1
|
Zhu W, Pan L, Cui X, Russo AC, Ray R, Pederson B, Wei X, Lin LL, Hafner H, Gregg B, Shrestha N, Liu C, Naji A, Arvan P, Sandoval DA, Lindberg I, Qi L, Reinert RB. SEL1L-HRD1 ER-Associated Degradation Facilitates Prohormone Convertase 2 Maturation and Glucagon Production in Islet α Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644437. [PMID: 40166183 PMCID: PMC11957139 DOI: 10.1101/2025.03.20.644437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Proteolytic cleavage of proglucagon by prohormone convertase 2 (PC2) is required for islet α cells to generate glucagon. However, the regulatory mechanisms underlying this process remain largely unclear. Here, we report that SEL1L-HRD1 endoplasmic reticulum (ER)-associated degradation (ERAD), a highly conserved protein quality control system responsible for clearing misfolded proteins from the ER, plays a key role in glucagon production by regulating turnover of the nascent proform of the PC2 enzyme (proPC2). Using a mouse model with SEL1L deletion in proglucagon-expressing cells, we observed a progressive decline in stimulated glucagon secretion and a reduction in pancreatic glucagon content. Mechanistically, we found that endogenous proPC2 is a substrate of SEL1L-HRD1 ERAD, and that degradation of misfolded proPC2 ensures the maturation of activation-competent proPC2 protein. These findings identify ERAD as a novel regulator of PC2 biology and an essential mechanism for maintaining α cell function.
Collapse
Affiliation(s)
- Wenzhen Zhu
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Linxiu Pan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Xianwei Cui
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Anna Chiara Russo
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Rohit Ray
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Brent Pederson
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Xiaoqiong Wei
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Liangguang Leo Lin
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Hannah Hafner
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brigid Gregg
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48105, USA
| | - Neha Shrestha
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Chengyang Liu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Darleen A. Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Nutrition Section, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore, Baltimore, MD 21201, USA
| | - Ling Qi
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Rachel B. Reinert
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
2
|
Jurca AD, Galea-Holhos LB, Jurca AA, Atasie D, Petchesi CD, Severin E, Jurca CM. Wolfram Syndrome Type I Case Report and Review-Focus on Early Diagnosis and Genetic Variants. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1064. [PMID: 39064493 PMCID: PMC11278941 DOI: 10.3390/medicina60071064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: Wolfram syndrome type 1 (OMIM# 222300; ORPHAcode 3463) is an extremely rare autosomal recessive syndrome with a 25% recurrence risk in children. It is characterized by the presence of juvenile-onset diabetes mellitus (DM), progressive optic atrophy (OA), diabetes insipidus (DI), and sensorineural deafness (D), often referred to by the acronym DIDMOAD. It is a severe neurodegenerative disease with a life expectancy of 39 years, with death occurring due to cerebral atrophy. For a positive diagnosis, the presence of diabetes mellitus and optic nerve atrophy is sufficient. The disease occurs because of pathogenic variants in the WFS1 gene. The aim of this article is to present a case report of Wolfram Syndrome Type I, alongside a review of genetic variants, clinical manifestations, diagnosis, therapy, and long-term management. Emphasizing the importance of early diagnosis and a multidisciplinary approach, the study aims to enhance understanding and improve outcomes for patients with this complex syndrome. Materials and Methods: A case of a 28-year-old patient diagnosed with DM at the age of 6 and with progressive optic atrophy at 26 years old is presented. Molecular diagnosis revealed the presence of a heterozygous nonsense variant WFS1 c.1943G>A (p.Trp648*), and a heterozygous missense variant WFS1 c.1675G>C (p.Ala559Pro). Results: The molecular diagnosis of the patient confirmed the presence of a heterozygous nonsense variant and a heterozygous missense variant in the WFS1 gene, correlating with the clinical presentation of Wolfram syndrome type 1. Both allelic variants found in our patient have been previously described in other patients, whilst this combination has not been described before. Conclusions: This case report and review underscores the critical role of early recognition and diagnosis in Wolfram syndrome, facilitated by genetic testing. By identifying pathogenic variants in the WFS1 gene, genetic testing not only confirms diagnosis but also guides clinical management and informs genetic counseling for affected families. Timely intervention based on genetic insights can potentially reduce the progressive multisystem manifestations of the syndrome, thereby improving the quality of life and outcomes for patients.
Collapse
Affiliation(s)
- Alexandru Daniel Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
| | - Larisa Bianca Galea-Holhos
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania
| | | | - Diter Atasie
- Departament II Medical Clinic, Faculty of Medicine, University “Lucian Blaga of Sibiu”, Lucian Blaga Street 2A, 550169 Sibiu, Romania;
| | - Codruta Diana Petchesi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN-ITHACA), 410469 Oradea, Romania
| | - Emilia Severin
- Genetics Department, “Carol Davila” University of Medicine and Pharmacy, 020027 Bucharest, District 2, Romania
| | - Claudia Maria Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN-ITHACA), 410469 Oradea, Romania
| |
Collapse
|
3
|
Blanc M, Habbouche L, Xiao P, Lebeaupin C, Janona M, Vaillant N, Irondelle M, Gilleron J, Murcy F, Rousseau D, Luci C, Barouillet T, Marchetti S, Lacas-Gervais S, Yvan-Charvet L, Gual P, Cardozo AK, Bailly-Maitre B. Bax Inhibitor-1 preserves pancreatic β-cell proteostasis by limiting proinsulin misfolding and programmed cell death. Cell Death Dis 2024; 15:334. [PMID: 38744890 PMCID: PMC11094198 DOI: 10.1038/s41419-024-06701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
The prevalence of diabetes steadily increases worldwide mirroring the prevalence of obesity. Endoplasmic reticulum (ER) stress is activated in diabetes and contributes to β-cell dysfunction and apoptosis through the activation of a terminal unfolded protein response (UPR). Our results uncover a new role for Bax Inhibitor-One (BI-1), a negative regulator of inositol-requiring enzyme 1 (IRE1α) in preserving β-cell health against terminal UPR-induced apoptosis and pyroptosis in the context of supraphysiological loads of insulin production. BI-1-deficient mice experience a decline in endocrine pancreatic function in physiological and pathophysiological conditions, namely obesity induced by high-fat diet (HFD). We observed early-onset diabetes characterized by hyperglycemia, reduced serum insulin levels, β-cell loss, increased pancreatic lipases and pro-inflammatory cytokines, and the progression of metabolic dysfunction. Pancreatic section analysis revealed that BI-1 deletion overburdens unfolded proinsulin in the ER of β-cells, confirmed by ultrastructural signs of ER stress with overwhelmed IRE1α endoribonuclease (RNase) activity in freshly isolated islets. ER stress led to β-cell dysfunction and islet loss, due to an increase in immature proinsulin granules and defects in insulin crystallization with the presence of Rod-like granules. These results correlated with the induction of autophagy, ER phagy, and crinophagy quality control mechanisms, likely to alleviate the atypical accumulation of misfolded proinsulin in the ER. In fine, BI-1 in β-cells limited IRE1α RNase activity from triggering programmed β-cell death through apoptosis and pyroptosis (caspase-1, IL-1β) via NLRP3 inflammasome activation and metabolic dysfunction. Pharmaceutical IRE1α inhibition with STF-083010 reversed β-cell failure and normalized the metabolic phenotype. These results uncover a new protective role for BI-1 in pancreatic β-cell physiology as a stress integrator to modulate the UPR triggered by accumulating unfolded proinsulin in the ER, as well as autophagy and programmed cell death, with consequences on β-cell function and insulin secretion. In pancreatic β-cells, BI-1-/- deficiency perturbs proteostasis with proinsulin misfolding, ER stress, terminal UPR with overwhelmed IRE1α/XBP1s/CHOP activation, inflammation, β-cell programmed cell death, and diabetes.
