1
|
Chen J, Hou X, Yang Y, Wang C, Zhou J, Miao J, Gong F, Ge F, Chen W. Immune checkpoint inhibitors-induced diabetes mellitus (review). Endocrine 2024; 86:451-458. [PMID: 38955861 DOI: 10.1007/s12020-024-03942-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have become extensively utilized in the early-stage treatment of various cancers, offering additional therapeutic possibilities for patients with advanced cancer. However, certain patient populations are susceptible to experiencing toxic adverse effects from ICIs, such as thyrotoxicosis, rashes, among others. Specifically, ICIDM, induced by immune checkpoint inhibitors, exhibits characteristics similar to insulin-dependent diabetes mellitus (Type 1 Diabetes Mellitus, T1DM). ICIDM is characterized by a rapid onset and may coincide with severe ketoacidosis. Despite a favorable response to insulin therapy, patients typically require lifelong insulin dependence. After discussing the autoimmune adverse effects and the specifics of ICIs-induced diabetes mellitus (ICIDM), it is important to note that certain patient populations are particularly susceptible to experiencing toxic adverse effects from ICIs. Specifically, ICIDM, which is triggered by immune checkpoint inhibitors, mirrors the characteristics of insulin-dependent diabetes mellitus (Type 1 Diabetes Mellitus, T1DM). This article conducts an in-depth analysis of the literature to explore the pathogenesis, disease progression, and treatment strategies applicable to diabetes induced by immune checkpoint inhibitors (ICIDM).
Collapse
Affiliation(s)
- Jiayi Chen
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Xiaochen Hou
- Academy of Biomedical Engineering, Kunming Medical University, Yunnan, 650500, China
| | - Yang Yang
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Chenxi Wang
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Jie Zhou
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Jingge Miao
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Fuhong Gong
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Fei Ge
- Department of Breast Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| | - Wenlin Chen
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China.
| |
Collapse
|
2
|
Azulay RS, Rodrigues V, Lago DCF, de Almeida AGFP, de Abreu JDMF, Matos L, Andrade C, Nascimento GC, Magalhães M, Facundo A, de Oliveira Neto CP, Sá AG, Silva DA, Gomes MB, Faria MDS. Relationship Between C-Peptide Levels, Clinical Features, and Serum Data in a Brazilian Type 1 Diabetes Population with Large Variations in Genomic Ancestry. Int J Mol Sci 2024; 25:11144. [PMID: 39456927 PMCID: PMC11508759 DOI: 10.3390/ijms252011144] [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: 09/08/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Type 1 diabetes (T1D) is a chronic disease characterized by the immune-mediated destruction of the pancreatic beta cells responsible for insulin production. The secreted insulin and C-peptide are equimolar. Due to its longer half-life, C-peptide has become a safer means of assessing the pancreatic reserve. C-peptide levels were evaluated in a population of patients with T1D, focusing on the relationship between this variable and other factors. In addition, the influence of C-peptide on metabolic control and microvascular complications was investigated. This cross-sectional study included 95 patients who had been diagnosed with T1D at least five years earlier. These patients were evaluated using a clinical demographic survey, anthropometric data, laboratory tests, and fundoscopy. This study showed that 29.5% of patients had residual insulin secretion, which correlated directly with their age at diagnosis. No statistically significant differences in metabolic control or microvascular complications were observed between the C-peptide level groups. In addition, our results indicate that ancestry does not influence the persistence of residual C-peptide function in our highly mixed population. It is recommended that future research consider incorporating new variables, such as HLA and pancreatic autoimmunity, as factors that may influence residual β-cell function.
Collapse
Affiliation(s)
- Rossana Sousa Azulay
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Vandilson Rodrigues
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Débora Cristina Ferreira Lago
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Ana Gregória Ferreira Pereira de Almeida
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Joana D’Arc Matos França de Abreu
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Lincoln Matos
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Caio Andrade
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis 65085-580, Brazil
| | - Gilvan Cortês Nascimento
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Marcelo Magalhães
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Alexandre Facundo
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Clariano Pires de Oliveira Neto
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Adriana Guimarães Sá
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
| | - Dayse Aparecida Silva
- DNA Diagnostic Laboratory (LDD), Rio de Janeiro State University (UERJ), Rio de Janeiro 20550-900, Brazil;
| | - Marília Brito Gomes
- Diabetes Unit, Rio de Janeiro State University (UERJ), Rio de Janeiro 20551-030, Brazil;
| | - Manuel dos Santos Faria
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís 65020-070, Brazil; (D.C.F.L.); (A.G.F.P.d.A.); (J.D.M.F.d.A.); (L.M.); (G.C.N.); (A.F.); (C.P.d.O.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (C.A.); (M.M.); (A.G.S.)
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis 65085-580, Brazil
| |
Collapse
|
3
|
Jansen TJP, Tokgöz S, Buitinga M, van Lith SAM, Joosten L, Frielink C, Smeets EMM, Stommel MWJ, van der Kolk MB, de Galan BE, Brom M, Boss M, Gotthardt M. Validation of radiolabelled exendin for beta cell imaging by ex vivo autoradiography and immunohistochemistry of human pancreas. EJNMMI Res 2024; 14:96. [PMID: 39405026 PMCID: PMC11480297 DOI: 10.1186/s13550-024-01159-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Estimation of beta cell mass is currently restricted to evaluating pancreatic tissue samples, which provides limited information. A non-invasive imaging technique that reliably quantifies beta cell mass enables monitoring of changes of beta cell mass during the progression of diabetes mellitus and may contribute to monitoring of therapy effectiveness. We assessed the specificity of radiolabelled exendin for beta cell mass quantification in humans. Fourteen adults with pancreas tumours were injected with 111In-labeled exendin-4 prior to pancreatic resection. In resected pancreas tissue, endocrine-exocrine ratios of tracer uptake were determined by digital autoradiography and accumulation of 111In-labeled exendin-4 was compared to insulin and GLP-1 receptor staining. Of four participants, abdominal single photon emission computed tomography/computed tomography (SPECT/CT) images were acquired to quantify pancreatic uptake in vivo RESULTS: Tracer uptake was predominantly present in the endocrine pancreas (endocrine-exocrine ratio: 3.6 [2.8-10.8]. Tracer accumulation showed overlap with insulin-positive regions, which overlapped with GLP-1 receptor positive areas. SPECT imaging showed pancreatic uptake of radiolabelled exendin in three participants. CONCLUSION Radiolabelled exendin specifically accumulates in the islets of Langerhans in human pancreas tissue. The clear overlap between regions positive for insulin and the GLP-1 receptor substantiate the beta cell specificity of the tracer. Radiolabelled exendin is therefore a valuable imaging agent for human beta cell mass quantification and has the potential to be used for a range of applications, including improvement of diabetes treatment by assessment of the effects of current and novel diabetes therapies on the beta cell mass. TRIAL REGISTRATION ClinicalTrials.gov NCT03889496, registered 26,032,019, URL https://clinicaltrials.gov/study/NCT03889496?term=NCT03889496 . CLINICALTRIALS gov NCT04733508, registered 02022021, URL https://clinicaltrials.gov/study/NCT04733508 .
Collapse
Affiliation(s)
- Theodorus J P Jansen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sevilay Tokgöz
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mijke Buitinga
- Nutrition and Movement Sciences, Maastricht University, Maastricht, The Netherlands
- Radiology and Nuclear Medicine, Maastricht UMC+, Maastricht, The Netherlands
| | - Sanne A M van Lith
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cathelijne Frielink
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esther M M Smeets
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Bastiaan E de Galan
- Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Internal Medicine, Maastricht UMC+, Maastricht, The Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands
| | - Maarten Brom
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marti Boss
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
4
|
Ikegami H, Noso S. Genetics of type-1 diabetes. Diabetol Int 2024; 15:688-698. [PMID: 39469551 PMCID: PMC11512969 DOI: 10.1007/s13340-024-00754-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/06/2024] [Indexed: 10/30/2024]
Abstract
Type-1 diabetes is a multifactorial disease characterized by genetic and environmental factors that contribute to its development and progression. Despite progress in the management of type-1 diabetes, the final goal of curing the disease is yet to be achieved. To establish effective methods for the prevention, intervention, and cure of the disease, the molecular mechanisms and pathways involved in its development and progression should be clarified. One effective approach is to identify genes responsible for disease susceptibility and apply information obtained from the function of genes in disease etiology for the protection, intervention, and cure of type-1 diabetes. In this review, we discuss the genetic basis of type-1 diabetes, along with prospects for its prevention, intervention, and cure for type-1 diabetes.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Professor Emeritus, Kindai University, Osaka-sayama, Japan
- Director of Health Administration Center and Nikkei Clinic, Human Resources, Nikkei Inc. Osaka Head Office, Osaka, Japan
| | - Shinsuke Noso
- Department of Endocrinology, Metabolism and Diabetes, Faculty of Medicine, Kindai University, Osaka-sayama, Japan
| |
Collapse
|
5
|
Glorioso KE, Stancill JS. Ex-CyT-ing Applications of Single-cell CyTOF to Human Pancreatic Islets in Diabetes Research. Endocrinology 2024; 165:bqae122. [PMID: 39276032 PMCID: PMC11448324 DOI: 10.1210/endocr/bqae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Affiliation(s)
- Kathryn E Glorioso
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jennifer S Stancill
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Niu F, Liu W, Ren Y, Tian Y, Shi W, Li M, Li Y, Xiong Y, Qian L. β-cell neogenesis: A rising star to rescue diabetes mellitus. J Adv Res 2024; 62:71-89. [PMID: 37839502 PMCID: PMC11331176 DOI: 10.1016/j.jare.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/08/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Diabetes Mellitus (DM), a chronic metabolic disease characterized by elevated blood glucose, is caused by various degrees of insulin resistance and dysfunctional insulin secretion, resulting in hyperglycemia. The loss and failure of functional β-cells are key mechanisms resulting in type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). AIM OF REVIEW Elucidating the underlying mechanisms of β-cell failure, and exploring approaches for β-cell neogenesis to reverse β-cell dysfunction may provide novel strategies for DM therapy. KEY SCIENTIFIC CONCEPTS OF REVIEW Emerging studies reveal that genetic susceptibility, endoplasmic reticulum (ER) stress, oxidative stress, islet inflammation, and protein modification linked to multiple signaling pathways contribute to DM pathogenesis. Over the past few years, replenishing functional β-cell by β-cell neogenesis to restore the number and function of pancreatic β-cells has remarkably exhibited a promising therapeutic approach for DM therapy. In this review, we provide a comprehensive overview of the underlying mechanisms of β-cell failure in DM, highlight the effective approaches for β-cell neogenesis, as well as discuss the current clinical and preclinical agents research advances of β-cell neogenesis. Insights into the challenges of translating β-cell neogenesis into clinical application for DM treatment are also offered.
Collapse
Affiliation(s)
- Fanglin Niu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wenxuan Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Department of Neurology, Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Wenzhen Shi
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Medical Research Center, the affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Man Li
- Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Yujia Li
- Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
7
|
Granlund L, Lundberg M. Loss of insulin-expressing extra-islet cells in type 1 diabetes is accompanied with increased number of glucagon-expressing extra-islet cells. Virchows Arch 2024:10.1007/s00428-024-03842-4. [PMID: 38922355 DOI: 10.1007/s00428-024-03842-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024]
Abstract
The presence of remaining insulin-positive cells in type 1 diabetes (T1D) is well-known. These cells are part of islets or appear as extra-islet insulin-positive cells scattered in the exocrine parenchyma. The latter are poorly described, and the presence of scattered endocrine cells expressing other islet hormones than insulin has not been explored. This study aimed to compare the extra-islet insulin- or glucagon-positive cells concerning their frequency, transcription-factor expression, and mitotic activity in subjects with and without T1D. Multispectral imaging was used to examine extra-islet cells by staining for insulin, glucagon, ARX, PDX1, and Ki67. This was done in well-preserved pancreatic tissue obtained from heart-beating organ donors with or without T1D. In three T1D donors, lobes with insulin-containing islets (ICI) were found. Within these, a higher frequency of extra-islet insulin-positive cells was observed compared to lobes with insulin-deficient islets (IDI). Increased frequency of glucagon-positive extra-islet cells was observed in donors with T1D (median 53 cells/mm2) when compared with non-diabetic donors (11 cells/mm2, p = 0.004). Proliferating endocrine cells were present in donors with, and without T1D, as demonstrated by Ki67-positive staining (0-3% of the cells expressing insulin or glucagon). The reduced frequency of extra-islet insulin-positive cells in lobes with IDI in donors with T1D suggests that the pathological mechanism causing beta cell demise in T1D affects entire lobes. The presence of an increased frequency of glucagon-positive extra-islet cells supports the notion of a preserved capacity to regenerate the endocrine pancreas in donors with T1D.
