Iα gene, we aimed to establish an in vitro pancreatic β cell proliferation model for screening therapeutic drugs of diabetes in the future. Pancreatic ductal epithelial cells were isolated from male Wistar rats, and induced to differentiate into pancreatic β cells. Immunofluorescence staining assay, western blot, RT-PCR analysis, and dithizone staining were used to characterize the cells. Rat Reg Iα protein was transiently expressed in vitro by transfection of HEK 293 cells with the PCMV6-entry-REG Ia plasmid, and expression was verified by RT-PCR analysis, proliferation assay, and apoptosis assay. The pancreatic β cell proliferation model was further validated by a proliferation assay using differentiated pancreatic β cells treated with transfection supernatant. Finally, we have successfully established an in vitro pancreatic β cells proliferation model using transiently expressed rat Reg Iα protein and differentiated pancreatic β cells from pancreatic ductal epithelial cells. This model could be used as a platform to screen new drugs for islet neogenesis to cure diabetes, especially Chinese herbal drugs in the future.
Collapse
Affiliation(s)
- Jing Jia
- Center for Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, Zhejiang Province, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
MASUI TAKASHI, OTA ICHIRO, ITAYA-HIRONAKA ASAKO, TAKEDA MAIKO, KASAI TAKAHIKO, YAMAUCHI AKIYO, SAKURAMOTO-TSUCHIDA SUMIYO, MIKAMI SHINJI, YANE KATSUNARI, TAKASAWA SHIN, HOSOI HIROSHI. Expression of REG III and prognosis in head and neck cancer. Oncol Rep 2013; 30:573-8. [DOI: 10.3892/or.2013.2521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/21/2013] [Indexed: 11/06/2022] Open
|
41
|
The involvement of interleukin-22 in the expression of pancreatic beta cell regenerative Reg genes. CELL REGENERATION 2013; 2:2. [PMID: 25408874 PMCID: PMC4230743 DOI: 10.1186/2045-9769-2-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 04/02/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND In Type 1 diabetes, the insulin-producing β-cells within the pancreatic islets of Langerhans are destroyed. We showed previously that immunotherapy with Bacillus Calmette-Guerin (BCG) or complete Freund's adjuvant (CFA) of non-obese diabetic (NOD) mice can prevent disease process and pancreatic β-cell loss. This was associated with increased islet Regenerating (Reg) genes expression, and elevated IL-22-producing Th17 T-cells in the pancreas. RESULTS We hypothesized that IL-22 was responsible for the increased Reg gene expression in the pancreas. We therefore quantified the Reg1, Reg2, and Reg3δ (INGAP) mRNA expression in isolated pre-diabetic NOD islets treated with IL-22. We measured IL-22, and IL-22 receptor(R)-α mRNA expression in the pancreas and spleen of pre-diabetic and diabetic NOD mice. Our results showed: 1) Reg1 and Reg2 mRNA abundance to be significantly increased in IL-22-treated islets in vitro; 2) IL-22 mRNA expression in the pre-diabetic mouse pancreas increased with time following CFA treatment; 3) a reduced expression of IL-22Rα following CFA treatment; 4) a down-regulation in Reg1 and Reg2 mRNA expression in the pancreas of pre-diabetic mice injected with an IL-22 neutralizing antibody; and 5) an increased islet β-cell DNA synthesis in vitro in the presence of IL-22. CONCLUSIONS We conclude that IL-22 may contribute to the regeneration of β-cells by up-regulating Regenerating Reg1 and Reg2 genes in the islets.