Collapse
Affiliation(s)
- Marina Blanc
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Lama Habbouche
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Peng Xiao
- Inflammation and Cell Death Signalling group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles (ULB), Bruxelles, Belgique
| | - Cynthia Lebeaupin
- Degenerative Diseases Program, Sanford Burnham Prebys, La Jolla, CA, 92037, USA
| | - Marion Janona
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Nathalie Vaillant
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Marie Irondelle
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Jérôme Gilleron
- Université Côte d'Azur, Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Adipo-Cible Research Study Group, Centre Méditerranéen de Médecine Moléculaire (C3M), Team «Insulin Resistance in Obesity and type 2 Diabetes», Nice, France
| | - Florent Murcy
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Déborah Rousseau
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Team «Chronic Liver Diseases Associated with Obesity and Alcohol», Nice, France
| | - Carmelo Luci
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Team «Chronic Liver Diseases Associated with Obesity and Alcohol», Nice, France
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Sandrine Marchetti
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Team «Metabolism, cancer and immune responses», Nice, France
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée, CCMA, Nice, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France
| | - Philippe Gual
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Team «Chronic Liver Diseases Associated with Obesity and Alcohol», Nice, France
| | - Alessandra K Cardozo
- Inflammation and Cell Death Signalling group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles (ULB), Bruxelles, Belgique
| | - Béatrice Bailly-Maitre
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Team "Hematometabolism and Metainflammation (HEMAMETABO), 06204, Nice, France.
| |
Collapse
|
4
|
Sayyed Kassem L, Rajpal A, Barreiro MV, Ismail‐Beigi F. Beta-cell function in type 2 diabetes (T2DM): Can it be preserved or enhanced? J Diabetes 2023; 15:817-837. [PMID: 37522521 PMCID: PMC10590683 DOI: 10.1111/1753-0407.13446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 08/01/2023] Open
Abstract
Type 2 diabetes (T2DM) is a complex metabolic disorder manifested by hyperglycemia, insulin resistance, and deteriorating beta-cell function. A way to prevent progression of the disease might be to enhance beta-cell function and insulin secretion. However, most previous studies examined beta-cell function while patients were using glycemia-lowering agents without an adequate period off medications (washout). In the present review we focus on studies with a washout period. We performed a literature search (2010 to June 2021) using beta-cell function and enhancement. The evidence shows that beta-cell function can be enhanced. Bariatric surgery and very low calorie diets show improvement in beta-cell function in many individuals. In addition, use of glucagon-like peptide-1 receptor agonists for prolonged periods (3 years or more) can also lead to improvement of beta-cell function. Further research is needed to understand the mechanisms leading to improved beta-cell function and identify agents that could enhance beta-cell function in patients with T2DM.
Collapse
Affiliation(s)
- Laure Sayyed Kassem
- Case Western Reserve UniversityClevelandOhioUSA
- Cleveland VA Medical CenterCase Western Reserve UniversityClevelandOhioUSA
| | - Aman Rajpal
- Case Western Reserve UniversityClevelandOhioUSA
- Cleveland VA Medical CenterCase Western Reserve UniversityClevelandOhioUSA
| | | | - Faramarz Ismail‐Beigi
- Case Western Reserve UniversityClevelandOhioUSA
- Cleveland VA Medical CenterCase Western Reserve UniversityClevelandOhioUSA
- University Hospitals of ClevelandClevelandOhioUSA
| |
Collapse
|
5
|
Li Z, Liu X, Zhang K, Zhao H, Luo P, Li D, Liu Z, Yuan H, Zhang B, Xie X, Shen C. Role and Mechanism of Endoplasmic Reticulum Stress in Mice Pancreatic Islet Dysfunction After Severe Burns. J Burn Care Res 2023; 44:1231-1240. [PMID: 36869805 DOI: 10.1093/jbcr/irad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Indexed: 03/05/2023]
Abstract
This study attempted to investigate the role and mechanism of endoplasmic reticulum (ER) stress in the islet dysfunction in mice after severe burns. C57BL/6 mice were randomly divided into the sham group, burn group, and burn+4-phenylbutyric acid (4-PBA) group. Mice were burned with full thickness of 30% total surface area (TBSA), and 4-PBA solution was intraperitoneally injected into mice in burn+4-PBA group. Glucose-stimulated insulin secretion (GSIS), Fasting blood glucose (FBG) and glucose tolerance were detected 24 hours post severe burns. The ER stress-related pathway markers immunoglobulin binding protein (BIP), X-box binding protein 1 (XBP1), phosphorylation-PKR-like ER kinase (p-PERK), phosphorylation-eukaryotic translation initiation factor 2α (p-eIF2α), CHOP, activating transcription factor 6 (ATF6), apoptosis-related protein Cleaved-Caspase 3, and islet cell apoptosis were measured. Mice were characterized with elevated FBG, decreased glucose tolerance and GSIS levels post severe burns. The expression of BIP, XBP1, p-PERK, p-eIF2α, CHOP, ATF6, Cleaved-Caspase 3, and islet cell apoptosis were increased significantly after severe burns. 4-PBA treatment contributed to decreased FBG, improved glucose tolerance, increased GSIS, inhibited islet ER stress, and reduced pancreatic islet cell apoptosis in mice post severe burns. ER stress occurs in islets of severely burned mice, which leads to increased apoptosis of islet cells, thus resulting in islet dysfunction.