Collapse
Affiliation(s)
- Louise Granlund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Marcus Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Webster KL, Mirmira RG. Beta cell dedifferentiation in type 1 diabetes: sacrificing function for survival? Front Endocrinol (Lausanne) 2024; 15:1427723. [PMID: 38904049 PMCID: PMC11187278 DOI: 10.3389/fendo.2024.1427723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
The pathogeneses of type 1 and type 2 diabetes involve the progressive loss of functional beta cell mass, primarily attributed to cellular demise and/or dedifferentiation. While the scientific community has devoted significant attention to unraveling beta cell dedifferentiation in type 2 diabetes, its significance in type 1 diabetes remains relatively unexplored. This perspective article critically analyzes the existing evidence for beta cell dedifferentiation in type 1 diabetes, emphasizing its potential to reduce beta cell autoimmunity. Drawing from recent advancements in both human studies and animal models, we present beta cell identity as a promising target for managing type 1 diabetes. We posit that a better understanding of the mechanisms of beta cell dedifferentiation in type 1 diabetes is key to pioneering interventions that balance beta cell function and immunogenicity.
Collapse
Affiliation(s)
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
9
|
Latres E, Greenbaum CJ, Oyaski ML, Dayan CM, Colhoun HM, Lachin JM, Skyler JS, Rickels MR, Ahmed ST, Dutta S, Herold KC, Marinac M. Evidence for C-Peptide as a Validated Surrogate to Predict Clinical Benefits in Trials of Disease-Modifying Therapies for Type 1 Diabetes. Diabetes 2024; 73:823-833. [PMID: 38349844 DOI: 10.2337/dbi23-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
Type 1 diabetes is a chronic autoimmune disease in which destruction of pancreatic β-cells causes life-threatening metabolic dysregulation. Numerous approaches are envisioned for new therapies, but limitations of current clinical outcome measures are significant disincentives to development efforts. C-peptide, a direct byproduct of proinsulin processing, is a quantitative biomarker of β-cell function that is not cleared by the liver and can be measured in the peripheral blood. Studies of quantitative measures of β-cell function have established a predictive relationship between stimulated C-peptide as a measure of β-cell function and clinical benefits. C-peptide levels at diagnosis are often high enough to afford glycemic control benefits associated with protection from end-organ complications of diabetes, and even lower levels offer protection from severe hypoglycemia in type 1 diabetes, as observed in large prospective cohort studies and interventional trials of islet transplantation. These observations support consideration of C-peptide not just as a biomarker of β-cell function but also as a specific, sensitive, feasible, and clinically meaningful outcome defining β-cell preservation or restoration for clinical trials of disease-modifying therapies. Regulatory acceptance of C-peptide as a validated surrogate for demonstration of efficacy would greatly facilitate development of disease-modifying therapies for type 1 diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | | | | | - Helen M Colhoun
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
| | - John M Lachin
- Biostatistics Center, George Washington University, Rockville, MD
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami, Miami, FL
| | - Michael R Rickels
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Simi T Ahmed
- New York Stem Cell Foundation Research Institute, New York, NY
| | | | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale School of Medicine, New Haven, CT
| | | |
Collapse
|
10
|
Uitbeijerse BS, Nijhoff MF, de Koning EJP. Comparison of an oral mixed meal plus arginine and intravenous glucose, GLP-1 plus arginine to unmask residual islet function in longstanding type 1 diabetes. Am J Physiol Endocrinol Metab 2024; 326:E673-E680. [PMID: 38446636 PMCID: PMC11376486 DOI: 10.1152/ajpendo.00030.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Residual beta cells are present in most patients with longstanding type 1 diabetes but it is unknown whether these beta cells react normally to different stimuli. Moreover a defect in proinsulin conversion and abnormal alpha cell response are also part of the islet dysfunction. A three-phase [euglycemia, hyperglycemia, and hyperglycemia + glucagon-like peptide 1 (GLP-1)] clamp was performed in patients with longstanding type 1 diabetes. Intravenous arginine boluses were administered at the end of each phase. On another day, a mixed meal stimulation test with a subsequent intravenous arginine bolus was performed. C-peptide was detectable in a subgroup of subjects at baseline (2/15) or only after stimulation (3/15). When detectable, C-peptide increased 2.9-fold [95% CI: 1.2-7.1] during the hyperglycemia phase and 14.1-fold [95% CI: 3.1-65.2] during the hyperglycemia + GLP-1 phase, and 22.3-fold [95% CI: 5.6-89.1] during hyperglycemia + GLP-1 + arginine phase when compared with baseline. The same subset of patients with a C-peptide response were identified during the mixed meal stimulation test as during the clamp. There was an inhibition of glucagon secretion (0.72-fold, [95% CI: 0.63-0.84]) during the glucose clamp irrespective of the presence of detectable beta cell function. Proinsulin was only present in a subset of subjects with detectable C-peptide (3/15) and proinsulin mimicked the C-peptide response to the different stimuli when detectable. Residual beta cells in longstanding type 1 diabetes respond adequately to different stimuli and could be of clinical benefit.NEW & NOTEWORTHY If beta cell function is detectable, the beta cells react relatively normal to the different stimuli except for the first phase response to intravenous glucose. An oral mixed meal followed by an intravenous arginine bolus can identify residual beta cell function/mass as well as the more commonly used glucose potentiated arginine-induced insulin secretion during a hyperglycemic clamp.
Collapse
Affiliation(s)
- Bas S Uitbeijerse
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Michiel F Nijhoff
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Bourgeois S, Coenen S, Degroote L, Willems L, Van Mulders A, Pierreux J, Heremans Y, De Leu N, Staels W. Harnessing beta cell regeneration biology for diabetes therapy. Trends Endocrinol Metab 2024:S1043-2760(24)00082-1. [PMID: 38644094 DOI: 10.1016/j.tem.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024]
Abstract
The pandemic scale of diabetes mellitus is alarming, its complications remain devastating, and current treatments still pose a major burden on those affected and on the healthcare system as a whole. As the disease emanates from the destruction or dysfunction of insulin-producing pancreatic β-cells, a real cure requires their restoration and protection. An attractive strategy is to regenerate β-cells directly within the pancreas; however, while several approaches for β-cell regeneration have been proposed in the past, clinical translation has proven challenging. This review scrutinizes recent findings in β-cell regeneration and discusses their potential clinical implementation. Hereby, we aim to delineate a path for innovative, targeted therapies to help shift from 'caring for' to 'curing' diabetes.
Collapse
Affiliation(s)
- Stephanie Bourgeois
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Sophie Coenen
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Laure Degroote
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Lien Willems
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Annelore Van Mulders
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Julie Pierreux
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Yves Heremans
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Nico De Leu
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; Endocrinology, Universiteit Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium; Endocrinology, ASZ Aalst, 9300 Aalst, Belgium.
| | - Willem Staels
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; Pediatric Endocrinology, Department of Pediatrics, KidZ Health Castle, Universiteit Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium.
| |
Collapse
|
12
|
Strage EM, Ley C, Westermark GT, Tengholm A. Insulin release from isolated cat islets of Langerhans. Domest Anim Endocrinol 2024; 87:106836. [PMID: 38141375 DOI: 10.1016/j.domaniend.2023.106836] [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] [Received: 06/08/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Feline diabetes mellitus is a common endocrine disease with increasing prevalence. It shows similarities with human type 2 diabetes and is characterized by insulin resistance and deficient insulin secretion. Moreover, cats and humans belong to the very few species that form amyloid depositions in the pancreatic islets. However, little is known about cat islet function and no studies have addressed insulin secretion from isolated islets ex vivo. The aim of this study was to establish a protocol for isolation of islets of Langerhans from pancreata of cats euthanized due to disease, and to evaluate insulin secretion responses to various physiological and pharmacological stimuli. Collagenase digestion of pancreatic tissue from 13 non-diabetic cats and two cats with diabetic ketoacidosis yielded individual islets surrounded by a layer of exocrine tissue that was reduced after two days in culture. Histological examination showed islet amyloid in pancreatic biopsies from most non-diabetic and in one diabetic cat. Islets from non-diabetic cats cultured at 5.5 mM glucose responded with increased insulin secretion to 16.7 mM glucose, 30 mM K+ and 20 µM of the sulfonylurea glipizide (2-3 times basal secretion at 3 mM glucose). The glucagon-like peptide-1 receptor agonist exendin-4 (100 nM) had no effect under basal conditions but potentiated glucose-triggered insulin release. Only one of nine islet batches from diabetic cats released detectable amounts of insulin, which was enhanced by exendin-4. Culture of islets from non-diabetic cats at 25 mM glucose impaired secretion both in response to glucose and K+ depolarization. In conclusion, we describe a procedure for isolation of islets from cat pancreas biopsies and demonstrate that isolated cat islets secrete insulin in response to glucose and antidiabetic drugs. The study provides a basis for future ex vivo studies of islet function relevant to the understanding of the pathophysiology and treatment of feline diabetes.
Collapse
Affiliation(s)
- Emma M Strage
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, P.O. Box 7054, Uppsala SE-750 07, Sweden.
| | - Cecilia Ley
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, P.O. Box 7028, Uppsala SE-750 07, Sweden; Department of Pathology and Wildlife Diseases, National Veterinary Institute (SVA), Uppsala SE-751 89, Sweden
| | - Gunilla T Westermark
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, P.O. Box 571, Uppsala SE-751 23, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, P.O. Box 571, Uppsala SE-751 23, Sweden
| |
Collapse
|
13
|
Davis TME, Peters KE, Davis W. Use of a type 1 genetic risk score for classification of diabetes type in young Australian adults: the Fremantle Diabetes Study Phase II. Intern Med J 2024; 54:494-498. [PMID: 38224531 DOI: 10.1111/imj.16328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
The applicability of a UK-validated genetic risk score (GRS) was assessed in 158 participants in the Fremantle Diabetes Study Phase II diagnosed between 20 and <40 years of age with type 1 or type 2 diabetes or latent autoimmune diabetes of adults (LADA). For type 1 versus type 2/LADA, the area under the receiver operating characteristic curve (AUC) was highest for serum C-peptide (0.93) and lowest for the GRS (0.66). Adding age at diagnosis and body mass index to C-peptide increased the AUC minimally (0.96). The GRS appears of modest diabetes diagnostic value in young Australians.
Collapse
Affiliation(s)
- Timothy M E Davis
- Medical School, University of Western Australia, Fremantle Hospital, Fremantle, Western Australia, Australia
- Department of Endocrinology and Diabetes, Fiona Stanley and Fremantle Hospitals Group, Perth, Western Australia, Australia
| | - Kirsten E Peters
- Medical School, University of Western Australia, Fremantle Hospital, Fremantle, Western Australia, Australia
- Proteomics International, Perth, Western Australia, Australia
| | - Wendy Davis
- Medical School, University of Western Australia, Fremantle Hospital, Fremantle, Western Australia, Australia
| |
Collapse
|
14
|
Liu W, Fang Y, Cai X, Zhu Y, Zhang M, Han X, Li J, Yin S, Cai D, Chen J, Wang L, Shi D, Ji L. Preserved C-peptide is common and associated with higher time in range in Chinese type 1 diabetes. Front Endocrinol (Lausanne) 2024; 15:1335913. [PMID: 38405156 PMCID: PMC10884320 DOI: 10.3389/fendo.2024.1335913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Objective The aim of this study is to determine the residual C-peptide level and to explore the clinical significance of preserved C-peptide secretion in glycemic control in Chinese individuals with type 1 diabetes (T1D). Research design and methods A total of 534 participants with T1D were enrolled and divided into two groups, low-C-peptide group (fasting C-peptide ≤10 pmol/L) and preserved-C-peptide group (fasting C-peptide >10 pmol/L), and clinical factors were compared between the two groups. In 174 participants who were followed, factors associated with C-peptide loss were also identified by Cox regression. In addition, glucose metrics derived from intermittently scanned continuous glucose monitoring were compared between individuals with low C-peptide and those with preserved C-peptide in 178 participants. Results The lack of preserved C-peptide was associated with longer diabetes duration, glutamic acid decarboxylase autoantibody, and higher daily insulin doses, after adjustment {OR, 1.10 [interquartile range (IQR), 1.06-1.14]; OR, 0.46 (IQR, 0.27-0.77); OR, 1.04 (IQR, 1.02-1.06)}. In the longitudinal analysis, the percentages of individuals with preserved C-peptide were 71.4%, 56.8%, 71.7%, 62.5%, and 22.2% over 5 years of follow-up. Preserved C-peptide was also associated with higher time in range after adjustment of diabetes duration [62.4 (IQR, 47.3-76.6) vs. 50.3 (IQR, 36.2-63.0) %, adjusted P = 0.003]. Conclusions Our results indicate that a high proportion of Chinese patients with T1D had preserved C-peptide secretion. Meanwhile, residual C-peptide was associated with favorable glycemic control, suggesting the importance of research on adjunctive therapy to maintain β-cell function in T1D.