Collapse
|
42
|
Abstract
Celiac disease (CD) is an autoimmune disorder, which damages the small intestine and is caused by ingestion of gluten in genetically susceptible individuals. The only known effective treatment is a lifelong gluten-free diet. Genetic risk factors have been identified and nearly all patients are HLA-DQ2 and/or HLA-DQ8 positive. Specific autoantibodies, IgA antitissue transglutaminase-2, antiendomysium, and antideaminated forms of gliadin peptide antibodies, are widely used as diagnostic aids in celiac patients. However, the discovery of new biomarkers may help in the diagnosis and follow-up of the disease. Recently, the molecule REG Iα, involved in tissue regeneration, has been proposed as a new biomarker of CD. REG Iα expression is increased in the target tissue and in the sera of celiac patients during damage and inflammation, decreasing after gluten-free diet. In this article we review the main biomarkers for diagnosis and monitoring of CD, focusing on the immune response-related mechanisms.
Collapse
|
43
|
Luo C, Li B, Liu L, Yin HP, Wang M, Liu JL. Transcriptional activation of Reg2 and Reg3β genes by glucocorticoids and interleukin-6 in pancreatic acinar and islet cells. Mol Cell Endocrinol 2013; 365:187-96. [PMID: 23147030 DOI: 10.1016/j.mce.2012.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/21/2012] [Accepted: 10/18/2012] [Indexed: 12/27/2022]
Abstract
Reg family proteins are expressed in the pancreas and involved in pancreatitis and islet β-cell growth. In order to explore transcriptional control, we transfected luciferase reporter genes driven by Reg promoters into acinar AR42J and islet MIN6 cells. Dexamethasone (DEX) significantly increased the promoter expression of Reg2 and Reg3β genes and the levels of endogenous Reg3β mRNA and protein in AR42J cells. DEX-induced promoter activation was inhibited by the inhibitor of poly(ADP-ribose) polymerase, nicotinamide. In MIN6 cells, DEX moderately stimulated the transcription of Reg3β but not Reg2 promoter. While IL-6 alone had no effect, coculture with DEX produced a remarkable synergism on Reg3β gene transcription, which was abolished by nicotinamide. Our results demonstrated a significant and direct stimulation of Reg2 and Reg3β genes by glucocorticoids, all three were activated in response to inflammation such as in pancreatitis. Prominent stimulation of specific Reg genes by glucocorticoids may constitute a functional synergism.
Collapse
Affiliation(s)
- Chen Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China; Fraser Laboratories for Diabetes Research, Department of Medicine, McGill University Health Centre, Montreal, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
β-Cell dysfunction is a critical component in the development of type 2 diabetes. Whilst both genetic and environmental factors contribute to the development of the disease, relatively little is known about the molecular network that is responsible for diet-induced functional changes in pancreatic β-cells. Recent genome-wide association studies for diabetes-related traits have generated a large number of candidate genes that constitute possible links between dietary factors and the genetic susceptibility for β-cell failure. Here, we summarize recent approaches for identifying nutritionally regulated transcripts in islets on a genome-wide scale. Polygenic mouse models for type 2 diabetes have been instrumental for investigating the mechanism of diet-induced β-cell dysfunction. Enhanced oxidative metabolism, triggered by a combination of dietary carbohydrates and fat, appears to play a critical role in the pathophysiology of diet-induced impairment of islets. More systematic studies of gene-diet interactions in β-cells of rodent models in combination with genetic profiling might reveal the regulatory circuits fundamental for the understanding of diet-induced impairments of β-cell function in humans.