Collapse
Affiliation(s)
- Zhisheng Li
- Jinzhou Medical University, Jinzhou, China
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xinzhu Liu
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Kun Zhang
- Jinzhou Medical University, Jinzhou, China
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongqing Zhao
- Jinzhou Medical University, Jinzhou, China
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Peng Luo
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Dawei Li
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhaoxing Liu
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huageng Yuan
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bohan Zhang
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaoye Xie
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chuan'an Shen
- Department of Burns and Plastic Surgery, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Di Giuseppe G, Ciccarelli G, Soldovieri L, Capece U, Cefalo CMA, Moffa S, Nista EC, Brunetti M, Cinti F, Gasbarrini A, Pontecorvi A, Giaccari A, Mezza T. First-phase insulin secretion: can its evaluation direct therapeutic approaches? Trends Endocrinol Metab 2023; 34:216-230. [PMID: 36858875 DOI: 10.1016/j.tem.2023.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023]
Abstract
Our work is aimed at unraveling the role of the first-phase insulin secretion in the natural history of type 2 diabetes mellitus (T2DM) and its interrelationship with insulin resistance and with β cell function and mass. Starting from pathophysiology, we investigate the impact of impaired secretion on glucose homeostasis and explore postmeal hyperglycemia as the main clinical feature, underlining its relevance in the management of the disease. We also review dietary and pharmacological approaches aimed at improving early secretory defects and restoring residual β cell function. Furthermore, we discuss possible approaches to detect early secretory defects in clinical practice. By providing a journey through human and animal data, we attempt a unification of the recent evidence in an effort to offer a new outlook on β cell secretion.
Collapse
Affiliation(s)
- Gianfranco Di Giuseppe
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Gea Ciccarelli
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura Soldovieri
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Umberto Capece
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara M A Cefalo
- Department of Clinical and Molecular Medicine, University of Rome - Sapienza, Rome, Italy
| | - Simona Moffa
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Enrico C Nista
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy; Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Michela Brunetti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesca Cinti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy; Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Giaccari
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Teresa Mezza
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy; Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
7
|
Serbis A, Rallis D, Giapros V, Galli-Tsinopoulou A, Siomou E. Wolfram Syndrome 1: A Pediatrician's and Pediatric Endocrinologist's Perspective. Int J Mol Sci 2023; 24:ijms24043690. [PMID: 36835101 PMCID: PMC9960967 DOI: 10.3390/ijms24043690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Wolfram syndrome 1 (WS1) is a rare autosomal recessive neurodegenerative disease caused by mutations in WFS1 and WFS2 genes that produce wolframin, a protein involved in endoplasmic reticulum calcium homeostasis and cellular apoptosis. Its main clinical features are diabetes insipidus (DI), early-onset non-autoimmune insulin-dependent diabetes mellitus (DM), gradual loss of vision due to optic atrophy (OA) and deafness (D), hence the acronym DIDMOAD. Several other features from different systems have been reported such as urinary tract, neurological, and psychiatric abnormalities. In addition, endocrine disorders that can appear during childhood and adolescence include primary gonadal atrophy and hypergonadotropic hypogonadism in males and menstrual cycle abnormalities in females. Further, anterior pituitary dysfunction with deficient GH and/or ACTH production have been described. Despite the lack of specific treatment for the disease and its poor life expectancy, early diagnosis and supportive care is important for timely identifying and adequately managing its progressive symptoms. The current narrative review focuses on the pathophysiology and the clinical features of the disease, with a special emphasis on its endocrine abnormalities that appear during childhood and adolescence. Further, therapeutic interventions that have been proven to be effective in the management of WS1 endocrine complications are discussed.
Collapse
Affiliation(s)
- Anastasios Serbis
- Department of Pediatrics, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
- Correspondence:
| | - Dimitrios Rallis
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Assimina Galli-Tsinopoulou
- Second Department of Pediatrics, School of Medicine, Faculty of Health Sciences, AHEPA University General Hospital, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Ekaterini Siomou
- Department of Pediatrics, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| |
Collapse
|
8
|
Brusco N, Sebastiani G, Di Giuseppe G, Licata G, Grieco GE, Fignani D, Nigi L, Formichi C, Aiello E, Auddino S, Quero G, Cefalo CMA, Cinti F, Mari A, Ferraro PM, Pontecorvi A, Alfieri S, Giaccari A, Dotta F, Mezza T. Intra-islet insulin synthesis defects are associated with endoplasmic reticulum stress and loss of beta cell identity in human diabetes. Diabetologia 2023; 66:354-366. [PMID: 36280617 PMCID: PMC9807540 DOI: 10.1007/s00125-022-05814-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 01/07/2023]
Abstract
AIMS/HYPOTHESIS Endoplasmic reticulum (ER) stress and beta cell dedifferentiation both play leading roles in impaired insulin secretion in overt type 2 diabetes. Whether and how these factors are related in the natural history of the disease remains, however, unclear. METHODS In this study, we analysed pancreas biopsies from a cohort of metabolically characterised living donors to identify defects in in situ insulin synthesis and intra-islet expression of ER stress and beta cell phenotype markers. RESULTS We provide evidence that in situ altered insulin processing is closely connected to in vivo worsening of beta cell function. Further, activation of ER stress genes reflects the alteration of insulin processing in situ. Using a combination of 17 different markers, we characterised individual pancreatic islets from normal glucose tolerant, impaired glucose tolerant and type 2 diabetic participants and reconstructed disease progression. CONCLUSIONS/INTERPRETATION Our study suggests that increased beta cell workload is accompanied by a progressive increase in ER stress with defects in insulin synthesis and loss of beta cell identity.
