1
|
Tijani OK, Moreno-Lopez M, Louvet I, Acosta-Montalvo A, Coddeville A, Gmyr V, Kerr-Conte J, Pattou F, Vantyghem MC, Saponaro C, Bonner C, Espiard S. Impact of therapeutic doses of prednisolone and other glucocorticoids on insulin secretion from human islets. ANNALES D'ENDOCRINOLOGIE 2025; 86:101676. [PMID: 39643079 DOI: 10.1016/j.ando.2024.101676] [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: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Glucocorticoid-induced diabetes (GCID) is a prevalent health issue, generally attributed to insulin resistance. High doses of dexamethasone (DEX) are known to inhibit glucose-stimulated insulin secretion (GSIS), but the effects of lower doses, commonly used in chronic therapy, and equipotent doses of other glucocorticoids (GCs) such as hydrocortisone (HC) and prednisone (PRED) remain underexplored. This study aimed to investigate these effects in vitro, and explore variations between patients. MATERIALS AND METHODS Dynamic perifusion assays were conducted on human islets to evaluate the impact of different GCs on GSIS. The islets were treated for 24h with 250nM PRED and other GCs at equipotent anti-inflammatory doses (HC: 1μM; DEX: 38nM). RESULTS In 11 human islet donor preparations, 250nM PRED, corresponding to a clinical oral dose of 5mg/day, significantly inhibited the first and second phase of GSIS: area under the curve (AUC) decreased by 32.3% (P<0.001), first phase by 41.5% (P<0.001), and second phase by 38.4% (P<0.001). Despite interindividual differences in GSIS response to PRED, no significant differences were observed according to body mass index, gender or age. Comparing the effects of GCs at equipotent anti-inflammatory doses, DEX had a more pronounced inhibitory effect on GSIS than HC or PRED. CONCLUSIONS In vitro, low-dose PRED treatment significantly impacted GSIS. DEX had a more unfavorable impact on GSIS than HC or PRED, indicating that metabolic effects do not align with anti-inflammatory potency.
Collapse
Affiliation(s)
- Omolara Khadijat Tijani
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Maria Moreno-Lopez
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Isaline Louvet
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Ana Acosta-Montalvo
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Anaïs Coddeville
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Valery Gmyr
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Julie Kerr-Conte
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - François Pattou
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Marie-Christine Vantyghem
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Chiara Saponaro
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France
| | - Caroline Bonner
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France.
| | - Stéphanie Espiard
- University of Lille, CHU de Lille, Inserm U1190, EGID, Institut Pasteur de Lille, 59000 Lille, France.
| |
Collapse
|
2
|
Mbaye EHA, Scott EA, Burke JA. From Edmonton to Lantidra and beyond: immunoengineering islet transplantation to cure type 1 diabetes. FRONTIERS IN TRANSPLANTATION 2025; 4:1514956. [PMID: 40182604 PMCID: PMC11965681 DOI: 10.3389/frtra.2025.1514956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing β cells within pancreatic islets, the specialized endocrine cell clusters of the pancreas. Islet transplantation has emerged as a β cell replacement therapy, involving the infusion of cadaveric islets into a patient's liver through the portal vein. This procedure offers individuals with T1D the potential to restore glucose control, reducing or even eliminating the need for exogenous insulin therapy. However, it does not address the underlying autoimmune condition responsible for T1D. The need for systemic immunosuppression remains the primary barrier to making islet transplantation a more widespread therapy for patients with T1D. Here, we review recent progress in addressing the key limitations of islet transplantation as a viable treatment for T1D. Concerns over systemic immunosuppression arise from its potential to cause severe side effects, including opportunistic infections, malignancies, and toxicity to transplanted islets. Recognizing the risks, the Edmonton protocol (2000) marked a shift away from glucocorticoids to prevent β cell damage specifically. This transition led to the development of combination immunosuppressive therapies and the emergence of less toxic immunosuppressive and anti-inflammatory drugs. More recent advances in islet transplantation derive from islet encapsulation devices, biomaterial platforms releasing immunomodulatory compounds or surface-modified with immune regulating ligands, islet engineering and co-transplantation with accessory cells. While most of the highlighted studies in this review remain at the preclinical stage using mouse and non-human primate models, they hold significant potential for clinical translation if a transdisciplinary research approach is prioritized.
Collapse
Affiliation(s)
- El Hadji Arona Mbaye
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
3
|
Dimas-Ramírez CA, Fortanell-Meza LA, San Agustín-Morales D, Brenner-Muslera E, Mejía-Vilet JM, Almeda-Valdes P, Vázquez-Cárdenas P, Merayo-Chalico J, Barrera-Vargas A. Steroid-induced diabetes in lupus nephritis patients: Classic risk factors or a different type of diabetes? Lupus 2025; 34:234-242. [PMID: 39844380 DOI: 10.1177/09612033251315976] [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] [Indexed: 01/24/2025]
Abstract
BackgroundGlucocorticoids are frequently employed in systemic lupus erythematosus (SLE) patients and play a critical role in the induction therapy of lupus nephritis (LN), despite their many side effects, including steroid-induced diabetes (SID). Information regarding SID in SLE patients is quite scant.PurposeThe aim of this study was to determine risk factors associated with the development of SID in patients with LN.Research Design A nested case-control study was conducted.Study sampleWe included patients with biopsy-proven LN, who received induction treatment with steroids.Data Collection and/or AnalysisOut of the total of 358 patients, 35 (9.7%) developed SID.ResultsPatients with SID had more metabolic risk factors, including the metabolic score for insulin resistance (METS-IR); more factors related with lupus activity, with higher SLEDAI and SLICC-DI scores; and lower cumulative pre-induction steroid dose. A higher percentage of patients who developed SID received steroid pulses and a lower percentage received antimalarials. After logistic regression, the variables significantly associated with the development of SID were the SLEDAI index (OR 1.25 [95% CI 1.04-1.50], p 0.01), SLICC-DI (OR 4.93 [95% CI 2.14-11.3], p < 0.001), METS-IR (OR 1.17 [95% CI 1.04-1.32], p 0.009), delta METS-IR at 6 months (OR 1.20 [95% CI 1.03-1.39], p 0.01), and the use of antimalarials (OR 0.14, [95% CI 0.02-0.85], p 0.03). After propensity score matching, METS-IR remained a significant predictor of SID. Patients with METS-IR >36.8 were at higher risk (OR: 2.83, 95% CI: 1.09-7.36, p = 0.034).ConclusionsIn conclusion, SDI development in patients receiving induction therapy for LN is associated with both classic metabolic risk factors and SLE-specific factors, and antimalarial use could be associated with a protective effect. Rheumatologists should be aware of this potential complication, in order to implement appropriate management strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Paloma Almeda-Valdes
- Department of Endocrinology and Metabolism, Metabolic Reseach Unit, INCMNSZ, Ciudad de Mexico, Mexico
| | | | | | | |
Collapse
|
4
|
Semprasert N, Maneethorn P, Kooptiwut S. The protective effect of imatinib against pancreatic β-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress. Sci Rep 2024; 14:17691. [PMID: 39085384 PMCID: PMC11291718 DOI: 10.1038/s41598-024-68429-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Glucocorticoids (GCs) are known to stimulate pancreatic beta (β)-cell apoptosis via several mechanisms, including oxidative stress. Our previous study suggested an increase in dexamethasone-induced pancreatic β-cell apoptosis via a reduction of glutathione S-transferase P1 (GSTP1), which is an antioxidant enzyme. Imatinib, which is a tyrosine kinase inhibitor, also exerts antioxidant effect. This study aims to test our hypothesis that imatinib would prevent pancreatic β-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress. Our results revealed that dexamethasone significantly increased apoptosis in INS-1 cells when compared to the control, and that imatinib significantly decreased INS-1 cell apoptosis induced by dexamethasone. Moreover, dexamethasone significantly increased superoxide production in INS-1 cells when compared to the control; however, imatinib, when combined with dexamethasone, significantly reduced superoxide production in INS-1 cells. Dexamethasone significantly decreased GSTP1, p-ERK1/2, and BCL2 protein expression, but significantly increased p-JNK, p-p38, and BAX protein expression in INS-1 cells-all compared to control. Importantly, imatinib significantly ameliorated the effect of dexamethasone on the expression of GSTP1, p-ERK1/2, p-JNK, p-p38 MAPK, BAX, and BCL2. Furthermore-6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX), which is a GSTP1 inhibitor, neutralized the protective effect of imatinib against pancreatic β-cell apoptosis induced by dexamethasone. In conclusion, imatinib decreases pancreatic β-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress.
Collapse
Affiliation(s)
- Namoiy Semprasert
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Petcharee Maneethorn
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand.
| |
Collapse
|
5
|
Dotan I, Rudman Y, Turjeman A, Akirov A, Steinmetz T, Calvarysky B, Diker Cohen T. Glucagon-like Peptide 1 Receptor Agonists and Cardiovascular Outcomes in Solid Organ Transplant Recipients With Diabetes Mellitus. Transplantation 2024; 108:e121-e128. [PMID: 38361246 DOI: 10.1097/tp.0000000000004945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
BACKGROUND Glucagon-like peptide 1 receptor agonists (GLP1-RAs) reduce cardiovascular events and mortality in type 2 diabetes. Limited data are available on diabetes treatment after solid organ transplantation. We aimed to explore the effect of GLP1-RAs on cardiovascular outcomes in transplanted recipients with diabetes. METHODS We extracted data on adult transplant recipients (kidney, lungs, liver, heart) insured in a large health maintenance organization. Death-censored patients with diabetes treated with GLP1-RAs were matched with nonusers. The primary outcome was a composite of major cardiovascular events (MACEs): a nonfatal cardiac event (myocardial infarction, stable/unstable angina, coronary bypass, and coronary angiography), ischemic stroke and all-cause mortality. Secondary outcomes were MACE or peripheral vascular disease (MACE-PVD), and all-cause mortality. Safety outcomes included biliopancreatic adverse events. RESULTS We included 318 patients (69% males, average age 58.3 ± 11.0 y) with a 3.1-y median follow-up. The incidence of MACE was 101 of 1000 patient-years in GLP1-RAs users compared with 134 of 1000 in controls (hazard ratio [HR] 0.46; 95% confidence interval [CI], 0.27-0.78). GLP1-RAs similarly reduced the risk of MACE-PVD (HR 0.53; 95% CI, 0.33-0.88) and the risk of all-cause mortality (HR 0.39; 95% CI, 0.18-0.84). Biliopancreatic adverse events occurred less in GLP1-RA users. CONCLUSIONS Transplant recipients with diabetes who used GLP1-RAs had lower risks for MACE and all-cause mortality. These results may profoundly implicate the daily management of posttransplant recipients with diabetes, a population with a high prevalence of cardiometabolic risk factors and cardiovascular death. Transplant patients are usually excluded from randomized controlled trials and, hence might be undertreated with disease-modifying drugs. Larger prospective studies are needed in this unique population.