Collapse
Affiliation(s)
- Wei Liu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Yayu Fang
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Yu Zhu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Mingxia Zhang
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Juan Li
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Sai Yin
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Deheng Cai
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Jing Chen
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Lei Wang
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Dawei Shi
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| |
Collapse
|
15
|
Kennedy EC, Hawkes CP. Approaches to Measuring Beta Cell Reserve and Defining Partial Clinical Remission in Paediatric Type 1 Diabetes. CHILDREN (BASEL, SWITZERLAND) 2024; 11:186. [PMID: 38397298 PMCID: PMC10887271 DOI: 10.3390/children11020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024]
Abstract
CONTEXT Type 1 diabetes (T1D) results from the autoimmune T-cell mediated destruction of pancreatic beta cells leading to insufficient insulin secretion. At the time of diagnosis of T1D, there is residual beta cell function that declines over the subsequent months to years. Recent interventions have been approved to preserve beta cell function in evolving T1D. OBJECTIVE The aim of this review is to summarise the approaches used to assess residual beta cell function in evolving T1D, and to highlight potential future directions. METHODS Studies including subjects aged 0 to 18 years were included in this review. The following search terms were used; "(type 1 diabetes) and (partial remission)" and "(type 1 diabetes) and (honeymoon)". References of included studies were reviewed to determine if additional relevant studies were eligible. RESULTS There are numerous approaches to quantifying beta cell reserve in evolving T1D. These include c-peptide measurement after a mixed meal or glucagon stimuli, fasting c-peptide, the urinary c-peptide/creatinine ratio, insulin dose-adjusted haemoglobin A1c, and other clinical models to estimate beta cell function. Other biomarkers may have a role, including the proinsulin/c-peptide ratio, cytokines, and microRNA. Studies using thresholds to determine if residual beta cell function is present often differ in values used to define remission. CONCLUSIONS As interventions are approved to preserve beta cell function, it will become increasingly necessary to quantify residual beta cell function in research and clinical contexts. In this report, we have highlighted the strengths and limitations of the current approaches.
Collapse
Affiliation(s)
- Elaine C Kennedy
- Department of Paediatrics and Child Health, University College Cork, T12 DC4A Cork, Ireland
- INFANT Research Centre, University College Cork, T12 DC4A Cork, Ireland
| | - Colin P Hawkes
- Department of Paediatrics and Child Health, University College Cork, T12 DC4A Cork, Ireland
- INFANT Research Centre, University College Cork, T12 DC4A Cork, Ireland
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Patil RS, Tupe RS. Communal interaction of glycation and gut microbes in diabetes mellitus, Alzheimer's disease, and Parkinson's disease pathogenesis. Med Res Rev 2024; 44:365-405. [PMID: 37589449 DOI: 10.1002/med.21987] [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: 09/23/2022] [Revised: 07/12/2023] [Accepted: 08/06/2023] [Indexed: 08/18/2023]
Abstract
Diabetes and its complications, Alzheimer's disease (AD), and Parkinson's disease (PD) are increasing gradually, reflecting a global threat vis-à-vis expressing the essentiality of a substantial paradigm shift in research and remedial actions. Protein glycation is influenced by several factors, like time, temperature, pH, metal ions, and the half-life of the protein. Surprisingly, most proteins associated with metabolic and neurodegenerative disorders are generally long-lived and hence susceptible to glycation. Remarkably, proteins linked with diabetes, AD, and PD share this characteristic. This modulates protein's structure, aggregation tendency, and toxicity, highlighting renovated attention. Gut microbes and microbial metabolites marked their importance in human health and diseases. Though many scientific shreds of evidence are proposed for possible change and dysbiosis in gut flora in these diseases, very little is known about the mechanisms. Screening and unfolding their functionality in metabolic and neurodegenerative disorders is essential in hunting the gut treasure. Therefore, it is imperative to evaluate the role of glycation as a common link in diabetes and neurodegenerative diseases, which helps to clarify if modulation of nonenzymatic glycation may act as a beneficial therapeutic strategy and gut microbes/metabolites may answer some of the crucial questions. This review briefly emphasizes the common functional attributes of glycation and gut microbes, the possible linkages, and discusses current treatment options and therapeutic challenges.
Collapse
Affiliation(s)
- Rahul Shivaji Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Rashmi Santosh Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Pune, Maharashtra, India
| |
Collapse
|
17
|
Leete P. Type 1 diabetes in the pancreas: A histological perspective. Diabet Med 2023; 40:e15228. [PMID: 37735524 DOI: 10.1111/dme.15228] [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] [Received: 08/14/2023] [Revised: 09/15/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
AIMS This review aims to introduce research in the pancreas to a broader audience. The pancreas is a heterocrine gland residing deep within our abdominal cavity. It is the home to our islets, which play a pivotal role in regulating metabolic homeostasis. Due to its structure and location, it is an impossible organ to study, in molecular detail, in living humans, and yet, understanding the pancreas is critical if we aim to characterise the immunopathology of type 1 diabetes (T1D) and one day prevent the triggering of the autoimmune attack associated with ß-cell demise. METHODS Over a 100 years ago, we began studying pancreatic histology using cadaveric samples and clever adaptations to microscopes. As histologists, some may say nothing much has changed. Nevertheless, our microscopes can now interrogate multiple proteins at molecular resolution. Images of pancreas sections are no longer constrained to a single field of view and can capture a thousands and thousands of cells. AI-image-analysis packages can analyse these massive data sets offering breakthrough findings. CONCLUSION This narrative review will provide an overview of pancreatic anatomy, and the importance of research focused on the pancreas in T1D. It will range from histological breakthroughs to briefly discussing the challenges associated with characterising the organ. I shall briefly introduce a selection of the available global biobanks and touch on the distinct pancreatic endotypes that differ immunologically and in ß-cell behaviour. Finally, I will introduce the idea of developing a collaborative tool aimed at developing a cohesive framework for characterising heterogeneity and stratifying endotypes in T1D more readily.
Collapse
Affiliation(s)
- Pia Leete
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
18
|
James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023; 78:101809. [PMID: 37734713 PMCID: PMC10622886 DOI: 10.1016/j.molmet.2023.101809] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND T1D is an autoimmune disease in which pancreatic islets of Langerhans are infiltrated by immune cells resulting in the specific destruction of insulin-producing islet beta cells. Our understanding of the factors leading to islet infiltration and the interplay of the immune cells with target beta cells is incomplete, especially in human disease. While murine models of T1D have provided crucial information for both beta cell and autoimmune cell function, the translation of successful therapies in the murine model to human disease has been a challenge. SCOPE OF REVIEW Here, we discuss current state of the art and consider knowledge gaps concerning the interface of the islet beta cell with immune infiltrates, with a focus on T cells. We discuss pancreatic and immune cell phenotypes and their impact on cell function in health and disease, which we deem important to investigate further to attain a more comprehensive understanding of human T1D disease etiology. MAJOR CONCLUSIONS The last years have seen accelerated development of approaches that allow comprehensive study of human T1D. Critically, recent studies have contributed to our revised understanding that the pancreatic beta cell assumes an active role, rather than a passive position, during autoimmune disease progression. The T cell-beta cell interface is a critical axis that dictates beta cell fate and shapes autoimmune responses. This includes the state of the beta cell after processing internal and external cues (e.g., stress, inflammation, genetic risk) that that contributes to the breaking of tolerance by hyperexpression of human leukocyte antigen (HLA) class I with presentation of native and neoepitopes and secretion of chemotactic factors to attract immune cells. We anticipate that emerging insights about the molecular and cellular aspects of disease initiation and progression processes will catalyze the development of novel and innovative intervention points to provide additional therapies to individuals affected by T1D.
Collapse
Affiliation(s)
- Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Alok V Joglekar
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amelia K Linnemann
- Center for Diabetes and Metabolic Diseases, and Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sally C Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
19
|
Minasian V, Nazari M. The association between type 1 diabetes and exercise/physical activity and prolongation of the honeymoon phase in patients. Life Sci 2023; 332:122114. [PMID: 37739162 DOI: 10.1016/j.lfs.2023.122114] [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: 08/06/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
In type 1 diabetes (T1D), pancreatic beta cells are destroyed by the immune system, causing chronic hyperglycemia and micro and macrovascular complications. However, some people experience a 'honeymoon' phase (or partial remission) after being diagnosed with type 1 diabetes. During this phase, a substantial amount of insulin is still produced by the pancreas, helping to reduce blood sugar levels and the requirement for external insulin. The clinical significance of this phase lies in the potential for pharmacological and non-pharmacological interventions during this time frame to either slow down or arrest beta-cell destruction. Clearly, we need to continue researching novel therapies like immunomodulatory agents, but we also need to look at potentially effective therapies with acceptable side effects that can serve as a complement to the medicines currently being studied. Physical activity and exercise, regardless of its type, is one of the factors its impact on the control of diabetes is being investigated and promising results have been achieved. Although there are still limited reports in this regard, there is some evidence to suggest that regular physical exercise could prolong the honeymoon period in both adults and children. In this review, having described the immune base of type 1 diabetes, we outline the benefits of exercise on the general health of individuals with T1D. Moreover, we centered on the honeymoon and current evidence suggesting the effects of physical activity and exercise on this phase duration.
Collapse
Affiliation(s)
- Vazgen Minasian
- Faculty of Sport Sciences, Department of Exercise physiology, University of Isfahan, Isfahan, Iran.
| | - Maryam Nazari
- Faculty of Sport Sciences, Department of Exercise physiology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
20
|
Cristelo C, Nunes R, Pinto S, Marques JM, Gama FM, Sarmento B. Targeting β Cells with Cathelicidin Nanomedicines Improves Insulin Function and Pancreas Regeneration in Type 1 Diabetic Rats. ACS Pharmacol Transl Sci 2023; 6:1544-1560. [PMID: 37854630 PMCID: PMC10580391 DOI: 10.1021/acsptsci.3c00218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Indexed: 10/20/2023]
Abstract
Type 1 diabetes (T1D) is an incurable condition with an increasing incidence worldwide, in which the hallmark is the autoimmune destruction of pancreatic insulin-producing β cells. Cathelicidin-based peptides have been shown to improve β cell function and neogenesis and may thus be relevant while developing T1D therapeutics. In this work, a cathelicidin-derived peptide, LLKKK18, was loaded in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), surface-functionalized with exenatide toward a GLP-1 receptor, aiming the β cell-targeted delivery of the peptide. The NPs present a mean size of around 100 nm and showed long-term stability, narrow size distribution, and negative ζ-potential (-10 mV). The LLKKK18 association efficiency and loading were 62 and 2.9%, respectively, presenting slow and sustained in vitro release under simulated physiologic fluids. Glucose-stimulated insulin release in the INS-1E cell line was observed in the presence of the peptide. In addition, NPs showed a strong association with β cells from isolated rat islets. After administration to diabetic rats, NPs induced a significant reduction of the hyperglycemic state, an improvement in the pancreatic insulin content, and glucose tolerance. Also remarkable, a considerable increase in the β cell mass in the pancreas was observed. Overall, this novel and versatile nanomedicine showed glucoregulatory ability and can pave the way for the development of a new generation of therapeutic approaches for T1D treatment.