Collapse
Affiliation(s)
- Noemi Brusco
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Guido Sebastiani
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Gianfranco Di Giuseppe
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giada Licata
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Giuseppina E Grieco
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Daniela Fignani
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Laura Nigi
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Caterina Formichi
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Elena Aiello
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Stefano Auddino
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Giuseppe Quero
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
- Pancreatic surgery unit, Pancreatic Advanced Research Center (CRMPG), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Chiara M A Cefalo
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Francesca Cinti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Pietro M Ferraro
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Sergio Alfieri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
- Pancreatic surgery unit, Pancreatic Advanced Research Center (CRMPG), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Andrea Giaccari
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Francesco Dotta
- Diabetes and Metabolic Disease Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy.
| | - Teresa Mezza
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| |
Collapse
|
9
|
Effects of Initial Combinations of Gemigliptin Plus Metformin Compared with Glimepiride Plus Metformin on Gut Microbiota and Glucose Regulation in Obese Patients with Type 2 Diabetes: The INTESTINE Study. Nutrients 2023; 15:nu15010248. [PMID: 36615904 PMCID: PMC9824054 DOI: 10.3390/nu15010248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
The efficacy and safety of medications can be affected by alterations in gut microbiota in human beings. Among antidiabetic medications, incretin-based therapy such as dipeptidyl peptidase 4 inhibitors might affect gut microbiomes, which are related to glucose metabolism. This was a randomized, controlled, active-competitor study that aimed to compare the effects of combinations of gemigliptin−metformin vs. glimepiride−metformin as initial therapies on gut microbiota and glucose homeostasis in drug-naïve patients with type 2 diabetes. Seventy drug-naïve patients with type 2 diabetes (mean age, 52.2 years) with a glycated hemoglobin (HbA1c) level ≥7.5% were assigned to either gemigliptin−metformin or glimepiride−metformin combination therapies for 24 weeks. Changes in gut microbiota, biomarkers linked to glucose regulation, body composition, and amino acid blood levels were investigated. Although both treatments decreased the HbA1c levels significantly, the gemigliptin−metformin group achieved HbA1c ≤ 7.0% without hypoglycemia or weight gain more effectively than did the glimepiride−metformin group (59% vs. 24%; p < 0.05). At the phylum level, the Firmicutes/Bacteroidetes ratio tended to decrease after gemigliptin−metformin therapy (p = 0.065), with a notable depletion of taxa belonging to Firmicutes, including Lactobacillus, Ruminococcus torques, and Streptococcus (all p < 0.05). However, regardless of the treatment modality, a distinct difference in the overall gut microbiome composition was noted between patients who reached the HbA1c target goal and those who did not (p < 0.001). Treatment with gemigliptin−metformin resulted in a higher achievement of the glycemic target without hypoglycemia or weight gain, better than with glimepiride−metformin; these improvements might be related to beneficial changes in gut microbiota.
Collapse
|
10
|
Zeitler P, El Ghormli L, Arslanian S, Caprio S, Isganaitis E, Kelsey MK, Weinstock RS, White NH, Drews K. Deterioration of Glycemic Control in Youth-Onset Type 2 Diabetes: What Are the Early and Late Predictors? J Clin Endocrinol Metab 2022; 107:e3384-e3394. [PMID: 35486388 PMCID: PMC9653021 DOI: 10.1210/clinem/dgac254] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE We examined predictors of early and late loss of glycemic control in individuals with youth-onset type 2 diabetes, as well as predictors of short-term deterioration in youth from the Treatment Options for type 2 Diabetes in Adolescents and Youth (TODAY) study. METHODS Demographic, physical, and biochemical measures at baseline and 48 months, and change over time, were examined in 584 participants separated into those with loss of glycemic control (sustained HbA1c ≥ 8%) before 48 months or at 48 months or later, and those who remained in control until the end of the study (median 6.8 years). Univariate and multivariate models, and receiver operating characteristic curve analyses were performed. RESULTS Approximately 45% of youth remained in control at 48 months; of these, 30% subsequently lost glycemic control prior to the end of follow-up. Predictors of early loss of glycemic control included baseline HbA1c, C-peptide index, oral disposition index, proinsulin, and proinsulin to insulin ratio. Predictors of late loss included baseline measures of insulin secretion and change in HbA1c and insulin processing at 48 months. A baseline HbA1c cutoff of ≥ 6.2% was optimally predictive of loss of glycemic control at any time, while an absolute rise in HbA1c > 0.5% related to loss of glycemic control within 3 to 6 months. CONCLUSION This analysis demonstrates that youth with type 2 diabetes at risk for loss of glycemic control, including impending rapid deterioration, can be identified using available clinical measures, allowing for closer monitoring of at-risk youth, and facilitating the design of research on better therapeutic options.
Collapse
Affiliation(s)
- Philip Zeitler
- University of Colorado
Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laure El Ghormli
- The Biostatistics Center, George Washington University, Rockville, MD 20852,USA
| | - Silva Arslanian
- University of Pittsburgh, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15213,USA
| | | | | | - Megan K Kelsey
- University of Colorado
Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ruth S Weinstock
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Neil H White
- Washington University in St. Louis School of Medicine, St. Louis, MO 63110,USA
| | - Kimberly Drews
- The Biostatistics Center, George Washington University, Rockville, MD 20852,USA
| |
Collapse
|
11
|
Hinzman CP, Singh B, Bansal S, Li Y, Iliuk A, Girgis M, Herremans KM, Trevino JG, Singh VK, Banerjee PP, Cheema AK. A multi-omics approach identifies pancreatic cancer cell extracellular vesicles as mediators of the unfolded protein response in normal pancreatic epithelial cells. J Extracell Vesicles 2022; 11:e12232. [PMID: 35656858 PMCID: PMC9164146 DOI: 10.1002/jev2.12232] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/22/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023] Open
Abstract
Although cancer-derived extracellular vesicles (cEVs) are thought to play a pivotal role in promoting cancer progression events, their precise effect on neighbouring normal cells is unknown. In this study, we investigated the impact of pancreatic cancer ductal adenocarcinoma (PDAC) derived EVs on recipient non-tumourigenic pancreatic normal epithelial cells upon internalization. We demonstrate that cEVs are readily internalized and induce endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) in treated normal pancreatic epithelial cells within 24 h. We further show that PDAC cEVs increase cell proliferation, migration, and invasion and that these changes are regulated at least in part, by the UPR mediator DDIT3. Subsequently, these cells release several inflammatory cytokines. Leveraging a layered multi-omics approach, we analysed EV cargo from a panel of six PDAC and two normal pancreas cell lines, using multiple EV isolation methods. We found that cEVs were enriched for an array of biomolecules which can induce or regulate ER stress and the UPR, including palmitic acid, sphingomyelins, metabolic regulators of tRNA charging and proteins which regulate trafficking and degradation. We further show that palmitic acid, at doses relevant to those found in cEVs, is sufficient to induce ER stress in normal pancreas cells. These results suggest that cEV cargo packaging may be designed to disseminate proliferative and invasive characteristics upon internalization by distant recipient normal cells, hitherto unreported. This study is among the first to highlight a major role for PDAC cEVs to induce stress in treated normal pancreas cells that may modulate a systemic response leading to altered phenotypes. These findings highlight the importance of EVs in mediating disease aetiology and open potential areas of investigation toward understanding the role of cEV lipids in promoting cell transformation in the surrounding microenvironment.