Collapse
Affiliation(s)
- Idit Dotan
- Institute of Endocrinology, Diabetes and Metabolism, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaron Rudman
- Institute of Endocrinology, Diabetes and Metabolism, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Turjeman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Research Authority, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Amit Akirov
- Institute of Endocrinology, Diabetes and Metabolism, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tali Steinmetz
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Nephrology, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Bronya Calvarysky
- Pharmacy, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Talia Diker Cohen
- Institute of Endocrinology, Diabetes and Metabolism, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
6
|
Ohmura H, Tobo T, Ando Y, Masuda T, Mimori K, Akashi K, Baba E. Case report: A rare case of triple negative breast cancer with development of acute pancreatitis due to dexamethasone during adjuvant chemotherapy. Front Oncol 2024; 14:1340419. [PMID: 38425339 PMCID: PMC10901989 DOI: 10.3389/fonc.2024.1340419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Here, we present the case of a 42-year-old female who developed acute pancreatitis due to dexamethasone during adjuvant chemotherapy for early triple negative breast cancer (TNBC). The patient received partial mastectomy and sentinel lymph node biopsy for early TNBC (cT1N0M0, cStage I) of the left breast. Dose-dense doxorubicin plus cyclophosphamide (ddAC) was administered as the adjuvant-chemotherapy; however, epigastralgia appeared on the fifth day of the first administration. A blood test showed a remarkable increase of serum pancreatic enzyme levels and computed tomography (CT) showed the swelling of pancreas and surrounding effusion, and she was diagnosed with moderate acute pancreatitis. As she had no history of excessive alcohol consumption or complication of cholelithiasis, dyslipidemia, or pancreatic neoplasm, drug-induced pancreatitis was suspected. Dexamethasone, which was administered as an antiemetic, was the suspected drug based on the drug administration history and previous report, and dexamethasone was discontinued from the second administration of ddAC. There was subsequently no recurrence of pancreatitis with no increase in serum pancreatic enzyme levels, and it was possible to complete adjuvant-chemotherapy. Alcohol, gallstones, dyslipidemia, and drugs have been reported as causes of pancreatitis; however, steroid-induced acute pancreatitis is extremely rare. We present the first case of acute pancreatitis induced by dexamethasone as the antiemetic.
Collapse
Affiliation(s)
- Hirofumi Ohmura
- Department of Oncology and Social Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Taro Tobo
- Department of Clinical Laboratory Medicine, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yuki Ando
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
7
|
Kutpruek S, Suksri K, Maneethorn P, Semprasert N, Yenchitsomanus PT, Kooptiwut S. Imatinib prevents dexamethasone-induced pancreatic β-cell apoptosis via decreased TRAIL and DR5. J Cell Biochem 2023; 124:1309-1323. [PMID: 37555250 DOI: 10.1002/jcb.30450] [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: 02/22/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023]
Abstract
Prolonged administration of dexamethasone, a potent anti-inflammatory drug, can lead to steroid-induced diabetes. Imatinib, a medication commonly prescribed for chronic myeloid leukemia (CML), has been shown to improve diabetes in CML patients. Our recent study demonstrated that dexamethasone induces pancreatic β-cell apoptosis by upregulating the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptor, death receptor 5 (DR5). We hypothesized that imatinib may protect against dexamethasone-induced pancreatic β-cell apoptosis by reducing the expression of TRAIL and DR5, thereby favorably modulating downstream effectors in apoptotic pathways. We test this hypothesis by assessing the effects of imatinib on dexamethasone-induced apoptosis in rat insulinoma cell line cells. As anticipated, dexamethasone treatment led to increased TRAIL and DR5 expression, as well as an elevation in superoxide production. Conversely, expression of the TRAIL decoy receptor (DcR1) was decreased. Moreover, key effectors in the extrinsic and intrinsic apoptosis pathways, such as B-cell lymphoma 2 (BCL-2) associated X (BAX), nuclear factor kappa B (NF-κb), P73, caspase 8, and caspase 9, were upregulated, while the antiapoptotic protein BCL-2 was downregulated. Interestingly and importantly, imatinib at a concentration of 10 µM reversed the effect of dexamethasone on TRAIL, DR5, DcR1, superoxide production, BAX, BCL-2, NF-κB, P73, caspase 3, caspase 8, and caspase 9. Similar effects of imatinib on dexamethasone-induced TRAIL and DR5 expression were also observed in isolated mouse islets. Taken together, our findings suggest that imatinib protects against dexamethasone-induced pancreatic β-cell apoptosis by reducing TRAIL and DR5 expression and modulating downstream effectors in the extrinsic and intrinsic apoptosis pathways.
Collapse
Affiliation(s)
- Suchanoot Kutpruek
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanchana Suksri
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Petcharee Maneethorn
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Namoiy Semprasert
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Research Department, Division of Molecular Medicine, Mahidol University, Bangkok, Thailand
| | - Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Lam SH, Liu HK, Chung SY, Chang JL, Hong MX, Kuo SC, Liaw CC. Diterpenoids and Their Glycosides from the Stems of Tinospora crispa with Beta-Cell Protective Activity. JOURNAL OF NATURAL PRODUCTS 2023; 86:1437-1448. [PMID: 37200063 DOI: 10.1021/acs.jnatprod.3c00114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Seven previously undescribed diterpenoids, tinocrisposides A-D (1-4) and borapetic acids A (5), B (6), and C (7), together with 16 known compounds, were isolated from the stem of Tinospora crispa (Menispermaceae). The structures of the new isolates were elucidated by spectroscopic and chemical methods. The β-cell protective effect of the tested compounds was examined on insulin-secreting BRIN-BD11 cells under dexamethasone treatment. Diterpene glycosides 12, 14-16, and 18 presented a substantial protective effect on BRIN-BD11 cells treated with dexamethasone in a dose-dependent manner. Compounds 4 and 17 with two sugar moieties exhibited clear protective effects on β-cells.
Collapse
Affiliation(s)
- Sio-Hong Lam
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hui-Kang Liu
- Division of Basic Chinese Medicine, National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
- Ph.D. Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Shih-Yuan Chung
- Department of Marine Biotechnology and Resource, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung 404, Taiwan
| | - Jia-Ling Chang
- Department of Science Application and Dissemination, National Taichung University of Education, Taichung 403, Taiwan
| | - Mao-Xuan Hong
- Department of Marine Biotechnology and Resource, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Sheng-Chu Kuo
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung 404, Taiwan
| | - Chih-Chuang Liaw
- Department of Marine Biotechnology and Resource, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
9
|
Zhang Z, Sun J, Guo M, Yuan X. Progress of new-onset diabetes after liver and kidney transplantation. Front Endocrinol (Lausanne) 2023; 14:1091843. [PMID: 36843576 PMCID: PMC9944581 DOI: 10.3389/fendo.2023.1091843] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Organ transplantation is currently the most effective treatment for end-stage organ failure. Post transplantation diabetes mellitus (PTDM) is a severe complication after organ transplantation that seriously affects the short-term and long-term survival of recipients. However, PTDM is often overlooked or poorly managed in its early stage. This article provides an overview of the incidence, and pathogenesis of and risk factors for PTDM, aiming to gain a deeper understanding of PTDM and improve the quality of life of recipients.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Urology, The People's Hospital of Linyi, Linyi, Shandong, China
| | - Jianyun Sun
- Department of Gastroenterology, The People's Hospital of Linyi, Linyi, Shandong, China
| | - Meng Guo
- National Key Laboratory of Medical Immunology &Institute of Immunology, Navy Medical University, Shanghai, China
| | - Xuemin Yuan
- Department of Gastroenterology, The People's Hospital of Linyi, Linyi, Shandong, China
| |
Collapse
|
10
|
Mehlich A, Bolanowski M, Mehlich D, Witek P. Medical treatment of Cushing's disease with concurrent diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1174119. [PMID: 37139336 PMCID: PMC10150952 DOI: 10.3389/fendo.2023.1174119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Cushing's disease (CD) is a severe endocrine disorder characterized by chronic hypercortisolaemia secondary to an overproduction of adrenocorticotropic hormone (ACTH) by a pituitary adenoma. Cortisol excess impairs normal glucose homeostasis through many pathophysiological mechanisms. The varying degrees of glucose intolerance, including impaired fasting glucose, impaired glucose tolerance, and Diabetes Mellitus (DM) are commonly observed in patients with CD and contribute to significant morbidity and mortality. Although definitive surgical treatment of ACTH-secreting tumors remains the most effective therapy to control both cortisol levels and glucose metabolism, nearly one-third of patients present with persistent or recurrent disease and require additional treatments. In recent years, several medical therapies demonstrated prominent clinical efficacy in the management of patients with CD for whom surgery was non-curative or for those who are ineligible to undergo surgical treatment. Cortisol-lowering medications may have different effects on glucose metabolism, partially independent of their role in normalizing hypercortisolaemia. The expanding therapeutic landscape offers new opportunities for the tailored therapy of patients with CD who present with glucose intolerance or DM, however, additional clinical studies are needed to determine the optimal management strategies. In this article, we discuss the pathophysiology of impaired glucose metabolism caused by cortisol excess and review the clinical efficacy of medical therapies of CD, with particular emphasis on their effects on glucose homeostasis.
Collapse
Affiliation(s)
- Anna Mehlich
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Marek Bolanowski
- Chair and Department of Endocrinology, Diabetes, and Isotope Treatment, Wroclaw Medical University, Wroclaw, Poland
| | - Dawid Mehlich
- Laboratory of Molecular OncoSignalling, International Institute of Molecular Mechanisms and Machines (IMol) Polish Academy of Sciences, Warsaw, Poland
- Doctoral School of Medical University of Warsaw, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Witek
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
- *Correspondence: Przemysław Witek,
| |
Collapse
|
11
|
Salehidoost R, Korbonits M. Glucose and lipid metabolism abnormalities in Cushing's syndrome. J Neuroendocrinol 2022; 34:e13143. [PMID: 35980242 DOI: 10.1111/jne.13143] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/15/2022] [Indexed: 11/30/2022]
Abstract
Prolonged excess of glucocorticoids (GCs) has adverse systemic effects leading to significant morbidities and an increase in mortality. Metabolic alterations associated with the high level of the GCs are key risk factors for the poor outcome. These include GCs causing excess gluconeogenesis via upregulation of key enzymes in the liver, a reduction of insulin sensitivity in skeletal muscle, liver and adipose tissue by inhibiting the insulin receptor signalling pathway, and inhibition of insulin secretion in beta cells leading to dysregulated glucose metabolism. In addition, chronic GC exposure leads to an increase in visceral adipose tissue, as well as an increase in lipolysis resulting in higher circulating free fatty acid levels and in ectopic fat deposition. Remission of hypercortisolism improves these metabolic changes, but very often does not result in full resolution of the abnormalities. Therefore, long-term monitoring of metabolic variables is needed even after the resolution of the excess GC levels.
Collapse
Affiliation(s)
- Rezvan Salehidoost
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
12
|
Yoshihara E. Adapting Physiology in Functional Human Islet Organogenesis. Front Cell Dev Biol 2022; 10:854604. [PMID: 35557947 PMCID: PMC9086403 DOI: 10.3389/fcell.2022.854604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of three-dimensional (3D)-structured functional human islets is expected to be an alternative cell source for cadaveric human islet transplantation for the treatment of insulin-dependent diabetes. Human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer infinite resources for newly synthesized human islets. Recent advancements in hPSCs technology have enabled direct differentiation to human islet-like clusters, which can sense glucose and secrete insulin, and those islet clusters can ameliorate diabetes when transplanted into rodents or non-human primates (NHPs). However, the generated hPSC-derived human islet-like clusters are functionally immature compared with primary human islets. There remains a challenge to establish a technology to create fully functional human islets in vitro, which are functionally and transcriptionally indistinguishable from cadaveric human islets. Understanding the complex differentiation and maturation pathway is necessary to generate fully functional human islets for a tremendous supply of high-quality human islets with less batch-to-batch difference for millions of patients. In this review, I summarized the current progress in the generation of 3D-structured human islets from pluripotent stem cells and discussed the importance of adapting physiology for in vitro functional human islet organogenesis and possible improvements with environmental cues.