Collapse
Affiliation(s)
- Cecília Cristelo
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- Centro
de Engenharia Biológica, Universidade
do Minho, Campus de Gualtar, Braga 4710-057, Portugal
- ICBAS
− Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto 4050-313, Portugal
| | - Rute Nunes
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- IUCS-CESPU, Instituto
Universitário de Ciências
da Saúde, Gandra 4585-116, Portugal
| | - Soraia Pinto
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- ICBAS
− Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto 4050-313, Portugal
| | - Joana Moreira Marques
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- Faculdade
de Farmácia, Universidade do Porto, Porto 4099-002, Portugal
| | - Francisco Miguel Gama
- Centro
de Engenharia Biológica, Universidade
do Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Bruno Sarmento
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- IUCS-CESPU, Instituto
Universitário de Ciências
da Saúde, Gandra 4585-116, Portugal
| |
Collapse
|
21
|
Murakami T, Nakamura T, Fujimoto H, Fujikura J, Shimizu Y, Miyake KK, Otani D, Sakaki K, Kiyobayashi S, Anazawa T, Nakamoto Y, Inagaki N. Noninvasive evaluation of donor and native pancreases following simultaneous pancreas-kidney transplantation using positron emission tomography/computed tomography. J Diabetes Investig 2023; 14:1187-1191. [PMID: 37377043 PMCID: PMC10512903 DOI: 10.1111/jdi.14045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
It is crucial to develop practical and noninvasive methods to assess the functional beta-cell mass in a donor pancreas, in which monitoring and precise evaluation is challenging. A patient with type 1 diabetes underwent noninvasive imaging following simultaneous kidney-pancreas transplantation with positron emission tomography/computed tomography (PET/CT) using an exendin-based probe, [18 F]FB(ePEG12)12-exendin-4. Following transplantation, PET imaging with [18 F]FB(ePEG12)12-exendin-4 revealed simultaneous and distinct accumulations in the donor and native pancreases. The pancreases were outlined at a reasonable distance from the surrounding organs using [18 F]FB(ePEG12)12-exendin-4 whole-body maximum intensity projection and axial PET images. At 1 and 2 h after [18 F]FB(ePEG12)12-exendin-4 administration, the mean standardized uptake values were 2.96 and 3.08, respectively, in the donor pancreas and 1.97 and 2.25, respectively, in the native pancreas. [18 F]FB(ePEG12)12-exendin-4 positron emission tomography imaging allowed repeatable and quantitative assessment of beta-cell mass following simultaneous kidney-pancreas transplantation.
Collapse
Affiliation(s)
- Takaaki Murakami
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Toshihiro Nakamura
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroyuki Fujimoto
- Radioisotope Research Center, Agency for Health, Safety and EnvironmentKyoto UniversityKyotoJapan
| | - Junji Fujikura
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yoichi Shimizu
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kanae K. Miyake
- Department of Advanced Medical Imaging Research, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Daisuke Otani
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kentaro Sakaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Sakura Kiyobayashi
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Takayuki Anazawa
- Division of Hepato‐Biliary‐Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of MedicineKyoto UniversityKyotoJapan
- Medical Research Institute Kitano Hospital, PIIF Tazuke‐kofukaiOsakaJapan
| |
Collapse
|
22
|
Krawczyk M, Burzynska-Pedziwiatr I, Wozniak LA, Bukowiecka-Matusiak M. Impact of Polyphenols on Inflammatory and Oxidative Stress Factors in Diabetes Mellitus: Nutritional Antioxidants and Their Application in Improving Antidiabetic Therapy. Biomolecules 2023; 13:1402. [PMID: 37759802 PMCID: PMC10526737 DOI: 10.3390/biom13091402] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by hyperglycaemia and oxidative stress. Oxidative stress plays a crucial role in the development and progression of diabetes and its complications. Nutritional antioxidants derived from dietary sources have gained significant attention due to their potential to improve antidiabetic therapy. This review will delve into the world of polyphenols, investigating their origins in plants, metabolism in the human body, and relevance to the antioxidant mechanism in the context of improving antidiabetic therapy by attenuating oxidative stress, improving insulin sensitivity, and preserving β-cell function. The potential mechanisms of, clinical evidence for, and future perspectives on nutritional antioxidants as adjuvant therapy in diabetes management are discussed.
Collapse
|
23
|
Sokołowska-Gadoux M, Jarosz-Chobot P, Polanska J, Kalemba A, Chobot A. Body mass index and partial remission in 119 children with type 1 diabetes-a 6-year observational study. Front Endocrinol (Lausanne) 2023; 14:1257758. [PMID: 37780631 PMCID: PMC10538636 DOI: 10.3389/fendo.2023.1257758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Background/objective This long-term study aimed to analyze the associations between BMI Z-score, HbA1c, and daily insulin requirement (DIR) and the prevalence and duration of partial remission (PR) in children and adolescents with type 1 diabetes (T1D). Methods After retrieving retrospective data for 195 patients from their health records at 24, 48, and 72 months after T1D diagnosis, the study group was comprised of 119 (57 girls) children with a complete dataset for all 6 years. PR was defined according to the ISPAD guidelines. Analyses were carried out in the whole group and subgroups according to PR duration: no PR at all (NPR), PR lasting less than 2 years (PR < 2), and PR at least 2 years (PR ≥ 2). Results PR was observed in 63% of the patients (78.9% of overweight and 100% of obese patients). NPR patients showed the lowest mean initial BMI Z-score [-0.65 ± 1.29 vs. 0.02 ± 1.42, (PR < 2), p = 0.01 and vs. 0.64 ± 1.43 (PR ≥ 2), p = 0.17]. The dissimilarity in BMI across patients declined over time. Within the NPR group, the initial mean BMI Z-score significantly increased within the first 2 years (unadjusted p < 0.001) and remained constant afterward. In the PR <2 group, the highest increase in BMI Z-score occurred after 4 years (p < 0.001) and then decreased (p = 0.04). In the PR ≥2, the BMI Z-score slightly decreased within the first 2 years (p = 0.02), then increased (p = 0.03) and remained unchanged for the last 2 years. Six years after T1D started, the mean DIRs do not differ among the patient groups (ANOVA p = 0.272). Conclusion During 6 years of follow-up, PR occurred in almost two-thirds of the studied children including almost all overweight and obese children. We observed a gradual normalization of the BMI Z-score at the end of the follow-up. BMI Z-score increased slightly in children with no remission initially but remained later constant until the end of observation. In both remitter groups, the increase in BMI Z-score appeared later when the protective honeymoon period ended. Regardless of BMI Z-score, the β-cell destruction process progresses, and after 6 years, the DIR is similar for all patients.
Collapse
Affiliation(s)
- Magdalena Sokołowska-Gadoux
- Department of Children’s Diabetology and Pediatrics, John Paul II Upper Silesian Child Health Centre, Katowice, Poland
| | - Przemysława Jarosz-Chobot
- Department of Children’s Diabetology and Pediatrics, Medical University of Silesia, Katowice, Poland
| | - Joanna Polanska
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Alicja Kalemba
- Department of Children’s Diabetology and Pediatrics, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agata Chobot
- Department of Pediatrics, Institute of Medical Sciences, Opole University, Opole, Poland
| |
Collapse
|
24
|
Abstract
Despite major advances over the past decade, prevention and treatment of type 1 diabetes mellitus (T1DM) remain suboptimal, with large and unexplained variations in individual responses to interventions. The current classification schema for diabetes mellitus does not capture the complexity of this disease or guide clinical management effectively. One of the approaches to achieve the goal of applying precision medicine in diabetes mellitus is to identify endotypes (that is, well-defined subtypes) of the disease each of which has a distinct aetiopathogenesis that might be amenable to specific interventions. Here, we describe epidemiological, clinical, genetic, immunological, histological and metabolic differences within T1DM that, together, suggest heterogeneity in its aetiology and pathogenesis. We then present the emerging endotypes and their impact on T1DM prediction, prevention and treatment.
Collapse
Affiliation(s)
- Maria J Redondo
- Paediatric Diabetes & Endocrinology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| | - Noel G Morgan
- Exeter Centre of Excellence for Diabetes Research (EXCEED), Department of Clinical and Biomedical and Science, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
25
|
Herat LY, Matthews JR, Hibbs M, Rakoczy EP, Schlaich MP, Matthews VB. SGLT1/2 inhibition improves glycemic control and multi-organ protection in type 1 diabetes. iScience 2023; 26:107260. [PMID: 37520739 PMCID: PMC10384225 DOI: 10.1016/j.isci.2023.107260] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/26/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Sodium glucose cotransporters (SGLTs) are transport proteins that are expressed throughout the body. Inhibition of SGLTs is a relatively novel therapeutic strategy to improve glycemic control and has been shown to promote cardiorenal benefits. Dual SGLT1/2 inhibitors (SGLT1/2i) such as sotagliflozin target both SGLT1 and 2 proteins. Sotagliflozin or vehicle was administered to diabetic Akimba mice for 8 weeks at a dose of 25 mg/kg/day. Urine glucose levels, water consumption, and body weight were measured weekly. Serum, kidney, pancreas, and brain tissue were harvested under terminal anesthesia. Tissues were assessed using immunohistochemistry or ELISA techniques. Treatment with sotagliflozin promoted multiple metabolic benefits in diabetic Akimba mice resulting in decreased blood glucose and improved polydipsia. Sotagliflozin also prevented mortalities associated with diabetes. Our data suggests that there is the possibility that combined SGLT1/2i may be superior to SGLT2i in controlling glucose homeostasis and provides protection of multiple organs affected by diabetes.
Collapse
Affiliation(s)
- Lakshini Yasaswi Herat
- Dobney Hypertension Centre, School of Biomedical Sciences – Royal Perth Hospital Unit / Royal Perth Hospital Medical Research Foundation, University of Western Australia, Crawley, WA 6009, Australia
| | - Jennifer Rose Matthews
- Dobney Hypertension Centre, School of Biomedical Sciences – Royal Perth Hospital Unit / Royal Perth Hospital Medical Research Foundation, University of Western Australia, Crawley, WA 6009, Australia
| | - Moira Hibbs
- Research Centre, Royal Perth Hospital, Perth, WA 6000, Australia
| | | | - Markus Peter Schlaich
- Dobney Hypertension Centre, Medical School – Royal Perth Hospital Unit / Royal Perth Hospital Medical Research Foundation, University of Western Australia, Crawley, WA 6009, Australia
- Department of Cardiology and Department of Nephrology, Royal Perth Hospital, Perth, WA 6000, Australia
| | - Vance Bruce Matthews
- Dobney Hypertension Centre, School of Biomedical Sciences – Royal Perth Hospital Unit / Royal Perth Hospital Medical Research Foundation, University of Western Australia, Crawley, WA 6009, Australia
| |
Collapse
|
26
|
Kattner N. Immune cell infiltration in the pancreas of type 1, type 2 and type 3c diabetes. Ther Adv Endocrinol Metab 2023; 14:20420188231185958. [PMID: 37529508 PMCID: PMC10387691 DOI: 10.1177/20420188231185958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/16/2023] [Indexed: 08/03/2023] Open
Abstract
The different types of diabetes differ in disease pathogenesis but share the impairment or loss of β-cell function leading to chronic hyperglycaemia. While immune cells are present throughout the whole pancreas in normality, their number and activation is increased in diabetes. Different patterns and composition of inflammation could be observed in type 1, type 2 and type 3c diabetes. Immune cells, pancreatic stellate cells and fibrosis were present in the islet microenvironment and could add to β-cell dysfunction and therefore development and progression of diabetes. First studies investigating the use of anti-inflammatory drugs demonstrate their ability to rescue remaining β-cell function and their potential benefit in diabetes treatment. This article provides an overview of immune cell infiltrates in different types of diabetes, highlights the knowledge of their impact on β-cell function and introduces the potential of immunomodulatory strategies.