Collapse
Affiliation(s)
- Charles P. Hinzman
- Department of BiochemistryMolecular and Cellular BiologyGeorgetown University Medical CentreWashingtonDCUSA
| | - Baldev Singh
- Department of OncologyLombardi Comprehensive Cancer CenterGeorgetown University Medical CentreWashingtonDCUSA
| | - Shivani Bansal
- Department of OncologyLombardi Comprehensive Cancer CenterGeorgetown University Medical CentreWashingtonDCUSA
| | - Yaoxiang Li
- Department of OncologyLombardi Comprehensive Cancer CenterGeorgetown University Medical CentreWashingtonDCUSA
| | - Anton Iliuk
- Tymora Analytical OperationsWest LafayetteINUSA
| | - Michael Girgis
- Department of OncologyLombardi Comprehensive Cancer CenterGeorgetown University Medical CentreWashingtonDCUSA
| | | | - Jose G. Trevino
- Division of Surgical OncologyVCU Massey Cancer CentreRichmondVAUSA
| | - Vijay K. Singh
- Department of Pharmacology and Molecular TherapeuticsSchool of MedicineUniformed Services University of the Health SciencesBethesdaMDUSA
- Armed Forces Radiobiology Research InstituteUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Partha P. Banerjee
- Department of BiochemistryMolecular and Cellular BiologyGeorgetown University Medical CentreWashingtonDCUSA
| | - Amrita K. Cheema
- Department of BiochemistryMolecular and Cellular BiologyGeorgetown University Medical CentreWashingtonDCUSA
- Department of OncologyLombardi Comprehensive Cancer CenterGeorgetown University Medical CentreWashingtonDCUSA
| |
Collapse
|
12
|
Rodríguez-Castelán J, Zepeda-Pérez D, Rojas-Juárez R, Aceves C, Castelán F, Cuevas-Romero E. Effects of hypothyroidism on the female pancreas involve the regulation of estrogen receptors. Steroids 2022; 181:108996. [PMID: 35245530 DOI: 10.1016/j.steroids.2022.108996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/22/2022] [Indexed: 11/25/2022]
Abstract
This study aimed to investigate the impact of short-time hypothyroidism on the expression of aromatase, estrogen receptors (ERα, β), and GPR30 in the pancreas of female rabbits. The formation of new islets and the expression of insulin, GLUT4, and lactate dehydrogenase (LDH) were also analyzed. This purpose is based on actions that thyroid hormones and estrogens have on β-cells differentiation, acinar cell function, and insulin secretion. Twelve Chinchilla-breed adult virgin female rabbits were divided into control (n = 6) and hypothyroid (n = 6; methimazole 10 mg/kg for 30 days) groups. In the complete pancreas, expressions of aromatase and estrogen receptors, as well as proinsulin, GLUT4, and LDH were determined by western blot. Characteristics of islets were measured in slices of the pancreas with immunohistochemistry for insulin. Islet and acinar cells express aromatase, ERα, ERβ, and GPR30. Hypothyroidism increased the expression of ERα and diminished that for aromatase, ERβ, and GPR30 in the pancreas. It also promoted a high number of extra small islets (new islets) and increased the expression of proinsulin and GLUT4 in the pancreas. Our results show that actions of thyroid hormones and estrogens on β-cells neogenesis, acinar cell function, and synthesis and secretion of insulin are linked. Thus, the effects of hypothyroidism on the pancreas could include summatory actions of thyroid hormones plus estrogens. Our findings indicate the importance of monitoring estrogen levels and actions on the pancreas of hypothyroid women, particularly when serum estrogen concentrations are affected such as menopausal, pregnant, and those with contraceptive use.
Collapse
Affiliation(s)
- Julia Rodríguez-Castelán
- Autonomous University of Tlaxcala, Tlaxcala, Tlaxcala, Mexico; Department of Cellular and Molecular Neurobiology, Institute of Neurobiology, Autonomous Nacional University of Mexico, Juriquilla, Querétaro, Mexico
| | | | | | - Carmen Aceves
- Department of Cellular and Molecular Neurobiology, Institute of Neurobiology, Autonomous Nacional University of Mexico, Juriquilla, Querétaro, Mexico
| | - Francisco Castelán
- Department of Cellular and Physiology, Institute of Biomedical Research, Autonomous Nacional University of Mexico, Mexico City, Mexico; Center Tlaxcala of Behavior Biology, Autonomous University of Tlaxcala, Tlaxcala, Tlaxcala, Mexico
| | - Estela Cuevas-Romero
- Center Tlaxcala of Behavior Biology, Autonomous University of Tlaxcala, Tlaxcala, Tlaxcala, Mexico.
| |
Collapse
|
13
|
Scully KJ, Wolfsdorf JI. Efficacy of GLP-1 Agonist Therapy in Autosomal Dominant WFS1-Related Disorder: A Case Report. Horm Res Paediatr 2021; 93:409-414. [PMID: 33075784 DOI: 10.1159/000510852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Wolfram syndrome is a rare neurodegenerative disorder, characterized by the presence of diabetes insipidus, diabetes mellitus, optic atrophy, and sensorineural deafness. The majority of cases are due to autosomal recessive biallelic variants in the WFS1 gene; however, pathogenic autosomal dominant (AD) mutations have also been described. Glucagon-like peptide (GLP-1) agonists have been studied in both animal models and humans with classic Wolfram syndrome. CASE We present a 15-year-old female with a personal and family history of congenital strabismus, bilateral cataracts, low-frequency sensorineural hearing loss, and diabetes mellitus. Trio whole exome sequencing revealed a previously unknown maternally inherited heterozygous variant in exon 8 of the WFS1 gene c.2605_2616del12 p.Ser869_His872del, leading to the diagnosis of AD WFS1-related disorder. Treatment with a GLP-1 agonist resulted in marked improvement in glycemic control and discontinuation of insulin therapy. This patient's response to a GLP-1 agonist provides suggestive indirect evidence for a role of WFS1 on β-cell endoplasmic reticulum stress and suggests that treatment with a GLP-1 agonist should be considered in patients with dominant forms of WS.