Collapse
Affiliation(s)
- Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Delangre E, Liu J, Tolu S, Maouche K, Armanet M, Cattan P, Pommier G, Bailbé D, Movassat J. Underlying mechanisms of glucocorticoid-induced β-cell death and dysfunction: a new role for glycogen synthase kinase 3. Cell Death Dis 2021; 12:1136. [PMID: 34876563 PMCID: PMC8651641 DOI: 10.1038/s41419-021-04419-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 11/08/2022]
Abstract
Glucocorticoids (GCs) are widely prescribed for their anti-inflammatory and immunosuppressive properties as a treatment for a variety of diseases. The use of GCs is associated with important side effects, including diabetogenic effects. However, the underlying mechanisms of GC-mediated diabetogenic effects in β-cells are not well understood. In this study we investigated the role of glycogen synthase kinase 3 (GSK3) in the mediation of β-cell death and dysfunction induced by GCs. Using genetic and pharmacological approaches we showed that GSK3 is involved in GC-induced β-cell death and impaired insulin secretion. Further, we unraveled the underlying mechanisms of GC-GSK3 crosstalk. We showed that GSK3 is marginally implicated in the nuclear localization of GC receptor (GR) upon ligand binding. Furthermore, we showed that GSK3 regulates the expression of GR at mRNA and protein levels. Finally, we dissected the proper contribution of each GSK3 isoform and showed that GSK3β isoform is sufficient to mediate the pro-apoptotic effects of GCs in β-cells. Collectively, in this work we identified GSK3 as a viable target to mitigate GC deleterious effects in pancreatic β-cells.
Collapse
Affiliation(s)
- Etienne Delangre
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Junjun Liu
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
- Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Stefania Tolu
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Kamel Maouche
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Mathieu Armanet
- Cell Therapy Unit, Saint-Louis hospital, AP-HP, and Université de Paris, Paris, France
| | - Pierre Cattan
- Cell Therapy Unit, Saint-Louis hospital, AP-HP, and Université de Paris, Paris, France
| | - Gaëlle Pommier
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Danielle Bailbé
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Jamileh Movassat
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France.
| |
Collapse
|
14
|
Elseady WS, Abd Ellatif RA, Estfanous RS, Emam MN, Keshk WA. New insight on the role of liraglutide in alleviating dexamethasone-induced pancreatic cytotoxicity via improving redox status, autophagy flux, and PI3K/Akt/Nrf2 signaling. Can J Physiol Pharmacol 2021; 99:1217-1225. [PMID: 34197718 DOI: 10.1139/cjpp-2021-0183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic glucocorticoids therapy is commonly complicated by steroid diabetes, although the underlying mechanisms are still elusive. Liraglutide, a glucagon-like peptide-1, was initially found to induce glycemic control and recently it was found to have many pleotropic effects; however, its role in pancreas remains unknown. The present study aims to estimate the protective role of liraglutide on dexamethasone-induced pancreatic cytotoxicity and hyperglycemia, highlighting the possible underlying biochemical, molecular, and cellular mechanisms. Twenty-eight male Wistar rats were involved in this study and were randomly divided into four groups. Group III and IV were treated with 1 mg/kg dexamethasone daily for 10 days. Group II and IV were treated with liraglutide in a dose of 0.8 mg/kg per day for 2 weeks. Pancreatic caspase-9, nuclear factor erythroid 2-related factor 2 (Nrf2), phospho-protein kinase-B (pAkt), and sequestrome 1 (p62) levels were assessed by immunoassay. Moreover, phosphoinositide 3-kinase (PI3K) expression by real-time PCR, microtubule-associated protein light chain 3 (LC3B) expression by immunohistochemistry, glycemic status, β-cell function by homoeostasis model assessment (HOMA) β index, and pancreatic redox status were assessed. Liraglutide improved blood glucose level, β-cell function, pancreatic caspase-9 level, redox status, and autophagy. Additionally, it increased pancreatic PI3K, pAkt, and Nrf2 levels. Moreover, preservation of pancreatic histological and the ultrastructural morphological features of β- and α-cells were observed. In conclusion, liraglutide protected against dexamethasone-induced pancreatic injury and hyperglycemia and decelerated the progression towards steroid diabetes via activating PI3K/Akt/Nrf2 signaling and autophagy flux pathways.
Collapse
Affiliation(s)
- Walaa S Elseady
- Department of Anatomy, Faculty of Medicine, Tanta University, Egypt
| | | | | | - Marwa N Emam
- Department of Physiology, Faculty of Medicine, Tanta University, Egypt
| | - Walaa A Keshk
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Egypt
| |
Collapse
|
15
|
Wang X, Brown NK, Wang B, Shariati K, Wang K, Fuchs S, Melero‐Martin JM, Ma M. Local Immunomodulatory Strategies to Prevent Allo-Rejection in Transplantation of Insulin-Producing Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2003708. [PMID: 34258870 PMCID: PMC8425879 DOI: 10.1002/advs.202003708] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/12/2021] [Indexed: 05/02/2023]
Abstract
Islet transplantation has shown promise as a curative therapy for type 1 diabetes (T1D). However, the side effects of systemic immunosuppression and limited long-term viability of engrafted islets, together with the scarcity of donor organs, highlight an urgent need for the development of new, improved, and safer cell-replacement strategies. Induction of local immunotolerance to prevent allo-rejection against islets and stem cell derived β cells has the potential to improve graft function and broaden the applicability of cellular therapy while minimizing adverse effects of systemic immunosuppression. In this mini review, recent developments in non-encapsulation, local immunomodulatory approaches for T1D cell replacement therapies, including islet/β cell modification, immunomodulatory biomaterial platforms, and co-transplantation of immunomodulatory cells are discussed. Key advantages and remaining challenges in translating such technologies to clinical settings are identified. Although many of the studies discussed are preliminary, the growing interest in the field has led to the exploration of new combinatorial strategies involving cellular engineering, immunotherapy, and novel biomaterials. Such interdisciplinary research will undoubtedly accelerate the development of therapies that can benefit the whole T1D population.
Collapse
Affiliation(s)
- Xi Wang
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Natalie K. Brown
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Bo Wang
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Kaavian Shariati
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Kai Wang
- Department of Cardiac SurgeryBoston Children's HospitalBostonMA02115USA
- Department of SurgeryHarvard Medical SchoolBostonMA02115USA
| | - Stephanie Fuchs
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| | - Juan M. Melero‐Martin
- Department of Cardiac SurgeryBoston Children's HospitalBostonMA02115USA
- Department of SurgeryHarvard Medical SchoolBostonMA02115USA
- Harvard Stem Cell InstituteCambridgeMA02138USA
| | - Minglin Ma
- Department of Biological and Environmental EngineeringCornell UniversityIthacaNY14853USA
| |
Collapse
|
16
|
Barekatain R, Chalvon-Demersay T, McLaughlan C, Lambert W. Intestinal Barrier Function and Performance of Broiler Chickens Fed Additional Arginine, Combination of Arginine and Glutamine or an Amino Acid-Based Solution. Animals (Basel) 2021; 11:2416. [PMID: 34438873 PMCID: PMC8388668 DOI: 10.3390/ani11082416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 01/01/2023] Open
Abstract
Two experiments were conducted to investigate the effect of arginine (Arg); the combination of Arg and glutamine (Gln); as well as an amino acid-based solution (MIX) containing Arg, Gln, threonine (Thr), and grape extract, on performance, intestinal permeability, and expression of selected mechanistic genes. Using 240 male Ross 308 off-sex broiler chickens, four experimental treatments were replicated six times with 10 birds per replicate. The experimental treatments included 5 g/kg Arg, 2.5 g/kg Arg and 2.5 g/kg Gln, and 1 g/kg MIX added to a basal diet as control. In the second study, the four dietary treatments were then given to 24 birds with or without a synthetic glucocorticoid, dexamethasone (DEX), as a gut dysfunction model. Feed conversion ratio was improved by all the supplemented treatments from day 7 to 35 of age (p < 0.001). DEX injections increased (p < 0.001) the intestinal permeability in all treatments, which tended to be reversed by Arg or MIX. Additional Arg, Arg-Gln, and MIX suppressed (p < 0.05) the overexpression of IL-1β generated by DEX. Feeding birds with MIX treatment increased (p < 0.05) expression of SGLT-1 and glutathione synthetase. In conclusion, tested amino acid supplements were effective in improving feed efficiency and restraining intestinal inflammation caused by DEX through IL-1β pathway.
Collapse
Affiliation(s)
- Reza Barekatain
- South Australian Research and Development Institute, Roseworthy Campus, University of Adelaide, Roseworthy, SA 5371, Australia;
| | | | - Clive McLaughlan
- South Australian Research and Development Institute, Roseworthy Campus, University of Adelaide, Roseworthy, SA 5371, Australia;
| | - William Lambert
- METEX NOOVISTAGO, 32 Rue Guersant, 75017 Paris, France; (T.C.-D.); (W.L.)
| |
Collapse
|
17
|
Current Pharmacological Intervention and Medical Management for Diabetic Kidney Transplant Recipients. Pharmaceutics 2021; 13:pharmaceutics13030413. [PMID: 33808901 PMCID: PMC8003701 DOI: 10.3390/pharmaceutics13030413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 01/02/2023] Open
Abstract
Hyperglycemia after kidney transplantation is common in both diabetic and non-diabetic patients. Both pretransplant and post-transplant diabetes mellitus are associated with increased kidney allograft failure and mortality. Glucose management may be challenging for kidney transplant recipients. The pathophysiology and pattern of hyperglycemia in patients following kidney transplantation is different from those with type 2 diabetes mellitus. In patients with pre-existing and post-transplant diabetes mellitus, there is limited data on the management of hyperglycemia after kidney transplantation. The following article discusses the nomenclature and diagnosis of pre- and post-transplant diabetes mellitus, the impact of transplant-related hyperglycemia on patient and kidney allograft outcomes, risk factors and potential pathogenic mechanisms of hyperglycemia after kidney transplantation, glucose management before and after transplantation, and modalities for prevention of post-transplant diabetes mellitus.
Collapse
|
18
|
Primavera R, Razavi M, Kevadiya BD, Wang J, Vykunta A, Di Mascolo D, Decuzzi P, Thakor AS. Enhancing islet transplantation using a biocompatible collagen-PDMS bioscaffold enriched with dexamethasone-microplates. Biofabrication 2021; 13. [PMID: 33455953 DOI: 10.1088/1758-5090/abdcac] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/15/2021] [Indexed: 01/01/2023]
Abstract
Islet transplantation is a promising approach to enable type 1 diabetic patients to attain glycemic control independent of insulin injections. However, up to 60% of islets are lost immediately following transplantation. To improve this outcome, islets can be transplanted within bioscaffolds, however, synthetic bioscaffolds induce an intense inflammatory reaction which can have detrimental effects on islet function and survival. In the present study, we first improved the biocompatibility of polydimethylsiloxane (PDMS) bioscaffolds by coating them with collagen. To reduce the inflammatory response to PDMS bioscaffolds, we then enriched the bioscaffolds with dexamethasone-loaded microplates (DEX-µScaffolds). These DEX-microplates have the ability to release DEX in a sustained manner over 7 weeks within a therapeutic range that does not affect the glucose responsiveness of the islets but which minimizes inflammation in the surrounding microenvironment. The bioscaffold showed excellent mechanical properties that enabled it to resist pore collapse thereby helping to facilitate islet seeding and its handling for implantation, and subsequent engraftment, within the epididymal fat pad (EFP). Following the transplantation of islets into the EFP of diabetic mice using DEX-µScaffolds there was a return in basal blood glucose to normal values by day 4, with normoglycemia maintained for 30 days. Furthermore, these animals demonstrated a normal dynamic response to glucose challenges with histological evidence showing reduced pro-inflammatory cytokines and fibrotic tissue surrounding DEX-µScaffolds at the transplantation site. In contrast, diabetic animals transplanted with either islets alone or islets in bioscaffolds without DEX microplates were not able to regain glycemic control during basal conditions with overall poor islet function. Taken together, our data show that coating PDMS bioscaffolds with collagen, and enriching them with DEX-microplates, significantly prolongs and enhances islet function and survival.