Collapse
Affiliation(s)
- Nicole Kattner
- Translational and Clinical Research Institute, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Harsunen M, Haukka J, Harjutsalo V, Mars N, Syreeni A, Härkönen T, Käräjämäki A, Ilonen J, Knip M, Sandholm N, Miettinen PJ, Groop PH, Tuomi T. Residual insulin secretion in individuals with type 1 diabetes in Finland: longitudinal and cross-sectional analyses. Lancet Diabetes Endocrinol 2023; 11:465-473. [PMID: 37290465 DOI: 10.1016/s2213-8587(23)00123-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Contrary to the presumption that type 1 diabetes leads to an absolute insulin deficiency, many individuals with type 1 diabetes have circulating C-peptide years after the diagnosis. We studied factors affecting random serum C-peptide concentration in individuals with type 1 diabetes and the association with diabetic complications. METHODS Our longitudinal analysis included individuals newly diagnosed with type 1 diabetes from Helsinki University Hospital (Helsinki, Finland) with repeated random serum C-peptide and concomitant glucose measurements from within 3 months of diagnosis and at least once later. The long-term cross-sectional analysis included data from participants from 57 centres in Finland who had type 1 diabetes diagnosed after 5 years of age, initiation of insulin treatment within 1 year from diagnosis, and a C-peptide concentration of less than 1·0 nmol/L (FinnDiane study) and patients with type 1 diabetes from the DIREVA study. We tested the association of random serum C-peptide concentrations and polygenic risk scores with one-way ANOVA, and association of random serum C-peptide concentrations, polygenic risk scores, and clinical factors with logistic regression. FINDINGS The longitudinal analysis included 847 participants younger than 16 years and 110 aged 16 years or older. In the longitudinal analysis, age at diagnosis strongly correlated with the decline in C-peptide secretion. The cross-sectional analysis included 3984 participants from FinnDiane and 645 from DIREVA. In the cross-sectional analysis, at a median duration of 21·6 years (IQR 12·5-31·2), 776 (19·4%) of 3984 FinnDiane participants had residual random serum C-peptide secretion (>0·02 nmol/L), which was associated with lower type 1 diabetes polygenic risk compared with participants without random serum C-peptide (p<0·0001). Random serum C-peptide was inversely associated with hypertension, HbA1c, and cholesterol, but also independently with microvascular complications (adjusted OR 0·61 [95% CI 0·38-0·96], p=0·033, for nephropathy; 0·55 [0·34-0·89], p=0·014, for retinopathy). INTERPRETATION Although children with multiple autoantibodies and HLA risk genotypes progressed to absolute insulin deficiency rapidly, many adolescents and adults had residual random serum C-peptide decades after the diagnosis. Polygenic risk of type 1 and type 2 diabetes affected residual random serum C-peptide. Even low residual random serum C-peptide concentrations seemed to be associated with a beneficial complications profile. FUNDING Folkhälsan Research Foundation; Academy of Finland; University of Helsinki and Helsinki University Hospital; Medical Society of Finland; the Sigrid Juselius Foundation; the "Liv and Hälsa" Society; Novo Nordisk Foundation; and State Research Funding via the Helsinki University Hospital, the Vasa Hospital District, Turku University Hospital, Vasa Central Hospital, Jakobstadsnejdens Heart Foundation, and the Medical Foundation of Vaasa.
Collapse
Affiliation(s)
- Minna Harsunen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jani Haukka
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Nina Mars
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anna Syreeni
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Taina Härkönen
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Annemari Käräjämäki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Diabetes unit of Ostrobothnia, Wellbeing Services County of Ostrobothnia, Vaasa, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Johanna Miettinen
- Translational Stem Cell Biology and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Diabetes, Central Medical School, Monash University, Melbourne, VIC, Australia
| | - Tiinamaija Tuomi
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Abdominal Center, Endocrinology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Lund, Sweden.
| |
Collapse
|
28
|
Dirr EW, Patel Y, Johnson RD, Otto KJ. The effects of targeted vagus nerve stimulation on glucose homeostasis in STZ-induced diabetic rodents. Front Neurosci 2023; 17:1179276. [PMID: 37397461 PMCID: PMC10309008 DOI: 10.3389/fnins.2023.1179276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
During type 1 diabetes, an autoimmune attack destroys pancreatic β-cells leading to the inability to maintain glucose homeostasis. These β-cells are neuroresponsive endocrine cells which normally secrete insulin partially in response to input from the vagus nerve. This neural pathway can be utilized as a point of therapeutic intervention by delivering exogenous stimulation to drive increased insulin secretion. In this study, a cuff electrode was implanted on the pancreatic branch of the vagus nerve just prior to pancreatic insertion in rats, and a continuous glucose meter was implanted into the descending aorta. Streptozotocin (STZ) was used to induce a diabetic state, and changes in blood glucose were assessed using various stimulation parameters. Stimulation driven changes in hormone secretion, pancreatic blood flow, and islet cell populations were assessed. We found increased changes in the rate of blood glucose change during stimulation which subsided after stimulation ended paired with increased concentration of circulating insulin. We did not observe increased pancreatic perfusion, which suggests that the modulation of blood glucose was due to the activation of b-cells rather than changes in the extra-organ transport of insulin. Pancreatic neuromodulation showed potentially protective effects by reducing deficits in islet diameter, and ameliorating insulin loss after STZ treatment.
Collapse
Affiliation(s)
- Elliott W. Dirr
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Yogi Patel
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Richard D. Johnson
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Kevin J. Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Department of Neurology, University of Florida, Gainesville, FL, United States
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, United States
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, United States
| |
Collapse
|
29
|
Perry DJ, Shapiro MR, Chamberlain SW, Kusmartseva I, Chamala S, Balzano-Nogueira L, Yang M, Brant JO, Brusko M, Williams MD, McGrail KM, McNichols J, Peters LD, Posgai AL, Kaddis JS, Mathews CE, Wasserfall CH, Webb-Robertson BJM, Campbell-Thompson M, Schatz D, Evans-Molina C, Pugliese A, Concannon P, Anderson MS, German MS, Chamberlain CE, Atkinson MA, Brusko TM. A genomic data archive from the Network for Pancreatic Organ donors with Diabetes. Sci Data 2023; 10:323. [PMID: 37237059 PMCID: PMC10219990 DOI: 10.1038/s41597-023-02244-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The Network for Pancreatic Organ donors with Diabetes (nPOD) is the largest biorepository of human pancreata and associated immune organs from donors with type 1 diabetes (T1D), maturity-onset diabetes of the young (MODY), cystic fibrosis-related diabetes (CFRD), type 2 diabetes (T2D), gestational diabetes, islet autoantibody positivity (AAb+), and without diabetes. nPOD recovers, processes, analyzes, and distributes high-quality biospecimens, collected using optimized standard operating procedures, and associated de-identified data/metadata to researchers around the world. Herein describes the release of high-parameter genotyping data from this collection. 372 donors were genotyped using a custom precision medicine single nucleotide polymorphism (SNP) microarray. Data were technically validated using published algorithms to evaluate donor relatedness, ancestry, imputed HLA, and T1D genetic risk score. Additionally, 207 donors were assessed for rare known and novel coding region variants via whole exome sequencing (WES). These data are publicly-available to enable genotype-specific sample requests and the study of novel genotype:phenotype associations, aiding in the mission of nPOD to enhance understanding of diabetes pathogenesis to promote the development of novel therapies.
Collapse
Affiliation(s)
- Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Sonya W Chamberlain
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Irina Kusmartseva
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Srikar Chamala
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Leandro Balzano-Nogueira
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Mingder Yang
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Jason O Brant
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA
| | - Maigan Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - MacKenzie D Williams
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Kieran M McGrail
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - James McNichols
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Leeana D Peters
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - John S Kaddis
- Department of Diabetes and Cancer Discovery Science, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Bobbie-Jo M Webb-Robertson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Desmond Schatz
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases and the Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alberto Pugliese
- Diabetes Research Institute, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33021, USA
| | - Patrick Concannon
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Genetics Institute, University of Florida, Gainesville, FL, 32601, USA
| | - Mark S Anderson
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Michael S German
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Chester E Chamberlain
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
30
|
Pieper T, Roth KDR, Glaser V, Riet T, Buitrago-Molina LE, Hagedorn M, Lieber M, Hust M, Noyan F, Jaeckel E, Hardtke-Wolenski M. Generation of Chimeric Antigen Receptors against Tetraspanin 7. Cells 2023; 12:1453. [PMID: 37296574 PMCID: PMC10252682 DOI: 10.3390/cells12111453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Adoptive transfer of antigen-specific regulatory T cells (Tregs) has shown promising results in the treatment of autoimmune diseases; however, the use of polyspecific Tregs has limited effects. However, obtaining a sufficient number of antigen-specific Tregs from patients with autoimmune disorders remains challenging. Chimeric antigen receptors (CARs) provide an alternative source of T cells for novel immunotherapies that redirect T cells independently of the MHC. In this study, we aimed to generate antibody-like single-chain variable fragments (scFv) and subsequent CARs against tetraspanin 7 (TSPAN7), a membrane protein highly expressed on the surface of pancreatic beta cells, using phage display technology. We established two methods for generating scFvs against TSPAN7 and other target structures. Moreover, we established novel assays to analyze and quantify their binding abilities. The resulting CARs were functional and activated specifically by the target structure, but could not recognize TSPAN7 on the surface of beta cells. Despite this, this study demonstrates that CAR technology is a powerful tool for generating antigen-specific T cells and provides new approaches for generating functional CARs.
Collapse
Affiliation(s)
- Tom Pieper
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Kristian Daniel Ralph Roth
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Medizinische Biotechnologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Viktor Glaser
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Riet
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department I of Internal Medicine, Tumor Genetics, University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50933 Cologne, Germany
| | - Laura Elisa Buitrago-Molina
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Maike Hagedorn
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Maren Lieber
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Medizinische Biotechnologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department of Liver Transplantation, Multi Organ Transplant Program, University Health Network, University of Toronto, Toronto, ON M5T 0S8, Canada
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 47057 Essen, Germany
| |
Collapse
|
31
|
Gabbay MAL, Crispim F, Dib SA. Residual β-cell function in Brazilian Type 1 diabetes after 3 years of diagnosis: prevalence and association with low presence of nephropathy. Diabetol Metab Syndr 2023; 15:51. [PMID: 36935525 PMCID: PMC10026390 DOI: 10.1186/s13098-023-01014-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/04/2023] [Indexed: 03/21/2023] Open
Abstract
BACKGROUND Persistence of β cell-function in Type 1 diabetes (T1D) is associated with glycaemia stability and lower prevalence of microvascular complications. We aimed to assess the prevalence of residual C- peptide secretion in long-term Brazilian childhood onset T1D receiving usual diabetes care and its association to clinical, metabolic variables and microvascular complications. METHODS A cross-sectional observational study with 138 T1D adults with ≥ 3 years from the diagnosis by routine diabetes care. Clinical, metabolic variables and microvascular complications were compared between positive ultra-sensitive fasting serum C-peptide (FCP +) and negative (FCP-) participants. RESULTS T1D studied had ≥ 3 yrs. of diagnosis and 60% had FCP > 1.15 pmol/L. FCP + T1D were older at diagnosis (10 vs 8 y.o; p = 0.03) and had less duration of diabetes (11 vs 15 y.o; p = 0.002). There was no association between the FCP + and other clinical and metabolic variable but there was inversely association with microalbuminuria (28.6% vs 13.4%, p = 0.03), regardless of HbA1c. FCP > 47 pmol/L were associated with nephropathy protection but were not related to others microvascular complications. CONCLUSION Residual insulin secretion is present in 60% of T1D with ≥ 3 years of diagnosis in routine diabetes care. FCP + was positively associated with age of diagnosis and negatively with duration of disease and microalbuminuria, regardless of HbA1c.
Collapse
Affiliation(s)
- Monica A L Gabbay
- Centre for Diabetes, Endocrinology Division, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.
- Molecular Biology Laboratory, Endocrinology Division, Department of Medicine Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.
| | - Felipe Crispim
- Molecular Biology Laboratory, Endocrinology Division, Department of Medicine Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sergio A Dib
- Centre for Diabetes, Endocrinology Division, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Thivolet C, Larger E, Cariou B, Renard E, Hanaire H, Benhamou PY, Guerci B, Mathiotte É, Chikh K. Dulaglutide and insulin microsecretion in people with type 1 diabetes (DIAMOND-GLP-1): A randomized double-blind placebo-controlled trial. DIABETES & METABOLISM 2023; 49:101433. [PMID: 36781064 DOI: 10.1016/j.diabet.2023.101433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/13/2023]
Affiliation(s)
- Charles Thivolet
- Centre du diabète DIAB-eCARE, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon France.