Collapse
Affiliation(s)
- Kevin J Scully
- Department of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA,
| | - Joseph I Wolfsdorf
- Department of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Herlea-Pana O, Eeda V, Undi RB, Lim HY, Wang W. Pharmacological Inhibition of Inositol-Requiring Enzyme 1α RNase Activity Protects Pancreatic Beta Cell and Improves Diabetic Condition in Insulin Mutation-Induced Diabetes. Front Endocrinol (Lausanne) 2021; 12:749879. [PMID: 34675883 PMCID: PMC8524045 DOI: 10.3389/fendo.2021.749879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
β-cell ER stress plays an important role in β-cell dysfunction and death during the pathogenesis of diabetes. Proinsulin misfolding is regarded as one of the primary initiating factors of ER stress and unfolded protein response (UPR) activation in β-cells. Here, we found that the ER stress sensor inositol-requiring enzyme 1α (IRE1α) was activated in the Akita mice, a mouse model of mutant insulin gene-induced diabetes of youth (MIDY), a monogenic diabetes. Normalization of IRE1α RNase hyperactivity by pharmacological inhibitors significantly ameliorated the hyperglycemic conditions and increased serum insulin levels in Akita mice. These benefits were accompanied by a concomitant protection of functional β-cell mass, as shown by the suppression of β-cell apoptosis, increase in mature insulin production and reduction of proinsulin level. At the molecular level, we observed that the expression of genes associated with β-cell identity and function was significantly up-regulated and ER stress and its associated inflammation and oxidative stress were suppressed in islets from Akita mice treated with IRE1α RNase inhibitors. This study provides the evidence of the in vivo efficacy of IRE1α RNase inhibitors in Akita mice, pointing to the possibility of targeting IRE1α RNase as a therapeutic direction for the treatment of diabetes.
Collapse
Affiliation(s)
- Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Hui-Ying Lim
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| |
Collapse
|
15
|
Egan AM, Laurenti MC, Hurtado Andrade MD, Dalla Man C, Cobelli C, Bailey KR, Vella A. Limitations of the fasting proinsulin to insulin ratio as a measure of β-cell health in people with and without impaired glucose tolerance. Eur J Clin Invest 2021; 51:e13469. [PMID: 33289929 PMCID: PMC8169515 DOI: 10.1111/eci.13469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The fasting proinsulin to insulin ratio is elevated in people with type 2 diabetes and has been suggested as a marker of β-cell health. However, its utility in discriminating between individuals with varying degrees of β-cell dysfunction is unclear. Proinsulin has a very different half-life to insulin and unlike insulin does not undergo hepatic extraction prior to reaching the systemic circulation. Given these limitations, we sought to examine the relationship between fasting and postprandial concentrations of β-cell polypeptides (proinsulin, insulin and C-peptide) in people with normal and impaired glucose tolerance in differing metabolic environments. DESIGN Subjects were studied on two occasions in random order while undergoing an oral challenge. During one study day, free fatty acids were elevated (to induce insulin resistance) by infusion of Intralipid with heparin. Proinsulin to insulin and proinsulin to C-peptide ratios were calculated for the 0-, 30-, 60- and 240-minute time points. Insulin action (Si) and β-cell responsivity (Φ) indices were calculated using the oral minimal model. RESULTS The fasting proinsulin to c-peptide or fasting proinsulin to insulin ratios did not differ between groups and did not predict subsequent β-cell responsivity to glucose during the glycerol or Intralipid study days in either group. CONCLUSIONS Among nondiabetic individuals, the fasting proinsulin to insulin ratio is not a useful marker of β-cell function.
Collapse
Affiliation(s)
- Aoife M Egan
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Marcello C Laurenti
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | | | - Chiara Dalla Man
- Department of Information Engineering, Università degli Studi di Padova, Padova, Italy
| | - Claudio Cobelli
- Department of Information Engineering, Università degli Studi di Padova, Padova, Italy
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Victor P, Sarada D, Ramkumar KM. Crosstalk between endoplasmic reticulum stress and oxidative stress: Focus on protein disulfide isomerase and endoplasmic reticulum oxidase 1. Eur J Pharmacol 2020; 892:173749. [PMID: 33245896 DOI: 10.1016/j.ejphar.2020.173749] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
Cellular stress and inflammation, establishing as disease pathology, have reached great heights in the last few decades. Stress conditions such as hyperglycemia, hyperlipidemia and lipoproteins are known to disturb proteostasis resulting in the accumulation of unfolded or misfolded proteins, alteration in calcium homeostasis culminating in unfolded protein response. Protein disulfide isomerase and endoplasmic reticulum oxidase-1 are the key players in protein folding. The protein folding process assisted by endoplasmic reticulum oxidase-1 results in the production of reactive oxygen species in the lumen of the endoplasmic reticulum. Production of reactive oxygen species beyond the quenching capacity of the antioxidant systems perturbs ER homeostasis. Endoplasmic reticulum stress also induces the production of cytokines leading to inflammatory responses. This has been proven to be the major causative factor for various pathophysiological states compared to other cellular triggers in diseases, which further manifests to increased oxidative stress, mitochondrial dysfunction, and altered inflammatory responses, deleterious to cellular physiology and homeostasis. Numerous studies have drawn correlations between the progression of several diseases in association with endoplasmic reticulum stress, redox protein folding, oxidative stress and inflammatory responses. This review aims to provide an insight into the role of protein disulfide isomerase and endoplasmic reticulum oxidase-1 in endoplasmic reticulum stress, unfolded protein response, mitochondrial dysfunction, and inflammatory responses, which exacerbate the progression of various diseases.