Collapse
Affiliation(s)
- A Chadt
- German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
45
|
Petropavlovskaia M, Daoud J, Zhu J, Moosavi M, Ding J, Makhlin J, Assouline-Thomas B, Rosenberg L. Mechanisms of action of islet neogenesis-associated protein: comparison of the full-length recombinant protein and a bioactive peptide. Am J Physiol Endocrinol Metab 2012; 303:E917-27. [PMID: 22850686 PMCID: PMC3469614 DOI: 10.1152/ajpendo.00670.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Islet neogenesis-associated protein (INGAP) was discovered in the partially duct-obstructed hamster pancreas as a factor inducing formation of new duct-associated islets. A bioactive portion of INGAP, INGAP(104-118) peptide (INGAP-P), has been shown to have neogenic and insulin-potentiating activity in numerous studies, including recent phase 2 clinical trials that demonstrated improved glucose homeostasis in both type 1 and type 2 diabetic patients. Aiming to improve INGAP-P efficacy and to understand its mechanism of action, we cloned the full-length protein (rINGAP) and compared the signaling events induced by the protein and the peptide in RIN-m5F cells that respond to INGAP with an increase in proliferation. Here, we show that, although both rINGAP and INGAP-P signal via the Ras/Raf/ERK pathway, rINGAP is at least 100 times more efficient on a molar basis than INGAP-P. For either ligand, ERK1/2 activation appears to be pertussis toxin sensitive, suggesting involvement of a G protein-coupled receptor(s). However, there are clear differences between the peptide and the protein in interactions with the cell surface and in the downstream signaling. We demonstrate that fluorescent-labeled rINGAP is characterized by clustering on the membrane and by slow internalization (≤5 h), whereas INGAP-P does not cluster and is internalized within minutes. Signaling by rINGAP appears to involve Src, in contrast to INGAP-P, which appears to activate Akt in addition to the Ras/Raf/ERK1/2 pathway. Thus our data suggest that interactions of INGAP with the cell surface are important to consider for further development of INGAP as a pharmacotherapy for diabetes.
Collapse
Affiliation(s)
- Maria Petropavlovskaia
- Department of Surgery, the Research Institute of the McGill University Health Center, McGill University, Montreal, Québec, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Gong L, Liu FQ, Wang Y, Hou XG, Zhang W, Qin WD, Zhang Y, Chen L, Zhang MX. Poly (ADP-ribose) transferase/polymerase-1-deficient mice resistant to age-dependent decrease in β-cell proliferation. Mol Med 2012; 18:816-24. [PMID: 22481269 DOI: 10.2119/molmed.2011.00458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 03/29/2012] [Indexed: 01/09/2023] Open
Abstract
Basal and adaptive β-cell regeneration capacity declines with old age, but the underlying molecular mechanisms remain incompletely understood. Poly (adenosine diphosphate [ADP]-ribose) polymerase 1 (PARP-1) is considered a multifunctional enzyme and transcription factor that regulates pancreatic β-cell death, regeneration and insulin secretion. We analyzed the capacity of β-cell regeneration in 2-month-old (young) and 12-month-old (old) wild-type (WT) and PARP-1⁻/⁻ mice before and after low-dose streptozotocin (STZ), a stimulus of β-cell regeneration and the underlying mechanism. Before STZ administration, young WT and PARP-1⁻/⁻ mice showed similar β-cell proliferation. By contrast, old WT but not old PARP-1⁻/⁻ mice showed severely restricted β-cell proliferation. In further assessment of the adaptive β-cell regeneration capacity with age, we observed that with a single low dose of STZ, young WT and PARP-1⁻/⁻ mice showed a similar increase in β-cell proliferation, with few changes in old WT mice. Surprisingly, adaptive β-cell proliferation capacity was significantly higher in old PARP-1⁻/⁻ mice than old WT mice after STZ administration. The ability of β-cell mass to expand was associated with increased levels of the regenerating (Reg) genes RegI and RegII but not RegIV. Therefore, PARP-1 is a key regulator in β-cell regeneration with advancing age in mice.