| | - Etienne Larger
- Department of Diabetology, Cochin Hospital AP-HP, University of Paris, Paris, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes F-44000, France
| | - Eric Renard
- Department of Endocrinology, Diabetes and Nutrition, Montpellier University Hospital, University of Montpellier, Montpellier, France
| | - Hélène Hanaire
- Department of Diabetology, Metabolic diseases and Nutrition, CHU Toulouse, University of Toulouse, Toulouse, France
| | - Pierre-Yves Benhamou
- Department of Endocrinology, Grenoble University Hospital, Grenoble Alpes University, INSERM U1055, LBFA, Grenoble, France
| | - Bruno Guerci
- Department of Endocrinology, Diabetology and Nutrition, CHRU of Nancy, Brabois Hospital, and ILCV Lorraine University, Vandoeuvre-les-Nancy, France
| | - Émilie Mathiotte
- Department of Clinical Research, pole IMER Hospices Civils de Lyon, Lyon, France
| | - Karim Chikh
- Department of biochemistry and molecular biology, Lyon-Sud Hospital, Hospices Civils de Lyon, Lyon, France, Université Claude-Bernard Lyon 1, Oullins, France
| |
Collapse
|
33
|
Irilouzadian R, Afaghi S, Esmaeili Tarki F, Rahimi F, Malekpour Alamadari N. Urinary c-peptide creatinine ratio (UCPCR) as a predictor of coronary artery disease in type 1 diabetes mellitus. Endocrinol Diabetes Metab 2023; 6:e413. [PMID: 36808709 PMCID: PMC10164436 DOI: 10.1002/edm2.413] [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: 12/14/2022] [Revised: 01/28/2023] [Accepted: 02/05/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Elevated C-peptide has been suggested as a risk factor for coronary artery disease (CAD). Elevated urinary C-peptide to creatinine ratio (UCPCR) as an alternative measurement is shown to be related to insulin secretion dysfunction; however, data regarding UCPCR predictive value for CAD in diabetes mellitus (DM) are scarce. Therefore, we aimed to assess the UCPCR association with CAD in type 1 DM (T1DM) patients. METHODS 279 patients previously diagnosed with T1DM included and categorized into two groups of CAD (n = 84) and without-CAD (n = 195). Furthermore, each group was divided into obese (body mass index (BMI) ≥ 30) and non-obese (BMI < 30) groups. Four models utilizing the binary logistic regression were designed to evaluate the role of UCPCR in CAD adjusted for well-known risk factors and mediators. RESULTS Median level of UCPCR was higher in CAD group compared to non-CAD group (0.07 vs. 0.04, respectively). Also, the well-acknowledged risk factors including being active smoker, hypertension, duration of diabetes, and body mass index (BMI) as well as higher levels of haemoglobin A1C (HbA1C), total cholesterol (TC), low-density lipoprotein (LDL) and estimated glomeruli filtration rate (e-GFR) had more significant pervasiveness in CAD patients. Based on multiple adjustments by logistic regression, UCPCR was a strong risk factor of CAD among T1DM patients independent of hypertension, demographic variables (gender, age, smoking, alcohol consumption), diabetes-related factors (diabetes duration, FBS, HbA1C), lipid profile (TC, LDL, HDL, TG) and renal-related indicators (creatinine, e-GFR, albuminuria, uric acid) in both patients with BMI≥30 and BMI < 30. CONCLUSION UCPCR is associated with clinical CAD, independent of CAD classic risk factors, glycaemic control, insulin resistance and BMI in type 1 DM patients.
Collapse
Affiliation(s)
- Rana Irilouzadian
- Burn Research Center, Iran university of medical sciences, Tehran, Iran
| | - Siamak Afaghi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Esmaeili Tarki
- Research institute of internal medicine, Shahid Modarres hospital, Shahid Beheshti university of medical sciences, Tehran, Iran
| | - Fatemehsadat Rahimi
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Malekpour Alamadari
- Department of Surgery, Clinical Research and Development Center, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Jansen TJP, Brom M, Boss M, Buitinga M, Tack CJ, van Meijel LA, de Galan BE, Gotthardt M. Importance of beta cell mass for glycaemic control in people with type 1 diabetes. Diabetologia 2023; 66:367-375. [PMID: 36394644 PMCID: PMC9669532 DOI: 10.1007/s00125-022-05830-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022]
Abstract
AIMS/HYPOTHESIS The role of beta cell mass in the balance of glucose control and hypoglycaemic burden in people with type 1 diabetes is unclear. We applied positron emission tomography (PET) imaging with radiolabelled exendin to compare beta cell mass among people with type 1 diabetes and either low glucose variability (LGV) or high glucose variability (HGV). METHODS All participants with either LGV (n=9) or HGV (n=7) underwent a mixed-meal tolerance test to determine beta cell function and wore a blinded continuous glucose monitor for a week. After an i.v. injection with [68Ga]Ga-NODAGA-exendin-4, PET images were acquired for the quantification of pancreatic uptake of radiolabelled exendin. The mean standardised uptake value (SUVmean) of the pancreas was used to determine the amount of beta cell mass. RESULTS Participants with LGV had lower HbA1c (46.0 mmol/mol [44.5-52.5] [6.4% (6.3-7)] vs 80 mmol/mol [69.0-110] [9.5% (8.5-12.2)], p=0.001) and higher time in range (TIR) (75.6% [73.5-90.3] vs 38.7% [25.1-48.5], p=0.002) than those with HGV. The SUVmean of the pancreas was higher for the LGV than for the HGV group (5.1 [3.6-5.6] vs 2.9 [2.1-3.4], p=0.008). The AUCC-peptide:AUCglucose ratio was numerically, but not statistically, higher in the LGV compared with the HGV group (2.7×10-2 [6.2×10-4-5.3×10-2] vs 9.3×10-4 [4.7×10-4-5.2×10-3], p=0.21). SUVmean correlated with the AUCC-peptide:AUCglucose ratio (Pearson r=0.64, p=0.01), as well as with the TIR (r=0.64, p=0.01) and the SD of interstitial glucose levels (r=-0.66, p=0.007). CONCLUSION/INTERPRETATION Our data show higher beta cell mass in people with type 1 diabetes and LGV than in those with HGV, independent of beta cell function.
Collapse
Affiliation(s)
- Theodorus J P Jansen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Maarten Brom
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marti Boss
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Mijke Buitinga
- Nutrition and Movement Sciences, Maastricht University, Maastricht, the Netherlands
- Radiology and Nuclear Medicine, Maastricht UMC+, Maastricht, the Netherlands
| | - Cees J Tack
- Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lian A van Meijel
- Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Internal Medicine, Maxima Medical Center, Veldhoven, the Netherlands
| | - Bastiaan E de Galan
- Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Internal Medicine, Maastricht UMC+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
35
|
Ginnard OZB, Redondo MJ, Lyons SK. Diabetes of Unclear Type in an Adolescent Boy With Multiple Islet-cell Autoantibody Positivity Successfully Managed With Glucagon-like Peptide-1 Receptor Agonist Alone: A Case Report. Can J Diabetes 2023; 47:90-93. [PMID: 36075851 PMCID: PMC10024250 DOI: 10.1016/j.jcjd.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 01/31/2023]
Abstract
Diabetes classification has traditionally considered type 1 and type 2 diabetes as 2 separate entities with different pathogenic mechanisms. However, clinicians and researchers see increasingly more exceptions to this conventional paradigm, leading to a concept of mixed phenotypes in diabetes classification. Herein we report the case of an adolescent with unclear diabetes type due to the presence of obesity, robust endogenous insulin production, multiple islet autoantibody positivity and severe hyperglycemia at diabetes diagnosis that has been successfully treated with liraglutide therapy alone. Our case report highlights the difficulty of diabetes classification and subsequent need for personalized medicine with regard to diabetes management.
Collapse
Affiliation(s)
- Olivia Z B Ginnard
- Section of Diabetes and Endocrinology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States.
| | - Maria J Redondo
- Section of Diabetes and Endocrinology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States
| | - Sarah K Lyons
- Section of Diabetes and Endocrinology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
36
|
Vollenbrock CE, Mul D, Dekker P, Birnie E, de Vries-Velraeds MMC, Boesten L, Groen J, Geelhoed-Duijvestijn PHLM, Aanstoot HJ, Wolffenbuttel BHR. Fasting and meal-stimulated serum C-peptide in long-standing type 1 diabetes mellitus. Diabet Med 2023; 40:e15012. [PMID: 36398450 PMCID: PMC10107202 DOI: 10.1111/dme.15012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
AIMS This study aims to evaluate the stability of C-peptide over time and to compare fasting C-peptide and C-peptide response after mixed-meal tolerance test (MMTT) at T90 or T120 with C-peptide area under the curve (AUC) in long-standing type 1 diabetes. METHODS We included 607 type 1 diabetes individuals with diabetes duration >5 years. C-peptide concentrations (ultrasensitive assay) were collected in the fasting state, and in a subpopulation after MMTT (T0, just prior to, T30-T60-T90-T120, 30-120 min after ingestion of mixed-meal) (n = 168). Fasting C-peptide concentrations (in n = 535) at Year 0 and Year 1 were compared. The clinical determinants associated with residual C-peptide secretion and the correspondence of C-peptide at MMTT T90 / T120 and total AUC were assessed. RESULTS A total of 153 participants (25%) had detectable fasting serum C-peptide (i.e ≥ 3.8 pmol/L). Fasting C-peptide was significantly lower at Year 1 (p < 0.001, effect size = -0.16). Participants with higher fasting C-peptide had a higher age at diagnosis and shorter disease duration and were less frequently insulin pump users. Overall, 109 of 168 (65%) participants had both non-detectable fasting and post-meal serum C-peptide concentrations. The T90 and T120 C-peptide values at MMTT were concordant with total AUC. In 17 (10%) individuals, C-peptide was only detectable at MMTT and not in the fasting state. CONCLUSIONS Stimulated C-peptide was detectable in an additional 10% of individuals compared with fasting in individuals with >5 years of diabetes duration. T90 and T120 MMTT measurements showed good concordance with the MMTT total AUC. Overall, there was a decrease of C-peptide at 1-year follow-up.
Collapse
Affiliation(s)
- Charlotte E Vollenbrock
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dick Mul
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | - Pim Dekker
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | - Erwin Birnie
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | | | - Lianne Boesten
- Department of Clinical Chemistry, IJsselland Ziekenhuis, Capelle aan den IJssel, The Netherlands
| | - Joost Groen
- Department of Clinical Chemistry, IJsselland Ziekenhuis, Capelle aan den IJssel, The Netherlands
| | | | - Henk-Jan Aanstoot
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Infante M, Ricordi C. The unique pathophysiological features of diabetes mellitus secondary to total pancreatectomy: proposal for a new classification distinct from diabetes of the exocrine pancreas. Expert Rev Endocrinol Metab 2023; 18:19-32. [PMID: 36692892 DOI: 10.1080/17446651.2023.2168645] [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] [Received: 09/07/2022] [Accepted: 01/11/2023] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Diabetes of the exocrine pancreas (DEP; a.k.a. pancreatic diabetes or pancreatogenic diabetes or type 3c diabetes mellitus or T3cDM) refers to different diabetes types resulting from disorders of the exocrine pancreas. DEP is characterized by the structural and functional loss of glucose-normalizing insulin secretion in the context of exocrine pancreatic dysfunction. Among these forms, new-onset diabetes mellitus secondary to total pancreatectomy (TP) has unique pathophysiological and clinical features, for which we propose a new nomenclature such as post-total pancreatectomy diabetes mellitus (PTPDM). AREAS COVERED TP results in the complete loss of pancreatic parenchyma, with subsequent absolute insulinopenia and lifelong need for exogenous insulin therapy. Patients with PTPDM also exhibit deficiency of glucagon, amylin and pancreatic polypeptide. These endocrine abnormalities, coupled with increased peripheral insulin sensitivity, deficiency of pancreatic enzymes and TP-related modifications of gastrointestinal anatomy, can lead to marked glucose variability and increased risk of iatrogenic (insulin-induced) severe hypoglycemic episodes ('brittle diabetes'). EXPERT OPINION We believe that diabetes mellitus secondary to TP should not be included in the DEP spectrum in light of its peculiar pathophysiological and clinical features. Therefore, we propose a new classification for this entity, that would likely provide more accurate prognosis and treatment strategies.
Collapse
Affiliation(s)
- Marco Infante
- Cell Transplant Center, Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL, USA
- Section of Diabetes and Metabolic Disorders, UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
- Diabetes Research Institute Federation (DRIF), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Rome, Italy
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
38
|
La Noce M, Nicoletti GF, Papaccio G, Del Vecchio V, Papaccio F. Insulitis in Human Type 1 Diabetic Pancreas: From Stem Cell Grafting to Islet Organoids for a Successful Cell-Based Therapy. Cells 2022; 11:3941. [PMID: 36497199 PMCID: PMC9740394 DOI: 10.3390/cells11233941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease with immune cells' islet infiltration (called "insulitis"), which leads to beta cell loss. Despite being the critical element of T1D occurrence and pathogenesis, insulitis is often present in a limited percentage of islets, also at diagnosis. Therefore, it is needed to define reproducible methods to detect insulitis and beta-cell decline, to allow accurate and early diagnosis and to monitor therapy. However, this goal is still far due to the morphological aspect of islet microvasculature, which is rather dense and rich, and is considerably rearranged during insulitis. More studies on microvasculature are required to understand if contrast-enhanced ultrasound sonography measurements of pancreatic blood-flow dynamics may provide a clinically deployable predictive marker to predict disease progression and therapeutic reversal in pre-symptomatic T1D patients. Therefore, it is needed to clarify the relation between insulitis and the dynamics of β cell loss and with coexisting mechanisms of dysfunction, according to clinical stage, as well as the micro vessels' dynamics and microvasculature reorganization. Moreover, the ideal cell-based therapy of T1D should start from an early diagnosis allowing a sufficient isolation of specific Procr+ progenitors, followed by the generation and expansion of islet organoids, which could be transplanted coupled to an immune-regulatory therapy which will permit the maintenance of pancreatic islets and an effective and long-lasting insulitis reversal.