Collapse
Affiliation(s)
- Paul Victor
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India
| | - Dronamraju Sarada
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India; Life Science Division, SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India.
| |
Collapse
|
17
|
Bender C, Rodriguez-Calvo T, Amirian N, Coppieters KT, von Herrath MG. The healthy exocrine pancreas contains preproinsulin-specific CD8 T cells that attack islets in type 1 diabetes. SCIENCE ADVANCES 2020; 6:6/42/eabc5586. [PMID: 33067232 PMCID: PMC7567597 DOI: 10.1126/sciadv.abc5586] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/03/2020] [Indexed: 05/03/2023]
Abstract
Preproinsulin (PPI) is presumably a crucial islet autoantigen found in patients with type 1 diabetes (T1D) but is also recognized by CD8+ T cells from healthy individuals. We quantified PPI-specific CD8+ T cells within different areas of the human pancreas from nondiabetic controls, autoantibody-positive donors, and donors with T1D to investigate their role in diabetes development. This spatial cellular quantitation revealed unusually high frequencies of autoreactive CD8+ T cells supporting the hypothesis that PPI is indeed a key autoantigen. To our surprise, PPI-specific CD8+ T cells were already abundantly present in the nondiabetic pancreas, thus questioning the dogma that T1D is caused by defective thymic deletion or systemic immune dysregulation. During T1D development, these cells accumulated in and around islets, indicating that an islet-specific trigger such as up-regulation of major histocompatibility complex class I might be essential to unmask beta cells to the immune system.
Collapse
Affiliation(s)
- Christine Bender
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Teresa Rodriguez-Calvo
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- The Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Institute of Diabetes Research, Munich-Neuherberg, Germany
| | - Natalie Amirian
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ken T Coppieters
- Global Research Project Management, Novo Nordisk, Måløv, Denmark
| | - Matthias G von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA.
- The Novo Nordisk Research Center Seattle Inc., Seattle, WA, USA
| |
Collapse
|
18
|
Sun J, Xiong Y, Li X, Haataja L, Chen W, Mir SA, Lv L, Madley R, Larkin D, Anjum A, Dhayalan B, Rege N, Wickramasinghe NP, Weiss MA, Itkin-Ansari P, Kaufman RJ, Ostrov DA, Arvan P, Liu M. Role of Proinsulin Self-Association in Mutant INS Gene-Induced Diabetes of Youth. Diabetes 2020; 69:954-964. [PMID: 32139596 PMCID: PMC7171958 DOI: 10.2337/db19-1106] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/22/2020] [Indexed: 02/06/2023]
Abstract
Abnormal interactions between misfolded mutant and wild-type (WT) proinsulin (PI) in the endoplasmic reticulum (ER) drive the molecular pathogenesis of mutant INS gene-induced diabetes of youth (MIDY). How these abnormal interactions are initiated remains unknown. Normally, PI-WT dimerizes in the ER. Here, we suggest that the normal PI-PI contact surface, involving the B-chain, contributes to dominant-negative effects of misfolded MIDY mutants. Specifically, we find that PI B-chain tyrosine-16 (Tyr-B16), which is a key residue in normal PI dimerization, helps confer dominant-negative behavior of MIDY mutant PI-C(A7)Y. Substitutions of Tyr-B16 with either Ala, Asp, or Pro in PI-C(A7)Y decrease the abnormal interactions between the MIDY mutant and PI-WT, rescuing PI-WT export, limiting ER stress, and increasing insulin production in β-cells and human islets. This study reveals the first evidence indicating that noncovalent PI-PI contact initiates dominant-negative behavior of misfolded PI, pointing to a novel therapeutic target to enhance PI-WT export and increase insulin production.
Collapse
Affiliation(s)
- Jinhong Sun
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Yi Xiong
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Leena Haataja
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Wei Chen
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Saiful A Mir
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Li Lv
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Rachel Madley
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Dennis Larkin
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Arfah Anjum
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Nischay Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
| | | | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Pamela Itkin-Ansari
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - David A Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Peter Arvan
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Ming Liu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Zhang IX, Raghavan M, Satin LS. The Endoplasmic Reticulum and Calcium Homeostasis in Pancreatic Beta Cells. Endocrinology 2020; 161:bqz028. [PMID: 31796960 PMCID: PMC7028010 DOI: 10.1210/endocr/bqz028] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 12/01/2019] [Indexed: 12/14/2022]
Abstract
The endoplasmic reticulum (ER) mediates the first steps of protein assembly within the secretory pathway and is the site where protein folding and quality control are initiated. The storage and release of Ca2+ are critical physiological functions of the ER. Disrupted ER homeostasis activates the unfolded protein response (UPR), a pathway which attempts to restore cellular equilibrium in the face of ER stress. Unremitting ER stress, and insufficient compensation for it results in beta-cell apoptosis, a process that has been linked to both type 1 diabetes (T1D) and type 2 diabetes (T2D). Both types are characterized by progressive beta-cell failure and a loss of beta-cell mass, although the underlying causes are different. The reduction of mass occurs secondary to apoptosis in the case of T2D, while beta cells undergo autoimmune destruction in T1D. In this review, we examine recent findings that link the UPR pathway and ER Ca2+ to beta cell dysfunction. We also discuss how UPR activation in beta cells favors cell survival versus apoptosis and death, and how ER protein chaperones are involved in regulating ER Ca2+ levels. Abbreviations: BiP, Binding immunoglobulin Protein ER; endoplasmic reticulum; ERAD, ER-associated protein degradation; IFN, interferon; IL, interleukin; JNK, c-Jun N-terminal kinase; KHE, proton-K+ exchanger; MODY, maturity-onset diabetes of young; PERK, PRKR-like ER kinase; SERCA, Sarco/Endoplasmic Reticulum Ca2+-ATPases; T1D, type 1 diabetes; T2D, type 2 diabetes; TNF, tumor necrosis factor; UPR, unfolded protein response; WRS, Wolcott-Rallison syndrome.