Collapse
Affiliation(s)
- Lei Gong
- Department of Endocrinology, Shandong University, Qilu Hospital, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Que YA, Delodder F, Guessous I, Graf R, Bain M, Calandra T, Liaudet L, Eggimann P. Pancreatic stone protein as an early biomarker predicting mortality in a prospective cohort of patients with sepsis requiring ICU management. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:R114. [PMID: 22748193 PMCID: PMC3580689 DOI: 10.1186/cc11406] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 07/02/2012] [Indexed: 12/16/2022]
Abstract
Introduction Biomarkers, such as C-reactive protein [CRP] and procalcitonin [PCT], are insufficiently sensitive or specific to stratify patients with sepsis. We investigate the prognostic value of pancreatic stone protein/regenerating protein (PSP/reg) concentration in patients with severe infections. Methods PSP/reg, CRP, PCT, tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL1-β), IL-6 and IL-8 were prospectively measured in cohort of patients ≥ 18 years of age with severe sepsis or septic shock within 24 hours of admission in a medico-surgical intensive care unit (ICU) of a community and referral university hospital, and the ability to predict in-hospital mortality was determined. Results We evaluated 107 patients, 33 with severe sepsis and 74 with septic shock, with in-hospital mortality rates of 6% (2/33) and 25% (17/74), respectively. Plasma concentrations of PSP/reg (343.5 vs. 73.5 ng/ml, P < 0.001), PCT (39.3 vs. 12.0 ng/ml, P < 0.001), IL-8 (682 vs. 184 ng/ml, P < 0.001) and IL-6 (1955 vs. 544 pg/ml, P < 0.01) were significantly higher in patients with septic shock than with severe sepsis. Of note, median PSP/reg was 13.0 ng/ml (IQR: 4.8) in 20 severely burned patients without infection. The area under the ROC curve for PSP/reg (0.65 [95% CI: 0.51 to 0.80]) was higher than for CRP (0.44 [0.29 to 0.60]), PCT 0.46 [0.29 to 0.61]), IL-8 (0.61 [0.43 to 0.77]) or IL-6 (0.59 [0.44 to 0.75]) in predicting in-hospital mortality. In patients with septic shock, PSP/reg was the only biomarker associated with in-hospital mortality (P = 0.049). Risk of mortality increased continuously for each ascending quartile of PSP/reg. Conclusions Measurement of PSP/reg concentration within 24 hours of ICU admission may predict in-hospital mortality in patients with septic shock, identifying patients who may benefit most from tailored ICU management.
Collapse
|
48
|
Kapur R, Højfeldt TW, Højfeldt TW, Rønn SG, Karlsen AE, Heller RS. Short-term effects of INGAP and Reg family peptides on the appearance of small β-cells clusters in non-diabetic mice. Islets 2012; 4:40-8. [PMID: 22395480 DOI: 10.4161/isl.18659] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Reg3 peptides INGAP-PP and human Reg3α/β (HIP) have been hypothesized to stimulate β-cell neogenesis in the pancreas. Administration of INGAP-PP has been shown to cause an increase in β-cell mass in multiple animal models, reverse streptozotocin (STZ) induced diabetes in mice and reduces HbA1c levels in type 2 diabetic humans. In this study, we have examined the ability of the INGAP-PP and HIP peptides to induce β-cell formation in vivo in normal mice through short-term administration of the peptides. We assessed the peptides ability to induce an increase in extra-islet insulin-positive cell clusters by looking at β-cell number by point counting morphometry on pancreata that had been randomized using the smooth fractionator principle in non-diabetic NMRI mice after short-term injections of the peptides (5 d). Five day continuous BrdU labeling was used to determine if the new β-cells were derived from replicating β-cells. Real time quantitative RT-PCR and immuno-histochemistry was used to analyze changes in pancreatic transcription factor expression. A 1.5- to 2-fold increase in the volume of small extra-islet insulin-positive clusters post 5 d treatment with INGAP-PP and HIP as compared with mice treated with a non-peptide control or scrambled peptide (p<0.05) (n = 7) was found. Five day continuous BrdU infusion during the 5 d period showed little or no incorporation in islets or small insulin clusters. Five days of treatment with INGAP-PP or HIP, showed a tendency toward increased levels of pancreatic progenitor markers such as Ngn3, Nkx6.1, Sox9 and Ins. These are the first studies to compare and indicate that the human Reg3 α/β (HIP) peptide has similar bioactivity in vivo as INGAP by causing formation of small β-cell clusters in extra-islet pancreatic tissue after only 5 d of treatment. Upregulation of pancreatic transcription factors may be part of the mechanism of action.