Collapse
Affiliation(s)
- Marcella La Noce
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “L. Vanvitelli”, Via L. de Crecchio 6, 80138 Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy
| | - Vitale Del Vecchio
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy
| | - Federica Papaccio
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Via Salvador Allende, 84081 Baronissi, Italy
| |
Collapse
|
39
|
Trawley S, Ward GM, Vogrin S, Colman PG, Fourlanos S, Grills CA, Lee MH, MacIsaac RJ, Alipoor AM, O'Neal DN, O'Regan NA, Sundararajan V, McAuley SA. Glucose profiles of older adults with type 1 diabetes using sensor-augmented pump therapy in Australia: pre-randomisation results from the ORACL study. THE LANCET. HEALTHY LONGEVITY 2022; 3:e839-e848. [PMID: 36410370 DOI: 10.1016/s2666-7568(22)00266-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Older adults with type 1 diabetes are recommended modified glucose targets. However, data on the effects of diabetes technology in older age are scarce. We assessed older adults established on sensor-augmented insulin pump therapy during clinical trial run-in and compared their continuous glucose monitoring (CGM) profiles with consensus recommendations. We aimed to provide insight into the applicability of currently recommended CGM-based targets while accounting for current Diabetes UK guidelines. METHODS In this analysis, adults aged 60 years or older with type 1 diabetes with a duration of at least 10 years and entering the Older Adult Closed Loop (ORACL) trial were studied. The trial was done at two tertiary hospitals in Australia. Individuals who were independent with diabetes self-management, as well as those receiving caregiver assistance for their diabetes management, were eligible for inclusion. Participants underwent baseline clinical assessment, which included medical history and examination, testing for frailty, functional ability, cognitive functioning, psychosocial wellbeing, and subjective sleep quality; fasting venous blood samples were collected for C-peptide, glucose, and glycated haemoglobin A1c measurement. Sensor-augmented pumps, carbohydrate-counting education, and diabetes education were provided to participants by diabetes nurse educators, dietitians, and endocrinologists experienced in type 1 diabetes clinical care. CGM data were subsequently collected for 2 weeks during sensor-augmented pump therapy. The ORACL trial is registered with the Australian New Zealand Clinical Trial Registry, ACTRN12619000515190. FINDINGS Our analysis included all 30 participants who completed the ORACL trial run-in-19 (63%) women and 11 (37%) men (mean age 67 years [SD 5], median diabetes duration 38 years [IQR 20-47], and insulin total daily dose 0·55 units [0·41-0·66] per kg bodyweight). Ten (33%) of 30 participants had impaired hypoglycaemia awareness and six (20%) were pre-frail; none were frail. The median CGM time in range 3·9-10·0 mmol/L was 71% (IQR 64-79). The time spent with glucose above 10·0 mmol/L was 27% (18-35) and above 13·9 mmol/L was 3·9% (2·4-10·2). The time with glucose below 3·9 mmol/L was 2·0% (1·2-3·1) and the time below 3·0 mmol/L was 0·2% (0·1-0·4). Only two (7%) of 30 participants met all CGM-based consensus recommendations modified for older adults. Time in hypoglycaemia was lower among the 16 participants with predictive low-glucose alerts enabled than among the 14 participants not using predictive low-glucose alerts (median difference -1·1 percentage points [95% CI -2·0 to -0·1]; p=0·038). This difference was even greater overnight (-2·3 percentage points [-3·2 to -1·0]; p=0·0018). One serious adverse event occurred (elective cardiac stent). INTERPRETATION Using sensor-augmented pumps after multidisciplinary education, this group of older adults without frailty achieved a time in range far exceeding minimum consensus recommendations. However, the current stringent hypoglycaemia recommendations for all older adults were not met. Predictive low alerts could reduce hypoglycaemia, particularly overnight. Investigation into the effectiveness of CGM-based targets that consider frailty, functional status, and diabetes therapies for older adults is warranted. FUNDING JDRF and Diabetes Australia.
Collapse
Affiliation(s)
- Steven Trawley
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Psychology, The Cairnmillar Institute, Melbourne, VIC Australia
| | - Glenn M Ward
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Sara Vogrin
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Peter G Colman
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Spiros Fourlanos
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, VIC, Australia; Australian Centre for Accelerating Diabetes Innovations, Melbourne, VIC, Australia
| | - Charlotte A Grills
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Melissa H Lee
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Richard J MacIsaac
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia; Australian Centre for Accelerating Diabetes Innovations, Melbourne, VIC, Australia
| | - Andisheh Mohammad Alipoor
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - David N O'Neal
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Niamh A O'Regan
- Department of Geriatric Medicine, Waterford Integrated Care for Older People, University Hospital Waterford, Waterford, Ireland
| | - Vijaya Sundararajan
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Public Health, La Trobe University, Melbourne, VIC Australia
| | - Sybil A McAuley
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia; Department of Psychology, The Cairnmillar Institute, Melbourne, VIC Australia.
| |
Collapse
|
40
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|
41
|
Liu W, Ma Y, Cai X, Zhu Y, Zhang M, Li J, Chen J, Shi D, Ji L. Preserved C-peptide secretion is associated with higher time in range (TIR) on intermittently scanned continuous glucose monitoring in Chinese adults with type 1 diabetes. Endocr Connect 2022; 11:e220244. [PMID: 36136936 PMCID: PMC9641764 DOI: 10.1530/ec-22-0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022]
Abstract
Objective To explore the relationship between C-peptide secretion and time in range (TIR) in adult patients with type 1 diabetes. Methods From December 2018 to December 2020, 76 type 1 diabetes participants were enrolled from the Department of Endocrinology and Metabolism of Peking University People's Hospital. All participants wore intermittently scanned continuous glucose monitoring (isCGM), and insulin dosage was adjusted according to standardized clinical procedures. Subjects were divided into low C-peptide group (<10 pmol/L) and preserved C-peptide group (10-200 pmol/L) based on fasting serum C-peptide levels. Differences of TIR, metrics related to glucose variability and hypoglycemic events were compared. Results A total of 94,846 isCGM values obtained from 39 male and 37 female participants were analyzed. Individuals with preserved C-peptide secretion had shorter diabetes duration (2.0 (0.5, 10.0) vs 10.0 (3.0, 18.3) years, P = 0.002). TIR was higher in the individuals with preserved C-peptide than those with decreased C-peptide (67.1% (54.2, 75.8) vs 45.5% (33.9, 56.1), P < 0.001), and time above range was significantly lower in those with preserved C-peptide (28.0% (15.6, 42.4) vs 49.4% (39.1, 64.2), P < 0.001). Preserved C-peptide was associated with lower glucose variability, as defined by s.d. (3.0 mmol/L (2.6, 3.4) vs 3.8 mmol/L (3.2, 4.3), P < 0.001) and interquartile range (4.3 mmol/L (3.1, 4.8) vs 5.3 mmol/L (4.5, 6.3), P < 0.001). Metrics related to hypoglycemia were not different between the two groups. Conclusion Preserved C-peptide secretion was associated with higher TIR and lower glucose variability in Chinese type 1 diabetes adults.
Collapse
Affiliation(s)
- Wei Liu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Yunke Ma
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Yu Zhu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Mingxia Zhang
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Juan Li
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Jing Chen
- School of Automation, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Dawei Shi
- School of Automation, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, People’s Republic of China
| |
Collapse
|
42
|
Abstract
First envisioned by early diabetes clinicians, a person-centred approach to care was an aspirational goal that aimed to match insulin therapy to each individual's unique requirements. In the 100 years since the discovery of insulin, this goal has evolved to include personalised approaches to type 1 diabetes diagnosis, treatment, prevention and prediction. These advances have been facilitated by the recognition of type 1 diabetes as an autoimmune disease and by advances in our understanding of diabetes pathophysiology, genetics and natural history, which have occurred in parallel with advancements in insulin delivery, glucose monitoring and tools for self-management. In this review, we discuss how these personalised approaches have improved diabetes care and how improved understanding of pathogenesis and human biology might inform precision medicine in the future.
Collapse
Affiliation(s)
- Alice L J Carr
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
43
|
Noso S, Babaya N, Hiromine Y, Taketomo Y, Niwano F, Yoshida S, Ikegami H. Metabolic signatures of β-cell destruction in type 1 diabetes. J Diabetes Investig 2022; 14:48-57. [PMID: 36227003 PMCID: PMC9807153 DOI: 10.1111/jdi.13926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/24/2022] [Accepted: 09/27/2022] [Indexed: 01/07/2023] Open
Abstract
AIMS/INTRODUCTION In the development of type 1 diabetes, metabolites are significantly altered and might be involved in β-cell destruction and protection. We aimed to identify new metabolic markers of β-cell destruction in type 1 diabetes patients. MATERIALS AND METHODS A total of 33 participants were recruited for this cross-sectional observational study: 23 with type 1 diabetes, seven with type 2 diabetes and three healthy controls. Those with type 1 diabetes were further subdivided into three groups: new-onset, microsecretors and complete lack of endogenous insulin in type 1 diabetes. RESULTS Metabolomic analysis identified a total of 737 peaks, and partial least square analysis was successful in discriminating between the three groups of type 1 diabetes. Among the factor loadings discriminating type 1 diabetes, 3-phenylpropionic acid (r = 0.80, P = 4.7E-6 ) and hypotaurine (r = -0.484, P = 1.9E-2 ) strongly contributed to identifying new-onset type 1 diabetes, and 5-methylcytosine to identifying complete-lack type 1 diabetes (r = 0.586, P = 6.5E-3 ). Reporter operating characteristics analysis, including all type 1 diabetes, type 2 diabetes and healthy controls, showed that high 3-phenylpropionic acid (Pc <0.0001) and low hypotaurine (Pc <0.0001) were useful for identifying new-onset type 1 diabetes, and high 5-methylcytosine (Pc = 0.002) for the complete-lack type 1 diabetes. CONCLUSIONS In the present study, metabolic signatures were shown to be useful in identifying type 1 diabetes at different clinical stages, and 3-phenylpropionic acid and hypotaurine are novel biomarkers for identifying new-onset type 1 diabetes, suggesting the involvement of the gut bacterial environment, anti-oxidant mechanisms through the hypotaurine-taurine pathway and methylated deoxyribonucleic acid fragmentation in the process of β-cell destruction.
Collapse
Affiliation(s)
- Shinsuke Noso
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| | - Naru Babaya
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| | - Yoshihisa Hiromine
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| | - Yasunori Taketomo
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| | - Fumimaru Niwano
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| | - Sawa Yoshida
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| | - Hiroshi Ikegami
- Department of Endocrinology, Metabolism and DiabetesKindai University Faculty of MedicineOsakaJapan
| |
Collapse
|
44
|
Kulkarni S, Posgai AL, Kusmartseva I, Wasserfall CH, Atkinson MA, Butler AE. Exocrine and Endocrine Inflammation Increases Cellular Replication in the Pancreatic Duct Compartment in Type 1 Diabetes. J Endocr Soc 2022; 6:bvac136. [PMID: 36249412 PMCID: PMC9557836 DOI: 10.1210/jendso/bvac136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Indexed: 01/21/2023] Open
Abstract
Context We recently demonstrated increased cellular proliferation in the pancreatic ductal gland (PDG) compartment of organ donors with type 1 diabetes, suggesting that PDGs may harbor progenitor cells capable of pancreatic regeneration. Objective We evaluated the impact of diabetes and pancreatic inflammation on PDG and interlobular duct (ILD) cellular proliferation and profiles. Methods Endocrine hormone expression (insulin, glucagon, somatostatin, pancreatic polypeptide) and proliferating Ki67+ cells were localized within the PDG and ILD compartments by multicolor immunohistochemistry in cross-sections from the head, body, and tail regions of pancreata from those with (n = 31) or without type 1 diabetes (n = 43). Whole-slide scanned images were analyzed using digital pathology. Results Type 1 diabetes donors with insulitis or histologically identified pancreatitis had increased cellular replication in the ILD and PDG compartments. Interestingly, while cellular proliferation within the pancreatic ductal tree was significantly increased in type 1 diabetes (PDG mean = 3.36%, SEM = 1.06; ILD mean = 2.78%, SEM = 0.97) vs nondiabetes(ND) subjects without pancreatic inflammation (PDG mean = 1.18%, SEM = 0.42; ILD mean = 0.74%, SEM = 0.15, P < 0.05), robust replication was also observed in ND donors with pancreatitis (PDG mean = 3.52%, SEM = 1.33; ILD mean = 2.18%, SEM = 0.54, P < 0.05). Few polyhormonal cells were present in the ILD (type 1 diabetes = 0.04 ± 0.02%; ND = 0.08 ± 0.03%, P = 0.40) or PDG compartment (type 1 diabetes = 0.02 ± 0.01%; ND = 0.08 ± 0.13%, P = 0.63). Conclusion These data suggest that increased pancreatic ductal cell replication is associated with sustained pancreatic inflammation; however, as replicating cells were hormone-negative, PDGs do not appear to represent a compelling endogenous source of hormone-positive endocrine cells.