Collapse
Affiliation(s)
- Rosita Primavera
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94305-5119, UNITED STATES
| | - Mehdi Razavi
- University of Central Florida, 6900 Lake Nona Blvd, Orlando, Florida, 32827, UNITED STATES
| | - Bhavesh D Kevadiya
- PEN, University of Nebraska Medical Center, Lab-3064,DRC-1,department of pharmacology and experimental neuroscience, Omaha, Nebraska, 68198, UNITED STATES
| | - Jing Wang
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94304, UNITED STATES
| | - Akshara Vykunta
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94304, UNITED STATES
| | - Daniele Di Mascolo
- Central Research Labs Genova, Istituto Italiano di Tecnologia, Via Morego, 30, Genova, Liguria, 16163, ITALY
| | - Paolo Decuzzi
- Istituto Italiano di Tecnologia, Via Morego, 30, Genova, Liguria, 16163, ITALY
| | - Avnesh S Thakor
- Radiology, Stanford University School of Medicine, 3155 Porter Drive, Stanford, California, 94304, UNITED STATES
| |
Collapse
|
19
|
Molecular Mechanisms of Glucocorticoid-Induced Insulin Resistance. Int J Mol Sci 2021; 22:ijms22020623. [PMID: 33435513 PMCID: PMC7827500 DOI: 10.3390/ijms22020623] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/29/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) are steroids secreted by the adrenal cortex under the hypothalamic-pituitary-adrenal axis control, one of the major neuro-endocrine systems of the organism. These hormones are involved in tissue repair, immune stability, and metabolic processes, such as the regulation of carbohydrate, lipid, and protein metabolism. Globally, GCs are presented as ‘flight and fight’ hormones and, in that purpose, they are catabolic hormones required to mobilize storage to provide energy for the organism. If acute GC secretion allows fast metabolic adaptations to respond to danger, stress, or metabolic imbalance, long-term GC exposure arising from treatment or Cushing’s syndrome, progressively leads to insulin resistance and, in fine, cardiometabolic disorders. In this review, we briefly summarize the pharmacological actions of GC and metabolic dysregulations observed in patients exposed to an excess of GCs. Next, we describe in detail the molecular mechanisms underlying GC-induced insulin resistance in adipose tissue, liver, muscle, and to a lesser extent in gut, bone, and brain, mainly identified by numerous studies performed in animal models. Finally, we present the paradoxical effects of GCs on beta cell mass and insulin secretion by the pancreas with a specific focus on the direct and indirect (through insulin-sensitive organs) effects of GCs. Overall, a better knowledge of the specific action of GCs on several organs and their molecular targets may help foster the understanding of GCs’ side effects and design new drugs that possess therapeutic benefits without metabolic adverse effects.
Collapse
|
20
|
Quattrocelli M, Zelikovich AS, Salamone IM, Fischer JA, McNally EM. Mechanisms and Clinical Applications of Glucocorticoid Steroids in Muscular Dystrophy. J Neuromuscul Dis 2021; 8:39-52. [PMID: 33104035 PMCID: PMC7902991 DOI: 10.3233/jnd-200556] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glucocorticoid steroids are widely used as immunomodulatory agents in acute and chronic conditions. Glucocorticoid steroids such as prednisone and deflazacort are recommended for treating Duchenne Muscular Dystrophy where their use prolongs ambulation and life expectancy. Despite this benefit, glucocorticoid use in Duchenne Muscular Dystrophy is also associated with significant adverse consequences including adrenal suppression, growth impairment, poor bone health and metabolic syndrome. For other forms of muscular dystrophy like the limb girdle dystrophies, glucocorticoids are not typically used. Here we review the experimental evidence supporting multiple mechanisms of glucocorticoid action in dystrophic muscle including their role in dampening inflammation and myofiber injury. We also discuss alternative dosing strategies as well as novel steroid agents that are in development and testing, with the goal to reduce adverse consequences of prolonged glucocorticoid exposure while maximizing beneficial outcomes.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Molecular Cardiovascular Biology Division, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aaron S Zelikovich
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Isabella M Salamone
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Julie A Fischer
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
21
|
Zhao Q, Zhou J, Pan Y, Ju H, Zhu L, Liu Y, Zhang Y. The difference between steroid diabetes mellitus and type 2 diabetes mellitus: a whole-body 18F-FDG PET/CT study. Acta Diabetol 2020; 57:1383-1393. [PMID: 32647998 PMCID: PMC7547981 DOI: 10.1007/s00592-020-01566-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
AIMS Steroid diabetes mellitus (SDM) is a metabolic syndrome caused by an increase in glucocorticoids, and its pathogenesis is unclear. 18F-FDG PET/CT can reflect the glucose metabolism of tissues and organs under living conditions. Here, PET/CT imaging of SDM and type 2 diabetes mellitus (T2DM) rats was used to visualize changes in glucose metabolism in the main glucose metabolizing organs and investigate the pathogenesis of SDM. METHODS SDM and T2DM rat models were established. During this time, PET/CT imaging was used to measure the %ID/g value of skeletal muscle and liver to evaluate glucose uptake. The pancreatic, skeletal muscle and liver were analyzed by immunohistochemistry. RESULTS SDM rats showed increased fasting blood glucose and insulin levels, hyperplasia of islet α and β cells, increased FDG uptake in skeletal muscle accompanied by an up-regulation of PI3Kp85α, IRS-1, and GLUT4, no significant changes in liver uptake, and that glycogen storage in the liver and skeletal muscle increased. T2DM rats showed atrophy of pancreatic islet β cells and decreased insulin levels, significantly reduced FDG uptake and glycogen storage in skeletal muscle and liver. CONCLUSIONS The pathogenesis of SDM is different from that of T2DM. The increased glucose metabolism of skeletal muscle may be related to the increased compensatory secretion of insulin. Glucocorticoids promote the proliferation of islet α cells and cause an increase in gluconeogenesis in the liver, which may cause increased blood glucose.
Collapse
Affiliation(s)
- Qingqing Zhao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Jinxin Zhou
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yu Pan
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Huijun Ju
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Liying Zhu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yang Liu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yifan Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
22
|
Xu W, Cui J, Zhou F, Bai M, Deng R, Wang W. Leonurine protects against dexamethasone-induced cytotoxicity in pancreatic β-cells via PI3K/Akt signaling pathway. Biochem Biophys Res Commun 2020; 529:652-658. [DOI: 10.1016/j.bbrc.2020.05.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/27/2023]
|
23
|
Kuppan P, Kelly S, Polishevska K, Hojanepesov O, Seeberger K, Korbutt GS, Pepper AR. Co-localized immune protection using dexamethasone-eluting micelles in a murine islet allograft model. Am J Transplant 2020; 20:714-725. [PMID: 31650674 DOI: 10.1111/ajt.15662] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/14/2019] [Accepted: 10/07/2019] [Indexed: 01/25/2023]
Abstract
The broad application of ß cell transplantation for type 1 diabetes is hindered by the requisite of lifelong systemic immunosuppression. This study examines the utility of localized islet graft drug delivery to subvert the inflammatory and adaptive immune responses. Herein, we have developed and characterized dexamethasone (Dex) eluting Food and Drug Administration-approved micro-Poly(lactic-co-glycolic acid) micelles and examined their efficacy in a fully major histocompatibility complex-mismatch murine islet allograft model. A clinically relevant dose of 46.6 ± 2.8 μg Dex per graft was confirmed when 2 mg of micelles was implemented. Dex-micelles + CTLA-4-Ig (n = 10) resulted in prolonged allograft function with 80% of the recipients demonstrating insulin independence for 60 days posttransplant compared to 40% in empty micelles + CTLA-4-Ig recipients (n = 10, P = .06). Recipients of this combination therapy (n = 8) demonstrated superior glucose tolerance profiles, compared to empty micelles + CTLA-4-Ig recipients (n = 4, P < .05), and significantly reduced localized intragraft proinflammatory cytokine expression. Histologically, increased insulin positive and FOXP3+ T cells were observed in Dex-micelles + CTLA-4-Ig grafts compared to empty micelles + CTLA-4-Ig grafts (P < .01 and P < .05, respectively). Localized drug delivery via micelles elution has the potential to alter the inflammatory environment, enhances allograft survival, and may be an important adjuvant approach to improve clinical islet transplantation outcomes.
Collapse
Affiliation(s)
- Purushothaman Kuppan
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Sandra Kelly
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Kateryna Polishevska
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Osmanmyrat Hojanepesov
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Seeberger
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Gregory S Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew R Pepper
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Riveline JP, Baz B, Nguewa JL, Vidal-Trecan T, Ibrahim F, Boudou P, Vicaut E, Brac de la Perrière A, Fetita S, Bréant B, Blondeau B, Tardy-Guidollet V, Morel Y, Gautier JF. Exposure to Glucocorticoids in the First Part of Fetal Life is Associated with Insulin Secretory Defect in Adult Humans. J Clin Endocrinol Metab 2020; 105:5609147. [PMID: 31665349 DOI: 10.1210/clinem/dgz145] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/25/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVE High glucocorticoid levels in rodents inhibit development of beta cells during fetal life and lead to insulin deficiency in adulthood. To test whether similar phenomena occur in humans, we compared beta-cell function in adults who were exposed to glucocorticoids during the first part of fetal life with that of nonexposed subjects. RESEARCH DESIGN AND METHODS The study was conducted in 16 adult participants exposed to glucocorticoids during the first part of fetal life and in 16 nonexposed healthy participants with normal glucose tolerance who were matched for age, sex, and body mass index (BMI). Exposed participants had been born to mothers who were treated with dexamethasone 1 to 1.5 mg/day from the sixth gestational week (GW) to prevent genital virilization in children at risk of 21-hydroxylase deficiency. We selected offspring of mothers who stopped dexamethasone before the 18th GW following negative genotyping of the fetus. Insulin and glucagon secretion were measured during an oral glucose tolerance test (OGTT) and graded intravenous (IV) glucose and arginine tests. Insulin sensitivity was measured by hyperinsulinemic-euglycemic-clamp. RESULTS Age, BMI, and anthropometric characteristics were similar in the 2 groups. Insulinogenic index during OGTT and insulin sensitivity during the clamp were similar in the 2 groups. In exposed subjects, insulin secretion during graded IV glucose infusion and after arginine administration decreased by 17% (P = 0.02) and 22% (P = 0.002), respectively, while glucagon secretion after arginine increased. CONCLUSION Overexposure to glucocorticoids during the first part of fetal life is associated with lower insulin secretion at adult age, which may lead to abnormal glucose tolerance later in life.