Collapse
Affiliation(s)
- Irina X Zhang
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan, Ann Arbor, MI
| | - Malini Raghavan
- Department of Microbiology and Immunology Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Leslie S Satin
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
20
|
Pallotta MT, Tascini G, Crispoldi R, Orabona C, Mondanelli G, Grohmann U, Esposito S. Wolfram syndrome, a rare neurodegenerative disease: from pathogenesis to future treatment perspectives. J Transl Med 2019; 17:238. [PMID: 31337416 PMCID: PMC6651977 DOI: 10.1186/s12967-019-1993-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Background Wolfram syndrome (WS), a rare genetic disorder, is considered the best prototype of endoplasmic reticulum (ER) diseases. Classical WS features are childhood-onset diabetes mellitus, optic atrophy, deafness, diabetes insipidus, neurological signs, and other abnormalities. Two causative genes (WFS1 and WFS2) have been identified. The transmission of the disease takes place in an autosomal recessive mode but autosomal dominant mutations responsible for WS-related disorders have been described. Prognosis is poor, death occurs at the median age of 39 years with a major cause represented by respiratory failure as a consequence of brain stem atrophy and neurodegeneration. The aim of this narrative review is to focus on etiology, pathogenesis and natural history of WS for an adequate patient management and for the discussion of future therapeutic interventions. Main body WS requires a multidisciplinary approach in order to be successfully treated. A prompt diagnosis decreases morbidity and mortality through prevention and treatment of complications. Being a monogenic pathology, WS represents a perfect model to study the mechanisms of ER stress and how this condition leads to cell death, in comparison with other prevalent diseases in which multiple factors interact to produce the disease manifestations. WS is also an important disease prototype to identify drugs and molecules associated with ER homeostasis. Evidence indicates that specific metabolic diseases (type 1 and type 2 diabetes), neurodegenerative diseases, atherosclerosis, inflammatory pathologies and also cancer are closely related to ER dysfunction. Conclusions Therapeutic strategies in WS are based on drug repurposing (i.e., investigation of approved drugs for novel therapeutic indications) with the aim to stop the progression of the disease by reducing the endoplasmic reticulum stress. An extensive understanding of WS from pathophysiology to therapy is fundamental and more studies are necessary to better manage this devastating disease and guarantee the patients a better quality of life and longer life expectancy.
Collapse
Affiliation(s)
- Maria Teresa Pallotta
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Giorgia Tascini
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Roberta Crispoldi
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Ciriana Orabona
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Giada Mondanelli
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Ursula Grohmann
- Pharmacology Section, Department of Experimental Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy.
| |
Collapse
|
21
|
Hao Y, Shen S, Yin F, Zhang Y, Liu J. Unfolded protein response is involved in geniposide‐regulating glucose‐stimulated insulin secretion in INS‐1 cells. Cell Biochem Funct 2019; 37:368-376. [DOI: 10.1002/cbf.3414] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/06/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Yanan Hao
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Shenli Shen
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Fei Yin
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Yonglan Zhang
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Jianhui Liu
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| |
Collapse
|
22
|
Arunagiri A, Haataja L, Pottekat A, Pamenan F, Kim S, Zeltser LM, Paton AW, Paton JC, Tsai B, Itkin-Ansari P, Kaufman RJ, Liu M, Arvan P. Proinsulin misfolding is an early event in the progression to type 2 diabetes. eLife 2019; 8:44532. [PMID: 31184302 PMCID: PMC6559786 DOI: 10.7554/elife.44532] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Biosynthesis of insulin – critical to metabolic homeostasis – begins with folding of the proinsulin precursor, including formation of three evolutionarily conserved intramolecular disulfide bonds. Remarkably, normal pancreatic islets contain a subset of proinsulin molecules bearing at least one free cysteine thiol. In human (or rodent) islets with a perturbed endoplasmic reticulum folding environment, non-native proinsulin enters intermolecular disulfide-linked complexes. In genetically obese mice with otherwise wild-type islets, disulfide-linked complexes of proinsulin are more abundant, and leptin receptor-deficient mice, the further increase of such complexes tracks with the onset of islet insulin deficiency and diabetes. Proinsulin-Cys(B19) and Cys(A20) are necessary and sufficient for the formation of proinsulin disulfide-linked complexes; indeed, proinsulin Cys(B19)-Cys(B19) covalent homodimers resist reductive dissociation, highlighting a structural basis for aberrant proinsulin complex formation. We conclude that increased proinsulin misfolding via disulfide-linked complexes is an early event associated with prediabetes that worsens with ß-cell dysfunction in type two diabetes. Our body fine-tunes the amount of sugar in our blood thanks to specialized ‘beta cells’ in the pancreas, which can release a hormone called insulin. To produce insulin, the beta cells first need to build an early version of the molecule – known as proinsulin – inside a cellular compartment called the endoplasmic reticulum. This process involves the formation of internal staples that keep the molecule of proinsulin folded correctly. Individuals developing type 2 diabetes have spikes of sugar in their blood, and so their bodies often respond by trying to make large amounts of insulin. After a while, the beta cells can fail to keep up, which brings on the full-blown disease. However, scientists have discovered that early in type 2 diabetes, the endoplasmic reticulum of beta cells can already show signs of stress; yet, the exact causes of this early damage are still unknown. To investigate this, Arunagiri et al. looked into whether proinsulin folds correctly during the earliest stages of type 2 diabetes. Biochemical experiments showed that even healthy beta cells contained some misfolded proinsulin molecules, where the molecular staples that should fold proinsulin internally were instead abnormally linking proinsulin molecules together. Further work revealed that the misfolded proinsulin was accumulating inside the endoplasmic reticulum. Finally, obese mice that were in the earliest stages of type 2 diabetes had the highest levels of abnormal proinsulin in their beta cells. Overall, the work by Arunagiri et al. suggests that large amounts of proinsulin molecules stapling themselves to each other in the endoplasmic reticulum of beta cells could be an early hallmark of the disease, and could make it get worse. A separate study by Jang et al. also shows that a protein that limits the misfolding of proinsulin is key to maintain successful insulin production in animals eating a Western-style, high fat diet. Hundreds of millions of people around the world have type 2 diabetes, and this number is rising quickly. Detecting and then fixing early problems associated with the condition may help to stop the disease in its track.
Collapse
Affiliation(s)
- Anoop Arunagiri
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Anita Pottekat
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Fawnnie Pamenan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Soohyun Kim
- Department of Biomedical Science and Technology, Konkuk University, Gwangjin-gu, Republic of Korea
| | - Lori M Zeltser
- Department of Pathology and Cell Biology, Naomi Berrie Diabetes Center, Columbia University, New York, United States
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Pamela Itkin-Ansari
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Ming Liu
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States.,Department of Endocrinology and Metabolism, Tianjin Medical University, Tianjin, China
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|