Collapse
Affiliation(s)
- Rahul Kapur
- Department of Beta Cell Regeneration; Hagedorn Research Institute; Gentofte, Denmark
| | | | | | - Sif Groth Rønn
- Department of Incretin Biology; Hagedorn Research Institute; Gentofte, Denmark
| | - Allan E Karlsen
- Department of Beta Cell Regeneration; Hagedorn Research Institute; Gentofte, Denmark
| | - R Scott Heller
- Department of Beta Cell Regeneration; Hagedorn Research Institute; Gentofte, Denmark
| |
Collapse
|
49
|
Parikh A, Stephan AF, Tzanakakis ES. Regenerating proteins and their expression, regulation and signaling. Biomol Concepts 2011; 3:57-70. [PMID: 22582090 DOI: 10.1515/bmc.2011.055] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The regenerating (Reg) protein family comprises C-type lectin-like proteins discovered independently during pancreatitis and pancreatic islet regeneration. However, an increasing number of studies provide evidence of participation of Reg proteins in the proliferation and differentiation of diverse cell types. Moreover, Reg family members are associated with various pathologies, including diabetes and forms of gastrointestinal cancer. These findings have led to the emergence of key roles for Reg proteins as anti-inflammatory, antiapoptotic and mitogenic agents in multiple physiologic and disease contexts. Yet, there are significant gaps in our knowledge regarding the regulation of expression of different Reg genes. In addition, the pathways relaying Reg-triggered signals, their targets and potential cross-talk with other cascades are still largely unknown. In this review, the expression patterns of different Reg members in the pancreas and extrapancreatic tissues are described. Moreover, factors known to modulate Reg levels in different cell types are discussed. Several signaling pathways, which have been implicated in conferring the effects of Reg ligands to date, are also delineated. Further efforts are necessary for elucidating the biological processes underlying the action of Reg proteins and their involvement in various maladies. Better understanding of the function of Reg genes and proteins will be beneficial in the design and development of therapies utilizing or targeting this protein group.
Collapse
Affiliation(s)
- Abhirath Parikh
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, NY 14260
| | | | | |
Collapse
|
50
|
Peterson KM, Guo X, Elkahloun AG, Mondal D, Bardhan PK, Sugawara A, Duggal P, Haque R, Petri WA. The expression of REG 1A and REG 1B is increased during acute amebic colitis. Parasitol Int 2011; 60:296-300. [PMID: 21586335 PMCID: PMC3124591 DOI: 10.1016/j.parint.2011.04.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 04/26/2011] [Accepted: 04/29/2011] [Indexed: 01/31/2023]
Abstract
Entamoeba histolytica, a protozoan parasite, is an important cause of diarrhea and colitis in the developing world. Amebic colitis is characterized by ulceration of the intestinal mucosa. We performed microarray analysis of intestinal biopsies during acute and convalescent amebiasis in order to identify genes potentially involved in tissue injury or repair. Colonic biopsy samples were obtained from 8 patients during acute E. histolytica colitis and again 60 days after recovery. Gene expression in the biopsies was evaluated using microarray, and confirmed by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR). REG 1A and REG 1B were the most up-regulated of all genes in the human intestine in acute versus convalescent E. histolytica disease: as determined by microarray, the levels of induction were 7.4-fold and 10.7 fold for REG 1A and B; p=0.003 and p=0.006 respectively. Increased expression of REG 1A and REG 1B protein in the colonic crypt epithelial cells during acute amebiasis was similarly observed by immunohistochemistry. Because REG 1 protein is anti-apoptotic and pro-proliferative, and since E. histolytica induces apoptosis of the intestinal epithelium as part of its disease process, we next tested if REG 1 might be protective during amebiasis by preventing parasite-induced apoptosis. Intestinal epithelial cells from REG 1-/- mice were found to be more susceptible to spontaneous, and parasite-induced, apoptosis in vitro (p=0.03). We concluded that REG 1A and REG 1B were upregulated during amebiasis and may function to protect the intestinal epithelium from parasite-induced apoptosis.
Collapse
Affiliation(s)
- Kristine M Peterson
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|