Collapse
Affiliation(s)
- Jean-Pierre Riveline
- Department of Diabetes and Endocrinology, Lariboisière Hospital, APHP, Paris, France
- Paris Diderot- Paris VII University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Baz Baz
- Department of Diabetes and Endocrinology, Lariboisière Hospital, APHP, Paris, France
| | - Jean-Louis Nguewa
- Department of Diabetes and Endocrinology, Lariboisière Hospital, APHP, Paris, France
| | - Tiphaine Vidal-Trecan
- Department of Diabetes and Endocrinology, Lariboisière Hospital, APHP, Paris, France
| | - Fidaa Ibrahim
- Unit of Hormonal Biology, Department of Biochemistry, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Philippe Boudou
- Unit of Hormonal Biology, Department of Biochemistry, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Eric Vicaut
- Assistance Publique-Hôpitaux de Paris, Clinical Research Unit, Fernand Widal Hospital, Sorbonne Paris Cité, Paris Diderot University, Paris, France
| | - Aude Brac de la Perrière
- Fédération d'endocrinologie Hopital Louis Pradel Groupement Hospitalier Est 28 av Doyen Lepine BRON
| | - Sabrina Fetita
- Department of Diabetes and Endocrinology, Lariboisière Hospital, APHP, Paris, France
| | - Bernadette Bréant
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Bertrand Blondeau
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Véronique Tardy-Guidollet
- Department of Biochemistry and Molecular Biology, Groupement Hospitalier Est 59 Boulevard Pinel Bron, France
| | - Yves Morel
- Department of Biochemistry and Molecular Biology, Groupement Hospitalier Est 59 Boulevard Pinel Bron, France
| | - Jean-François Gautier
- Department of Diabetes and Endocrinology, Lariboisière Hospital, APHP, Paris, France
- Paris Diderot- Paris VII University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
25
|
Calcium Signaling in ß-cell Physiology and Pathology: A Revisit. Int J Mol Sci 2019; 20:ijms20246110. [PMID: 31817135 PMCID: PMC6940736 DOI: 10.3390/ijms20246110] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic beta (β) cell dysfunction results in compromised insulin release and, thus, failed regulation of blood glucose levels. This forms the backbone of the development of diabetes mellitus (DM), a disease that affects a significant portion of the global adult population. Physiological calcium (Ca2+) signaling has been found to be vital for the proper insulin-releasing function of β-cells. Calcium dysregulation events can have a dramatic effect on the proper functioning of the pancreatic β-cells. The current review discusses the role of calcium signaling in health and disease in pancreatic β-cells and provides an in-depth look into the potential role of alterations in β-cell Ca2+ homeostasis and signaling in the development of diabetes and highlights recent work that introduced the current theories on the connection between calcium and the onset of diabetes.
Collapse
|
26
|
Dei-Adomakoh YA, Akpalu J, Yawson AE, Ekem I, Reynolds M, Atiase Y. Steroid-induced dysglycaemia in patients with haematological disorders a ten-year review in a tertiary hospital in Ghana. Ghana Med J 2019; 53:156-162. [PMID: 31481812 PMCID: PMC6697763 DOI: 10.4314/gmj.v53i2.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Glucocorticoids (steroids) play a key role in the management of multiple medical conditions including haematological disorders. This study looked at the prevalence of steroid induced dysglycaemia in patients with haematological disorders receiving steroids as part of their treatment with the view of modifying its use and selection of patients where necessary. Methods A retrospective review of haematology patients on treatment regimens including steroids. Information extracted included, demographic characteristics, clinical information such as age, gender, haematological disorder, type of steroid, daily and cumulative dose of steroid, duration of therapy, family history of diabetes and alcohol use. Results The case records of 351 haematology patients were reviewed. However, eight patients with dysglycaemia before therapy were excluded. The median age of patients was 51.0 ± 26.0(IQR: Interquartile Range) years, with an age range of 13 to 87 years, and a female: male ratio of 1.2: 1 (p= 0.778). The prevalence of Steroid-Induced Dysglycaemia (SID) was 3.79% with a mean diagnosis interval of 8.8 + 2.1 months. Overall, 245 (71.4%) patients were on continuous steroids. Among the 13 patients who developed SID, 11 (84.6%) were on continuous steroids. In the majority of the patients (97.1%) there was no family history of diabetes in a first degree relative. Significant differences were found between patients with normoglycaemia and those with dysglycaemia with respect to age (p=0.049) and duration of steroid therapy (p=0.024). Conclusion The prevalence of steroid-induced dysglycaemia is relatively low among Ghanaian patients with haematological disorders on steroid based chemotherapy. Funding None declared
Collapse
Affiliation(s)
| | - Josephine Akpalu
- Korle Bu teaching hospital, Department of Medicine, Accra, Ghana
| | - Alfred E Yawson
- University of Ghana Medical School, Department of Community, Accra, Ghana.,National AIDS/STI Control Program, Clinical Care Unit, Ghana Health Service, Accra, Ghana
| | - Ivy Ekem
- University of Cape Coast, CoHAS, School of Medical Sciences, Department of Haematology, UCC SMS University of Cape Coast Cape Coast, Cape Coast
| | | | - Yacoba Atiase
- Korle Bu teaching hospital, Department of Medicine, Accra, Ghana
| |
Collapse
|
27
|
Tsai SF, Chen CH. Management of Diabetes Mellitus in Normal Renal Function, Renal Dysfunction and Renal Transplant Recipients, Focusing on Glucagon-Like Peptide-1 Agonist: A Review Based upon Current Evidence. Int J Mol Sci 2019; 20:ijms20133152. [PMID: 31261624 PMCID: PMC6651241 DOI: 10.3390/ijms20133152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/23/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
Diabetes Mellitus (DM) is a leading cause of both Cardiovascular Disease (CVD) and End-stage Renal Disease (ESRD). After 2008, there has been much evidence presented, and recently the guidelines for sugar control have changed to focus on being more disease orientated. GLP-1 Receptor Agonists (GLP-1R) and sodium glucose cotransporter-2 inhibitors are suggested as the first line towards fighting all DM, CVD and ESRD. However, the benefits of GLP-1R in organ transplantation recipients remain very limited. No clinical trials have been designed for this particular population. GLP-1R, a gastrointestinal hormone of the incretin family, possesses antidiabetic, antihypertensive, anti-inflammatory, anti-apoptotic and immunomodulatory actions. There are few drug–drug interactions, with delayed gastric emptying being the major concern. The trough level of tacrolimus may not be significant but should still be closely monitored. There are some reasons which support GLP-1R in recipients seeking glycemic control. Post-transplant DM is due to an impaired β-cell function and glucose-induced glucagon suppression during hyperglycemia, which can be reversed by GLP-1R. GLP-1R infusion tends to relieve immunosuppressant related toxicity. Until now, in some cases, glycemic control and body weight reduction can be anticipated with GLP-1R. Additional renal benefits have also been reported. Side effects of hypoglycemia and gastrointestinal discomfort were rarely reported. In conclusion, GLP-1R could be implemented for recipients while closely monitoring their tacrolimus levels and any potential side effects. Any added benefits, in addition to sugar level control, still require more well-designed studies to prove their existence.
Collapse
Affiliation(s)
- Shang-Feng Tsai
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Department of Life Science, Tunghai University, Taichung 407, Taiwan
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan.
- Department of Life Science, Tunghai University, Taichung 407, Taiwan.
| |
Collapse
|
28
|
Akbib S, Stichelmans J, Stangé G, Ling Z, Assefa Z, Hellemans KH. Glucocorticoids and checkpoint tyrosine kinase inhibitors stimulate rat pancreatic beta cell proliferation differentially. PLoS One 2019; 14:e0212210. [PMID: 30779812 PMCID: PMC6380609 DOI: 10.1371/journal.pone.0212210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cell therapy for diabetes could benefit from the identification of small-molecule compounds that increase the number of functional pancreatic beta cells. Using a newly developed screening assay, we previously identified glucocorticoids as potent stimulators of human and rat beta cell proliferation. We now compare the stimulatory action of these steroid hormones to a selection of checkpoint tyrosine kinase inhibitors that were also found to activate the cell cycle-in beta cells and analyzed their respective effects on DNA-synthesis, beta cell numbers and expression of cell cycle regulators. Our data using glucocorticoids in combination with a receptor antagonist, mifepristone, show that 48h exposure is sufficient to allow beta cells to pass the cell cycle restriction point and to become committed to cell division regardless of sustained glucocorticoid-signaling. To reach the end-point of mitosis another 40h is required. Within 14 days glucocorticoids stimulate up to 75% of the cells to undergo mitosis, which indicates that these steroid hormones act as proliferation competence-inducing factors. In contrast, by correlating thymidine-analogue incorporation to changes in absolute cell numbers, we show that the checkpoint kinase inhibitors, as compared to glucocorticoids, stimulate DNA-synthesis only during a short time-window in a minority of cells, insufficient to give a measurable increase of beta cell numbers. Glucocorticoids, but not the kinase inhibitors, were also found to induce changes in the expression of checkpoint regulators. Our data, using checkpoint kinase-specific inhibitors further point to a role for Chk1 and Cdk1 in G1/S transition and progression of beta cells through the cell cycle upon stimulation with glucocorticoids.
Collapse
Affiliation(s)
- Sarah Akbib
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jordy Stichelmans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Stangé
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Zhidong Ling
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Beta Cell Bank, University Hospital Brussels, Brussels, Belgium
| | - Zerihun Assefa
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine H. Hellemans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| |
Collapse
|
29
|
Shariff AI, Syed S, Shelby RA, Force J, Clarke JM, D'Alessio D, Corsino L. Novel cancer therapies and their association with diabetes. J Mol Endocrinol 2019; 62:R187-R199. [PMID: 30532995 DOI: 10.1530/jme-18-0002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/25/2018] [Indexed: 12/29/2022]
Abstract
Over the last decade, there has been a shift in the focus of cancer therapy from conventional cytotoxic drugs to therapies more specifically directed to cancer cells. These novel therapies include immunotherapy, targeted therapy and precision medicine, each developed in great part with a goal of limiting collateral destruction of normal tissues, while enhancing tumor destruction. Although this approach is sound in theory, even new, specific therapies have some undesirable, 'off target effects', in great part due to molecular pathways shared by neoplastic and normal cells. One such undesirable effect is hyperglycemia, which results from either the loss of immune tolerance and autoimmune destruction of pancreatic β-cells or dysregulation of the insulin signaling pathway resulting in insulin resistance. These distinct pathogenic mechanisms lead to clinical presentations similar to type 1 (T1DM) and type 2 (T2DM) diabetes mellitus. Both types of diabetes have been reported in patients across clinical trials, and data on the mechanism(s) for developing hyperglycemia, prevalence, prognosis and effect on cancer mortality is still emerging. With the rapidly expanding list of clinical indications for new cancer therapies, it is essential to understand the impact of their adverse effects. In this review, we focus on hyperglycemia and diabetes related to cancer therapies, describe what is known about mechanism(s) leading to dysregulated glucose metabolism and provide a guide to management of complex oncology patients with a new diagnosis of diabetes.
Collapse
Affiliation(s)
- Afreen Idris Shariff
- Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sohail Syed
- Virginia Commonwealth University, Richmond, Virginia, USA
| | - Rebecca A Shelby
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jeremy Force
- Division of Medical Oncology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jeffrey Melson Clarke
- Division of Medical Oncology, Duke University School of Medicine, Durham, North Carolina, USA
| | - David D'Alessio
- Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Leonor Corsino
- Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
30
|
Yue Y, Huo F, Yue P, Meng X, Salamanca JC, Escobedo JO, Strongin RM, Yin C. In Situ Lysosomal Cysteine-Specific Targeting and Imaging during Dexamethasone-Induced Apoptosis. Anal Chem 2018; 90:7018-7024. [DOI: 10.1021/acs.analchem.8b01406] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | | | - Ping Yue
- Department of Chemistry, Anhui University, Hefei 230601, China
| | - Xiangming Meng
- Department of Chemistry, Anhui University, Hefei 230601, China
| | - James C. Salamanca
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Jorge O. Escobedo
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Robert M. Strongin
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | | |
Collapse
|
31
|
Bonaventura A, Montecucco F. Steroid-induced hyperglycemia: An underdiagnosed problem or clinical inertia? A narrative review. Diabetes Res Clin Pract 2018; 139:203-220. [PMID: 29530386 DOI: 10.1016/j.diabres.2018.03.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/26/2018] [Accepted: 03/02/2018] [Indexed: 01/08/2023]
Abstract
Corticosteroids are widely diffused drugs. An important side effect is the impairment of glycemic control both in patients with known diabetes and in normoglycemic ones potentially leading to steroid-induced diabetes mellitus (SIDM). In this review based on papers released on PubMed, MEDLINE, and EMBASE from January 2015 to October 2017, we summarized and discussed main updates about the definition, the diagnosis, and the pathophysiology of steroid-induced hyperglycemia (SIH), with a look to new therapies. Main alterations responsible for the diabetogenic effect of corticosteroids are a negative impact on insulin sensitivity along with a derangement on insulin secretion, explaining the typical post-prandial hyperglycemia linked to the promotion of gluconeogenesis. An early and precise diagnosis of SIH and/or SIDM is necessary, but current criteria do not seem sensible enough. As an afterthought, the treatment should be reasoned and tailored according to proposed glycemic thresholds and patient comorbidities, choosing between antidiabetic oral drugs and insulin, the latter being preferable among hospitalized patients. SIDM and SIH are frequent problems, but often underdiagnosed due to old diagnostic criteria. Dedicated guidelines universally shared are mandatory in order to harmonize the treatment of these conditions, thus overtaking single therapeutic strategies mostly arising from literature.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
32
|
King T, Faiman B. Steroid-Associated Side Effects: A Symptom Management Update on Multiple Myeloma Treatment
. Clin J Oncol Nurs 2017; 21:240-249. [PMID: 28315528 DOI: 10.1188/17.cjon.240-249] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND One constant and relatively unchanged aspect of treatment of multiple myeloma (MM) is the use of glucocorticosteroids, or steroids, which can cause a wide range of adverse side effects and harm patients' quality of life.
. OBJECTIVES The objective of this study was to provide updated recommendations on the management of steroid-associated side effects in patients with MM.
. METHODS A study of steroid-associated side effects in MM treatment regimens was reviewed to provide updated recommendations to healthcare professionals. FINDINGS Identifying the side effects of steroids and managing them promptly contribute to the success of steroid-containing regimens for patients with MM.
Collapse
|
33
|
Lemos NE, Brondani LDA, Dieter C, Rheinheimer J, Bouças AP, Leitão CB, Crispim D, Bauer AC. Use of additives, scaffolds and extracellular matrix components for improvement of human pancreatic islet outcomes in vitro: A systematic review. Islets 2017; 9:73-86. [PMID: 28678625 PMCID: PMC5624286 DOI: 10.1080/19382014.2017.1335842] [Citation(s) in RCA: 11] [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] [Received: 03/08/2017] [Revised: 05/16/2017] [Accepted: 05/24/2017] [Indexed: 01/31/2023] Open
Abstract
Pancreatic islet transplantation is an established treatment to restore insulin independence in type 1 diabetic patients. Its success rates have increased lately based on improvements in immunosuppressive therapies and on islet isolation and culture. It is known that the quality and quantity of viable transplanted islets are crucial for the achievement of insulin independence and some studies have shown that a significant number of islets are lost during culture time. Thus, in an effort to improve islet yield during culture period, researchers have tested a variety of additives in culture media as well as alternative culture devices, such as scaffolds. However, due to the use of different categories of additives or devices, it is difficult to draw a conclusion on the benefits of these strategies. Therefore, the aim of this systematic review was to summarize the results of studies that described the use of medium additives, scaffolds or extracellular matrix (ECM) components during human pancreatic islets culture. PubMed and Embase repositories were searched. Of 5083 articles retrieved, a total of 37 articles fulfilled the eligibility criteria and were included in the review. After data extraction, articles were grouped as follows: 1) "antiapoptotic/anti-inflammatory/antioxidant," 2) "hormone," 3) "sulphonylureas," 4) "serum supplements," and 5) "scaffolds or ECM components." The effects of the reviewed additives, ECM or scaffolds on islet viability, apoptosis and function (glucose-stimulated insulin secretion - GSIS) were heterogeneous, making any major conclusion hard to sustain. Overall, some "antiapoptotic/anti-inflammatory/antioxidant" additives decreased apoptosis and improved GSIS. Moreover, islet culture with ECM components or scaffolds increased GSIS. More studies are needed to define the real impact of these strategies in improving islet transplantation outcomes.
Collapse
Affiliation(s)
- Natália Emerim Lemos
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Letícia de Almeida Brondani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristine Dieter
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jakeline Rheinheimer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Paula Bouças
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristiane Bauermann Leitão
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daisy Crispim
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Andrea Carla Bauer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
34
|
Seggelke SA, Lindsay MC, Hazlett I, Sanagorski R, Eckel RH, Low Wang CC. Cardiovascular Safety of Antidiabetic Drugs in the Hospital Setting. Curr Diab Rep 2017; 17:64. [PMID: 28699089 DOI: 10.1007/s11892-017-0884-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE OF REVIEW Patients with diabetes and/or stress hyperglycemia requires good glycemic control in the hospital setting, often requiring the use of glucose-lowering therapy. Standard-of-care dictates that non-insulin therapy be discontinued, with insulin therapy initiated using a basal-bolus approach. However, insulin is associated with a high risk for hypoglycemia and medical errors. Alternatives to insulin are needed in the inpatient setting, but the cardiovascular (CV) safety of non-insulin therapy is a concern. RECENT FINDINGS Most studies of antidiabetic drugs have been performed in the outpatient setting, and except for insulin therapy, trials in the inpatient setting have been insufficient to establish CV safety. Randomized controlled trials support the safety of insulin with more moderate glycemic control in the hospital, when hypoglycemia is minimized. Two recent multicenter randomized controlled clinical trials support the safety of sitagliptin, a dipeptidylpeptidase-4 inhibitor (DPP4i), in hospitalized patients, although the sample sizes were likely too small to detect CV events. Small trials suggest a possible CV benefit of glucagon-like peptide-1 receptor agonist therapy. A paucity of evidence and presence of side effects and cautions with insulin secretagogues, sodium glucose-co-transporter-2 inhibitors, and metformin preclude their routine use in the hospital setting. Available evidence is inadequate to evaluate the CV safety of most antidiabetic drug classes in the hospital setting. However, preliminary data from randomized clinical trials suggest the potential safety of the DPP4i sitagliptin.
Collapse
Affiliation(s)
- Stacey A Seggelke
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, MS8106, RC-1 South, Room 7103, 12801 East 17th Avenue, Aurora, CO, 80045, USA
| | - Mark C Lindsay
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, MS8106, RC-1 South, Room 7103, 12801 East 17th Avenue, Aurora, CO, 80045, USA
| | - Ingrid Hazlett
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, MS8106, RC-1 South, Room 7103, 12801 East 17th Avenue, Aurora, CO, 80045, USA
| | - Rebecca Sanagorski
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, MS8106, RC-1 South, Room 7103, 12801 East 17th Avenue, Aurora, CO, 80045, USA
| | - Robert H Eckel
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, MS8106, RC-1 South, Room 7103, 12801 East 17th Avenue, Aurora, CO, 80045, USA
| | - Cecilia C Low Wang
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, MS8106, RC-1 South, Room 7103, 12801 East 17th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
35
|
Kawamori D, Shirakawa J, Liew CW, Hu J, Morioka T, Duttaroy A, Burkey B, Kulkarni RN. GLP-1 signalling compensates for impaired insulin signalling in regulating beta cell proliferation in βIRKO mice. Diabetologia 2017; 60:1442-1453. [PMID: 28526921 PMCID: PMC5508991 DOI: 10.1007/s00125-017-4303-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/18/2017] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS We aimed to investigate potential interactions between insulin and glucagon-like peptide (GLP)-1 signalling pathways in the regulation of beta cell-cycle dynamics in vivo, in the context of the therapeutic potential of GLP-1 to modulate impaired beta cell function. METHODS Beta cell-specific insulin receptor knockout (βIRKO) mice, which exhibit beta cell dysfunction and an age-dependent decrease in beta cell mass, were treated with the dipeptidyl peptidase-4 inhibitor vildagliptin. Following this, glucose homeostasis and beta cell proliferation were evaluated and underlying molecular mechanisms were investigated. RESULTS The sustained elevation in circulating GLP-1 levels, caused by treatment of the knockout mice with vildagliptin for 6 weeks, significantly improved glucose tolerance secondary to enhanced insulin secretion and proliferation of beta cells. Treating βIRKO beta cell lines with the GLP-1 analogue, exendin-4, promoted Akt phosphorylation and protein expression of cyclins A, D1 and E two- to threefold, in addition to cyclin D2. Pancreases from the vildagliptin-treated βIRKO mice exhibited increased cyclin D1 expression, while cyclin D2 expression was impaired. CONCLUSIONS/INTERPRETATION Activation of GLP-1 signalling compensates for impaired growth factor (insulin) signalling and enhances expression of cyclins to promote beta cell proliferation. Together, these data indicate the potential of GLP-1-related therapies to enhance beta cell proliferation and promote beneficial outcomes in models with dysfunctional beta cells.
Collapse
Affiliation(s)
- Dan Kawamori
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Medical Education Center, Faculty of Medicine, Osaka University, Osaka, Japan
| | - Jun Shirakawa
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
| | - Chong Wee Liew
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang Hu
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
| | - Tomoaki Morioka
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Alokesh Duttaroy
- Cardiovascular & Metabolic Diseases, Novartis Institutes for Biomedical Research, Inc., Cambridge, MA, USA
| | | | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Room 410, One Joslin Place, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
36
|
Dubois-Laforgue D. [Post-transplantation diabetes mellitus in kidney recipients]. Nephrol Ther 2017; 13 Suppl 1:S137-S146. [PMID: 28577736 DOI: 10.1016/j.nephro.2017.01.011] [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: 12/22/2016] [Revised: 01/15/2017] [Accepted: 01/17/2017] [Indexed: 10/19/2022]
Abstract
Post-transplantation diabetes mellitus is defined as diabetes that is diagnosed in grafted patients. It affects 20 to 30 % of kidney transplant recipients, with a high incidence in the first year. The increasing age at transplantation and the rising incidence of obesity may increase its prevalence in the next years. Post-transplantation diabetes mellitus is associated with poor outcomes, such as mortality, cardiovascular events or graft dysfunction. Its occurrence is mainly related to immunosuppressive agents, affecting both insulin secretion and sensibility. Immunosuppressants may be iatrogenic, and as such, induce an early and transient diabetes. They may also precociously determine a permanent diabetes, acting here as a promoting factor in patients proned to the development of type 2 diabetes. Lastly, they may behave, far from transplantation, as an additional risk factor for type 2 diabetes. The screening, management and prognosis of these different subtypes of post-transplantation diabetes mellitus will be different.
Collapse
Affiliation(s)
- Danièle Dubois-Laforgue
- Service de diabétologie, hôpital Cochin-Port Royal, 123, boulevard Port-Royal, 75014 Paris, France; Inserm U1016, institut Cochin, 22, rue Méchain, 75014 Paris, France.
| |
Collapse
|
37
|
Scaroni C, Zilio M, Foti M, Boscaro M. Glucose Metabolism Abnormalities in Cushing Syndrome: From Molecular Basis to Clinical Management. Endocr Rev 2017; 38:189-219. [PMID: 28368467 DOI: 10.1210/er.2016-1105] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/15/2017] [Indexed: 12/13/2022]
Abstract
An impaired glucose metabolism, which often leads to the onset of diabetes mellitus (DM), is a common complication of chronic exposure to exogenous and endogenous glucocorticoid (GC) excess and plays an important part in contributing to morbidity and mortality in patients with Cushing syndrome (CS). This article reviews the pathogenesis, epidemiology, diagnosis, and management of changes in glucose metabolism associated with hypercortisolism, addressing both the pathophysiological aspects and the clinical and therapeutic implications. Chronic hypercortisolism may have pleiotropic effects on all major peripheral tissues governing glucose homeostasis. Adding further complexity, both genomic and nongenomic mechanisms are directly induced by GCs in a context-specific and cell-/organ-dependent manner. In this paper, the discussion focuses on established and potential pathologic molecular mechanisms that are induced by chronically excessive circulating levels of GCs and affect glucose homeostasis in various tissues. The management of patients with CS and DM includes treating their hyperglycemia and correcting their GC excess. The effects on glycemic control of various medical therapies for CS are reviewed in this paper. The association between DM and subclinical CS and the role of screening for CS in diabetic patients are also discussed.
Collapse
Affiliation(s)
- Carla Scaroni
- Endocrinology Unit, Department of Medicine, DIMED, University of Padova, Via Ospedale 105, 35128 Padua, Italy
| | - Marialuisa Zilio
- Endocrinology Unit, Department of Medicine, DIMED, University of Padova, Via Ospedale 105, 35128 Padua, Italy
| | - Michelangelo Foti
- Department of Cell Physiology & Metabolism, Centre Médical Universitaire, 1 Rue Michel Servet, 1211 Genèva, Switzerland
| | - Marco Boscaro
- Endocrinology Unit, Department of Medicine, DIMED, University of Padova, Via Ospedale 105, 35128 Padua, Italy
| |
Collapse
|
38
|
Nayak DK, Saravanan PB, Bansal S, Naziruddin B, Mohanakumar T. Autologous and Allogenous Antibodies in Lung and Islet Cell Transplantation. Front Immunol 2016; 7:650. [PMID: 28066448 PMCID: PMC5179571 DOI: 10.3389/fimmu.2016.00650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/14/2016] [Indexed: 01/02/2023] Open
Abstract
The field of organ transplantation has undoubtedly made great strides in recent years. Despite the advances in donor-recipient histocompatibility testing, improvement in transplantation procedures, and development of aggressive immunosuppressive regimens, graft-directed immune responses still pose a major problem to the long-term success of organ transplantation. Elicitation of immune responses detected as antibodies to mismatched donor antigens (alloantibodies) and tissue-restricted self-antigens (autoantibodies) are two major risk factors for the development of graft rejection that ultimately lead to graft failure. In this review, we describe current understanding on genesis and pathogenesis of antibodies in two important clinical scenarios: lung transplantation and transplantation of islet of Langerhans. It is evident that when compared to any other clinical solid organ or cellular transplant, lung and islet transplants are more susceptible to rejection by combination of allo- and autoimmune responses.
Collapse
Affiliation(s)
- Deepak Kumar Nayak
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | | | - Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | | | | |
Collapse
|
39
|
Xu W, Zhang Y, Bai M, Zhou F, Deng R, Ji X, Zhang J, Liu Y, Zhou L, Wang X. Glucose enhances rat islet function via stimulating CART expression. Biochem Biophys Res Commun 2016; 481:84-89. [PMID: 27823935 DOI: 10.1016/j.bbrc.2016.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 12/30/2022]
Abstract
Cocaine- and amphetamine-regulated transcript (CART) is an anorexigenic peptide widely expressed in the central and peripheral nervous systems, as well as in endocrine cells. CART is markedly upregulated in the β-cells of several rodent models of type-2 diabetes. The stimulatory effect of exogenous CART peptide on insulin secretion is cAMP dependent. Glucose is the most important regulator of islet function. However, the role of CART in glucose-potentiated insulin secretion remains unclear. Here, our results showed that glucose time- and dose-dependently elicited CART mRNA expression in rat islets. Both the glucokinase agonist GKA50 and the long-acting GLP-1 analogue exendin-4 increased CART mRNA expression. The protein kinase A (PKA) inhibitor H89 and the inactivation of cAMP response element-binding protein (CREB) suppressed forskolin-stimulated CART mRNA expression. Furthermore, CART overexpression amplified insulin secretion from rat islets in response to glucose and forskolin, and ameliorated dexamethasone-impaired insulin secretion. These findings suggest that islet-derived CART is involved, at least in part, in high glucose-potentiated pancreatic β-cell function.
Collapse
Affiliation(s)
- Wan Xu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yuqing Zhang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Mengyao Bai
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Feiye Zhou
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Ruyuan Deng
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Xueying Ji
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Juan Zhang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yun Liu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Libin Zhou
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Xiao Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
40
|
Gilor C, Niessen S, Furrow E, DiBartola S. What's in a Name? Classification of Diabetes Mellitus in Veterinary Medicine and Why It Matters. J Vet Intern Med 2016; 30:927-40. [PMID: 27461721 PMCID: PMC5108445 DOI: 10.1111/jvim.14357] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 04/07/2016] [Accepted: 05/16/2016] [Indexed: 12/14/2022] Open
Abstract
Diabetes Mellitus (DM) is a syndrome caused by various etiologies. The clinical manifestations of DM are not indicative of the cause of the disease, but might be indicative of the stage and severity of the disease process. Accurately diagnosing and classifying diabetic dogs and cats by the underlying disease process is essential for current and future studies on early detection, prevention, and treatment of underlying disease. Here, we review the current etiology-based classification of DM and definitions of DM types in human medicine and discuss key points on the pathogenesis of each DM type and prediabetes. We then review current evidence for application of this etiology-based classification scheme in dogs and cats. In dogs, we emphasize the lack of consistent evidence for autoimmune DM (Type 1) and the possible importance of other DM types such as DM associated with exocrine pancreatic disease. While most dogs are first examined because of DM in an insulin-dependent state, early and accurate diagnosis of the underlying disease process could change the long-term outcome and allow some degree of insulin independence. In cats, we review the appropriateness of using the umbrella term of Type 2 DM and differentiating it from DM secondary to other endocrine disease like hypersomatotropism. This differentiation could have crucial implications on treatment and prognosis. We also discuss the challenges in defining and diagnosing prediabetes in cats.
Collapse
Affiliation(s)
- C. Gilor
- Department of Veterinary Clinical SciencesCollege of Veterinary MedicineThe Ohio State UniversityColumbusOH
| | - S.J.M. Niessen
- Department of Clinical Science and ServicesRoyal Veterinary CollegeUniversity of LondonNorth MymmsHertfordshireUK
| | - E. Furrow
- Department of Veterinary Clinical SciencesCollege of Veterinary MedicineUniversity of MinnesotaSt. PaulMN
| | - S.P. DiBartola
- Department of Veterinary Clinical SciencesCollege of Veterinary MedicineThe Ohio State UniversityColumbusOH
| |
Collapse
|
41
|
Adrenal-Derived Hormones Differentially Modulate Intestinal Immunity in Experimental Colitis. Mediators Inflamm 2016; 2016:4936370. [PMID: 27403034 PMCID: PMC4923585 DOI: 10.1155/2016/4936370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 05/19/2016] [Indexed: 01/09/2023] Open
Abstract
The adrenal glands are able to modulate immune responses through neuroimmunoendocrine interactions and cortisol secretion that could suppress exacerbated inflammation such as in inflammatory bowel disease (IBD). Therefore, here we evaluated the role of these glands in experimental colitis induced by 3% dextran sulfate sodium (DSS) in C57BL/6 mice subjected to adrenalectomy, with or without glucocorticoid (GC) replacement. Mice succumbed to colitis without adrenals with a higher clinical score and augmented systemic levels of IL-6 and lower LPS. Furthermore, adrenalectomy negatively modulated systemic regulatory markers. The absence of adrenals resulted in augmented tolerogenic lamina propria dendritic cells but no compensatory local production of corticosterone and decreased mucosal inflammation associated with increased IFN-γ and FasL in the intestine. To clarify the importance of GC in this scenario, GC replacement in adrenalectomized mice restored different markers to the same degree of that observed in DSS group. Finally, this is the first time that adrenal-derived hormones, especially GC, were associated with the differential local modulation of the gut infiltrate, also pointing to a relationship between adrenalectomy and the modulation of systemic regulatory markers. These findings may elucidate some neuroimmunoendocrine mechanisms that dictate colitis outcome.
Collapse
|
42
|
Abstract
In response to stress, the central nervous system initiates a signaling cascade, which leads to the production of glucocorticoids (GCs). GCs act through the glucocorticoid receptor (GR) to coordinate the appropriate cellular response with the primary goal of mobilizing the storage forms of carbon precursors to generate a continuous glucose supply for the brain. Although GCs are critical for maintaining energy homeostasis, excessive GC stimulation leads to a number of undesirable side effects, including hyperglycemia, insulin resistance, fatty liver, obesity, and muscle wasting leading to severe metabolic dysfunction. Summarized below are the diverse metabolic roles of glucocorticoids in energy homeostasis and dysregulation, focusing specifically on glucose, lipid, and protein metabolism.
Collapse
Affiliation(s)
- Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
43
|
Zhang C, Li L, Zhao B, Jiao A, Li X, Sun N, Zhang J. Ghrelin Protects against Dexamethasone-Induced INS-1 Cell Apoptosis via ERK and p38MAPK Signaling. Int J Endocrinol 2016; 2016:4513051. [PMID: 27190513 PMCID: PMC4844876 DOI: 10.1155/2016/4513051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/10/2016] [Accepted: 03/23/2016] [Indexed: 11/17/2022] Open
Abstract
Glucocorticoid excess induces apoptosis of islet cells, which may result in diabetes. In this study, we investigated the protective effect of ghrelin on dexamethasone-induced INS-1 cell apoptosis. Our data showed that ghrelin (0.1 μM) inhibited dexamethasone-induced (0.1 μM) apoptosis of INS-1 cells and facilitated cell proliferation. Moreover, ghrelin upregulated Bcl-2 expression, downregulated Bax expression, and decreased caspase-3 activity. The protective effect of ghrelin against dexamethasone-induced INS-1 cell apoptosis was mediated via growth hormone secretagogue receptor 1a. Further studies revealed that ghrelin increased ERK activation and decreased p38MAPK expression after dexamethasone treatment. Ghrelin-mediated protection of dexamethasone-induced apoptosis of INS-1 cells was attenuated using the ERK inhibitor U0126 (10 μM), and cell viability increased using the p38MAPK inhibitor SB203580 (10 μM). In conclusion, ghrelin could protect against dexamethasone-induced INS-1 cell apoptosis, at least partially via GHS-R1a and the signaling pathway of ERK and p38MAPK.
Collapse
Affiliation(s)
- Chengshuo Zhang
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, First Hospital of China Medical University, Shenyang 110001, China
| | - Le Li
- Hepatobiliary Surgery Department, Chifeng Municipal Hospital, Chifeng 024000, China
| | - Bochao Zhao
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, First Hospital of China Medical University, Shenyang 110001, China
| | - Ao Jiao
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, First Hospital of China Medical University, Shenyang 110001, China
| | - Xin Li
- Department of General Surgery, Fourth Hospital of China Medical University, Shenyang 110032, China
| | - Ning Sun
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, First Hospital of China Medical University, Shenyang 110001, China
| | - Jialin Zhang
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, First Hospital of China Medical University, Shenyang 110001, China
- *Jialin Zhang:
| |
Collapse
|
44
|
Easley JT, Nelson JW, Mellas RE, Sommakia S, Wu C, Trump B, Baker OJ. Aspirin-Triggered Resolvin D1 Versus Dexamethasone in the Treatment of Sjögren's Syndrome-Like NOD/ShiLtJ Mice - A Pilot Study. ACTA ACUST UNITED AC 2015; 1. [PMID: 27110599 DOI: 10.23937/2469-5726/1510027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Resolvin D1 (RvD1) and its aspirin-triggered epimeric form (AT-RvD1) are endogenous lipid mediators (derived from docosahexaenoic acid, DHA) that control the duration and magnitude of inflammation in models of complex diseases. Our previous studies demonstrated that RvD1-mediated signaling pathways are expressed and active in salivary glands from rodents and humans. Furthermore, treatment of salivary cells with RvD1 blocked TNF-α-mediated inflammatory signals and improved epithelial integrity. The purpose of this pilot study was to determine the feasibility of treatment with AT-RvD1 versus dexamethasone (DEX) on inflammation (i.e., lymphocytic infiltration, cytokine expression and apoptosis) observed in submandibular glands (SMG) from the NOD/ShiLtJ Sjögren's syndrome (SS) mouse model before experimenting with a larger population. NOD/ShiLtJ mice were treated intravenously with NaCl (0.9%, negative control), AT-RvD1 (0.01-0.1 mg/kg) or DEX (4.125-8.25 mg/kg) twice a week for 14 weeks beginning at 4 weeks of age. At 18 weeks of age, SMG were collected for pathological analysis and detection of SS-associated inflammatory genes. The AT-RvD1 treatment alone did not affect lymphocytic infiltration seen in NOD/ShiLtJ mice while DEX partially prevented lymphocytic infiltration. Interestingly, both AT-RvD1 and DEX caused downregulation of SS-associated inflammatory genes and reduction of apoptosis. Results from this pilot study suggest that a systemic treatment with AT-RvD1 and DEX alone attenuated inflammatory responses observed in the NOD/ShiLtJ mice; therefore, they may be considered as potential therapeutic tools in treating SS patients when used alone or in combination.
Collapse
Affiliation(s)
- Justin T Easley
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| | - Joel W Nelson
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| | - Rachel E Mellas
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| | - Salah Sommakia
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| | - Chunhua Wu
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| | - Bryan Trump
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| | - Olga J Baker
- School of Dentistry, University of Utah, Salt Lake City, UT 84108-1201, USA
| |
Collapse
|
45
|
Guo B, Zhang W, Xu S, Lou J, Wang S, Men X. GSK-3β mediates dexamethasone-induced pancreatic β cell apoptosis. Life Sci 2015; 144:1-7. [PMID: 26606859 DOI: 10.1016/j.lfs.2015.11.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/29/2015] [Accepted: 11/19/2015] [Indexed: 01/02/2023]
Abstract
AIMS Glucocorticoids, such as dexamethasone, are widely used anti-inflammatory drugs. Their use is frequently associated with the development of steroid- associated diabetes. Pancreatic β-cell dysfunction has been suggested to be one of the main causes of steroid-associated diabetes. However, the mechanism is not fully understood. Glycogen synthase kinase-3β (GSK-3β) is a multifunctional serine/threonine kinase and plays an important role in energy metabolism, cell growth and apoptosis. Therefore, the contribution of GSK-3β in dexamethasone-induced pancreatic β-cell apoptosis was determined in the present study. MAIN METHODS The effect of dexamethasone treatment on rat pancreatic β-cell line (INS-1) apoptosis (determined by TUNEL and Flow Cytometry), generation of reactive oxidative stress (ROS), and the phosphorylation status of GSK-3β was determined. The inhibitory effect of GSK-3β inhibitor-lithium chloride (LiCl) on dexamethasone-induced β-cell apoptosis was also evaluated. KEY FINDINGS Dexamethasone (0.1 μM) treatment induced INS-1 apoptosis, which was associated with increased GSK-3β activation and increased NOX4-derived ROS generation. Pretreatment of INS-1 with LiCl inhibited dexamethasone induced ROS generation and INS-1 apoptosis. SIGNIFICANCE This study provides a new mechanism of Dex induced pancreatic β cell apoptosis and may serve as a new therapeutic option for treating GC induced diabetes.
Collapse
Affiliation(s)
- Bin Guo
- Department of Pathophysiology, North China University of Science and Technology,Tangshan 063000, People's Republic of China
| | - Wenjian Zhang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Shiqing Xu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Jinning Lou
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA.
| | - Xiuli Men
- Department of Pathophysiology, North China University of Science and Technology,Tangshan 063000, People's Republic of China.
| |
Collapse
|
46
|
Regulation of Glucose Homeostasis by Glucocorticoids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215992 DOI: 10.1007/978-1-4939-2895-8_5] [Citation(s) in RCA: 421] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are steroid hormones that regulate multiple aspects of glucose homeostasis. Glucocorticoids promote gluconeogenesis in liver, whereas in skeletal muscle and white adipose tissue they decrease glucose uptake and utilization by antagonizing insulin response. Therefore, excess glucocorticoid exposure causes hyperglycemia and insulin resistance. Glucocorticoids also regulate glycogen metabolism. In liver, glucocorticoids increase glycogen storage, whereas in skeletal muscle they play a permissive role for catecholamine-induced glycogenolysis and/or inhibit insulin-stimulated glycogen synthesis. Moreover, glucocorticoids modulate the function of pancreatic α and β cells to regulate the secretion of glucagon and insulin, two hormones that play a pivotal role in the regulation of blood glucose levels. Overall, the major glucocorticoid effect on glucose homeostasis is to preserve plasma glucose for brain during stress, as transiently raising blood glucose is important to promote maximal brain function. In this chapter we will discuss the current understanding of the mechanisms underlying different aspects of glucocorticoid-regulated mammalian glucose homeostasis.
Collapse
|
47
|
Carlessi R, Lemos NE, Dias AL, Oliveira FS, Brondani LA, Canani LH, Bauer AC, Leitão CB, Crispim D. Exendin-4 protects rat islets against loss of viability and function induced by brain death. Mol Cell Endocrinol 2015; 412:239-50. [PMID: 25976662 DOI: 10.1016/j.mce.2015.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Islet quality loss after isolation from brain-dead donors still hinders the implementation of human islet transplantation for treatment of type 1 diabetes. In this scenario, systemic inflammation elicited by donor brain death (BD) is among the main factors influencing islet viability and functional impairment. Exendin-4 is largely recognized to promote anti-inflammatory and cytoprotective effects on β-cells. Therefore, we hypothesized that administration of exendin-4 to brain-dead donors might improve islet survival and insulin secretory capabilities. Here, using a rat model of BD, we demonstrate that exendin-4 administration to the brain-dead donors increases both islet viability and glucose-stimulated insulin secretion. In this model, exendin-4 treatment produced a significant decrease in interleukin-1β expression in the pancreas. Furthermore, exendin-4 treatment increased the expression of superoxide dismutase-2 and prevented BD-induced elevation in uncoupling protein-2 expression. Such observations were accompanied by a reduction in gene expression of two genes often associated with endoplasmic reticulum (ER) stress response in freshly isolated islets from treated animals, C/EBP homologous protein and immunoglobulin heavy-chain binding protein. As ER stress response has been shown to be triggered by and to participate in cytokine-induced β-cell death, we suggest that exendin-4 might exert its beneficial effects through alleviation of pancreatic inflammation and oxidative stress, which in turn could prevent islet ER stress and β-cell death. Our findings might unveil a novel strategy to preserve islet quality from brain-dead donors. After testing in the human pancreatic islet transplantation setting, this approach might sum to the ongoing effort to achieve consistent and successful single-donor islet transplantation.
Collapse
Affiliation(s)
- Rodrigo Carlessi
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; School of Biomedical Sciences, CHIRI - Biosciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Natália E Lemos
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana L Dias
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Fernanda S Oliveira
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Letícia A Brondani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luis H Canani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andrea C Bauer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Cristiane B Leitão
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
48
|
Chowdhury S, Grimm L, Gong YJK, Wang B, Li B, Srikant CB, Gao ZH, Liu JL. Decreased 11β-Hydroxysteroid Dehydrogenase 1 Level and Activity in Murine Pancreatic Islets Caused by Insulin-Like Growth Factor I Overexpression. PLoS One 2015; 10:e0136656. [PMID: 26305481 PMCID: PMC4549276 DOI: 10.1371/journal.pone.0136656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 08/06/2015] [Indexed: 12/21/2022] Open
Abstract
We have reported a high expression of IGF-I in pancreatic islet β-cells of transgenic mice under the metallothionein promoter. cDNA microarray analysis of the islets revealed that the expression of 82 genes was significantly altered compared to wild-type mice. Of these, 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), which is responsible for the conversion of inert cortisone (11-dehydrocorticosterone, DHC in rodents) to active cortisol (corticosterone) in the liver and adipose tissues, has not been identified previously as an IGF-I target in pancreatic islets. We characterized the changes in its protein level, enzyme activity and glucose-stimulated insulin secretion. In freshly isolated islets, the level of 11β-HSD1 protein was significantly lower in MT-IGF mice. Using dual-labeled immunofluorescence, 11β-HSD1 was observed exclusively in glucagon-producing, islet α-cells but at a lower level in transgenic vs. wild-type animals. MT-IGF islets also exhibited reduced enzymatic activities. Dexamethasone (DEX) and DHC inhibited glucose-stimulated insulin secretion from freshly isolated islets of wild-type mice. In the islets of MT-IGF mice, 48-h pre-incubation of DEX caused a significant decrease in insulin release, while the effect of DHC was largely blunted consistent with diminished 11β-HSD1 activity. In order to establish the function of intracrine glucocorticoids, we overexpressed 11β-HSD1 cDNA in MIN6 insulinoma cells, which together with DHC caused apoptosis and a significant decrease in proliferation. Both effects were abolished with the treatment of an 11β-HSD1 inhibitor. Our results demonstrate an inhibitory effect of IGF-I on 11β-HSD1 expression and activity within the pancreatic islets, which may mediate part of the IGF-I effects on cell proliferation, survival and insulin secretion.
Collapse
Affiliation(s)
- Subrata Chowdhury
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Larson Grimm
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Ying Jia Kate Gong
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Beixi Wang
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Bing Li
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Coimbatore B. Srikant
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Zu-hua Gao
- Department of Pathology, the Research Institute of McGill University Health Centre, Montreal, Canada
| | - Jun-Li Liu
- Fraser Laboratories for Diabetes Research, Department of Medicine, the Research Institute of McGill University Health Centre, Montreal, Canada
- Montreal Diabetes Research Centre, Montreal, Canada
| |
Collapse
|
49
|
Takahashi K, Ogura N, Tomoki R, Eda T, Okada H, Kato R, Iwai S, Ito K, Kuyama K, Kondoh T. Applicability of human dental follicle cells to bone regeneration without dexamethasone: an in vivo pilot study. Int J Oral Maxillofac Surg 2015; 44:664-9. [DOI: 10.1016/j.ijom.2014.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 10/27/2014] [Accepted: 11/07/2014] [Indexed: 11/24/2022]
|
50
|
Glucocorticoids suppress GLP-1 secretion: possible contribution to their diabetogenic effects. Clin Sci (Lond) 2015; 129:405-14. [PMID: 25853863 DOI: 10.1042/cs20140719] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/08/2015] [Indexed: 12/23/2022]
Abstract
Evidence indicates that subtle abnormalities in GC (glucocorticoid) plasma concentrations and/or in tissue sensitivity to GCs are important in the metabolic syndrome, and it is generally agreed that GCs induce insulin resistance. In addition, it was recently reported that short-term exposure to GCs reduced the insulinotropic effects of the incretin GLP-1 (glucagon-like peptide 1). However, although defective GLP-1 secretion has been correlated with insulin resistance, potential direct effects of GCs on GLP-1-producing L-cell function in terms of GLP-1 secretion and apoptosis have not been studied in any greater detail. In the present study, we sought to determine whether GCs could exert direct effects on GLP-1-producing L-cells in terms of GLP-1 secretion and cell viability. We demonstrate that the GR (glucocorticoid receptor) is expressed in GLP-1-producing cells, where GR activation in response to dexamethasone induces SGK1 (serum- and glucocorticoid-inducible kinase 1) expression, but did not influence preproglucagon expression or cell viability. In addition, dexamethasone treatment of enteroendocrine GLUTag cells reduced GLP-1 secretion induced by glucose, 2-deoxy-D-glucose, fructose and potassium, whereas the secretory response to a phorbol ester was unaltered. Furthermore, in vivo administration of dexamethasone to rats reduced the circulating levels of GLP-1 concurrent with induction of insulin resistance and glucose intolerance. We can conclude that GR activation in GLP-1-producing cells will diminish the secretory responsiveness of these cells to subsequent carbohydrate stimulation. These effects may not only elucidate the pathogenesis of steroid diabetes, but could ultimately contribute to the identification of novel molecular targets for controlling incretin secretion.
Collapse
|