Collapse
Affiliation(s)
- Shweta Kulkarni
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Irina Kusmartseva
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Alexandra E Butler
- Department of Research, Royal College of Surgeons in Ireland-Bahrain, 15503 Adliya, Bahrain
| |
Collapse
|
45
|
Nowak C, Lind M, Sumnik Z, Pelikanova T, Nattero-Chavez L, Lundberg E, Rica I, Martínez-Brocca MA, Ruiz de Adana M, Wahlberg J, Hanas R, Hernandez C, Clemente-León M, Gómez-Gila A, Ferrer Lozano M, Sas T, Pruhova S, Dietrich F, Puente-Marin S, Hannelius U, Casas R, Ludvigsson J. Intralymphatic GAD-Alum (Diamyd®) Improves Glycemic Control in Type 1 Diabetes With HLA DR3-DQ2. J Clin Endocrinol Metab 2022; 107:2644-2651. [PMID: 35665810 PMCID: PMC9721339 DOI: 10.1210/clinem/dgac343] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Indexed: 01/07/2023]
Abstract
AIMS Residual beta cell function in type 1 diabetes (T1D) is associated with lower risk of complications. Autoantigen therapy with GAD-alum (Diamyd) given in 3 intralymphatic injections with oral vitamin D has shown promising results in persons with T1D carrying the human leukocyte antigen (HLA) DR3-DQ2 haplotype in the phase 2b trial DIAGNODE-2. We aimed to explore the efficacy of intralymphatic GAD-alum on blood glucose recorded by continuous glucose monitoring (CGM). METHODS DIAGNODE-2 (NCT03345004) was a multicenter, randomized, placebo-controlled, double-blind trial of 109 recent-onset T1D patients aged 12 to 24 years with GAD65 antibodies and fasting C-peptide > 0.12 nmol/L, which randomized patients to 3 intralymphatic injections of 4 μg GAD-alum and oral vitamin D, or placebo. We report results for exploratory endpoints assessed by 14-day CGM at months 0, 6, and 15. Treatment arms were compared by mixed-effects models for repeated measures adjusting for baseline values. RESULTS We included 98 patients with CGM recordings of sufficient quality (DR3-DQ2-positive patients: 27 GAD-alum-treated and 15 placebo-treated). In DR3-DQ2-positive patients, percent of time in range (TIR, 3.9-10 mmol/L) declined less between baseline and month 15 in GAD-alum-treated compared with placebo-treated patients (-5.1% and -16.7%, respectively; P = 0.0075), with reduced time > 13.9 mmol/L (P = 0.0036), and significant benefits on the glucose management indicator (P = 0.0025). No differences were detected for hypoglycemia. GAD-alum compared to placebo lowered the increase in glycemic variability (standard deviation) observed in both groups (P = 0.0219). Change in C-peptide was correlated with the change in TIR. CONCLUSIONS Intralymphatic GAD-alum improves glycemic control in recently diagnosed T1D patients carrying HLA DR3-DQ2.
Collapse
Affiliation(s)
- Christoph Nowak
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14183 Huddinge, Sweden
- Diamyd Medical AB, 11135 Stockholm, Sweden
| | - Marcus Lind
- Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden, Sahlgrenska University Hospital, Gothenburg and NU-Hospital Group, S41553, Uddevalla, Sweden
| | - Zdenek Sumnik
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, 15000 Prague, Czech Republic
| | - Terezie Pelikanova
- Diabetes Centre of the Institute of Clinical and Experimental Medicine, 14000 Prague, Czech Republic
| | - Lía Nattero-Chavez
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Elena Lundberg
- Institution of Clinical Science, Department of Pediatrics, Umeå University, Norrland University Hospital, 93451 Umeå, Sweden
| | - Itxaso Rica
- Department of Pediatric Endocrinology, Cruces University Hospital, 48902 Bilbao, Ciberdem, Spain
| | - Maria A Martínez-Brocca
- Department of Endocrinology, Virgen Macarena Hospital, Department of Endocrinology and Nutrition, Virgen Macarena University Hospital, 41009 Sevilla, Spain
| | - MariSol Ruiz de Adana
- Diabetes Unit, Department of Endocrinology and Nutrition, Ibima, Ciberdem, General University Hospital, 29010 Malaga, Spain
| | - Jeanette Wahlberg
- Department of Endocrinology in Linköping and Department of Health, Medicine and Caring Sciences, Linköping University, 58183 Linköping, Sweden
- Department of Internal Medicine, School of Health and Medical Sciences, Örebro University, 70281 Örebro, Sweden
| | - Ragnar Hanas
- Department of Pediatrics, NU Hospital Group, 45153 Uddevalla, Sweden
| | - Cristina Hernandez
- Department of Endocrinology and Nutrition, Vall d’Hebron Hospital, 08035 Barcelona, Ciberdem, Spain
| | - Maria Clemente-León
- Department of Endocrinology, Pediatric Service, Vall d’Hebron Hospital, 08035 Barcelona, CibererSpain
| | - Ana Gómez-Gila
- Pediatric Endocrinology Service, Virgen del Rocío University Hospital, 41013 Sevilla, Spain
| | - Marta Ferrer Lozano
- Department of Pediatric Endocrinology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Theo Sas
- Diabeter, National Treatment and Research Center for Children, Adolescents and Young Adults with type 1 diabetes, and Department of Pediatric Endocrinology, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Stepanka Pruhova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, 15000 Prague, Czech Republic
| | - Fabricia Dietrich
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Sara Puente-Marin
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, 58183 Linköping, Sweden
| | | | - Rosaura Casas
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences and Crown Princess Victoria Children´s Hospital, Linköping University, 58183 Linköping, Sweden
| | - Johnny Ludvigsson
- Correspondence: Johnny Ludvigsson, MD, PhD, Professor of Pediatrics, Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, and Crown Princess Victoria Children’s Hospital, Linköping University, SE-58185 Linköping, Sweden.
| |
Collapse
|
46
|
Carr AL, Inshaw JR, Flaxman CS, Leete P, Wyatt RC, Russell LA, Palmer M, Prasolov D, Worthington T, Hull B, Wicker LS, Dunger DB, Oram RA, Morgan NG, Todd JA, Richardson SJ, Besser RE. Circulating C-Peptide Levels in Living Children and Young People and Pancreatic β-Cell Loss in Pancreas Donors Across Type 1 Diabetes Disease Duration. Diabetes 2022; 71:1591-1596. [PMID: 35499624 PMCID: PMC9233242 DOI: 10.2337/db22-0097] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/16/2022] [Indexed: 11/13/2022]
Abstract
C-peptide declines in type 1 diabetes, although many long-duration patients retain low, but detectable levels. Histological analyses confirm that β-cells can remain following type 1 diabetes onset. We explored the trends observed in C-peptide decline in the UK Genetic Resource Investigating Diabetes (UK GRID) cohort (N = 4,079), with β-cell loss in pancreas donors from the network for Pancreatic Organ donors with Diabetes (nPOD) biobank and the Exeter Archival Diabetes Biobank (EADB) (combined N = 235), stratified by recently reported age at diagnosis endotypes (<7, 7-12, ≥13 years) across increasing diabetes durations. The proportion of individuals with detectable C-peptide declined beyond the first year after diagnosis, but this was most marked in the youngest age group (<1-year duration: age <7 years: 18 of 20 [90%], 7-12 years: 107 of 110 [97%], ≥13 years: 58 of 61 [95%] vs. 1-5 years postdiagnosis: <7 years: 172 of 522 [33%], 7-12 years: 604 of 995 [61%], ≥13 years: 225 of 289 [78%]). A similar profile was observed in β-cell loss, with those diagnosed at younger ages experiencing more rapid loss of islets containing insulin-positive (insulin+) β-cells <1 year postdiagnosis: age <7 years: 23 of 26 (88%), 7-12 years: 32 of 33 (97%), ≥13 years: 22 of 25 (88%) vs. 1-5 years postdiagnosis: <7 years: 1 of 12 (8.3%), 7-12 years: 7 of 13 (54%), ≥13 years: 7 of 8 (88%). These data should be considered in the planning and interpretation of intervention trials designed to promote β-cell retention and function.
Collapse
Affiliation(s)
- Alice L.J. Carr
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
- Corresponding authors: Alice Carr, , Rachel Besser, , or Sarah Richardson,
| | - Jamie R.J. Inshaw
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Christine S. Flaxman
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Pia Leete
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Rebecca C. Wyatt
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Lydia A. Russell
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Matthew Palmer
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Dmytro Prasolov
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Thomas Worthington
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Bethany Hull
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Linda S. Wicker
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - David B. Dunger
- Department of Paediatrics, Addenbrooke's Hospital, University of Cambridge, Cambridge, U.K
| | - Richard A. Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Noel G. Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - John A. Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
- National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
| | - Sarah J. Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
- Corresponding authors: Alice Carr, , Rachel Besser, , or Sarah Richardson,
| | - Rachel E.J. Besser
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
- National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
- Corresponding authors: Alice Carr, , Rachel Besser, , or Sarah Richardson,
| |
Collapse
|
47
|
Ikegami H, Hiromine Y, Noso S. Insulin-dependent diabetes mellitus in older adults: Current status and future prospects. Geriatr Gerontol Int 2022; 22:549-553. [PMID: 35711119 PMCID: PMC9542793 DOI: 10.1111/ggi.14414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 11/27/2022]
Abstract
The recent increase in life expectancy has resulted in an increase in the number of older adults with diabetes mellitus. In addition to type 2 diabetes, in which aging is a well‐known risk factor, individuals with type 1 and other types of diabetes are also increasing owing to longevity in the general population and improved prognosis of the disease and comorbidities. Insulin‐dependent state in type 1 diabetes and other types of diabetes, such as diabetes after pancreatectomy, inevitably requires insulin treatment for survival; however, daily injection of insulin is often hampered in older adults due to impaired cognitive function or limited activities of daily living. In this review, we aimed to discuss the current situation of insulin‐dependent diabetes mellitus in older adults and highlight future prospects. Geriatr Gerontol Int 2022; 22: 549–553.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Department of Endocrinology, Metabolism and Diabetes, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yoshihisa Hiromine
- Department of Endocrinology, Metabolism and Diabetes, Kindai University Faculty of Medicine, Osaka, Japan
| | - Shinsuke Noso
- Department of Endocrinology, Metabolism and Diabetes, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
48
|
Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, Pastori RL, Russ HA, Wagner BK, Dominguez-Bendala J. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022; 60:101477. [PMID: 35331962 PMCID: PMC8987999 DOI: 10.1016/j.molmet.2022.101477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stem cell therapies are finally coming of age as a viable alternative to pancreatic islet transplantation for the treatment of insulin-dependent diabetes. Several clinical trials using human embryonic stem cell (hESC)-derived β-like cells are currently underway, with encouraging preliminary results. Remaining challenges notwithstanding, these strategies are widely expected to reduce our reliance on human isolated islets for transplantation procedures, making cell therapies available to millions of diabetic patients. At the same time, advances in our understanding of pancreatic cell plasticity and the molecular mechanisms behind β-cell replication and regeneration have spawned a multitude of translational efforts aimed at inducing β-cell replenishment in situ through pharmacological means, thus circumventing the need for transplantation. SCOPE OF REVIEW We discuss here the current state of the art in hESC transplantation, as well as the parallel quest to discover agents capable of either preserving the residual mass of β-cells or inducing their proliferation, transdifferentiation or differentiation from progenitor cells. MAJOR CONCLUSIONS Stem cell-based replacement therapies in the mold of islet transplantation are already around the corner, but a permanent cure for type 1 diabetes will likely require the endogenous regeneration of β-cells aided by interventions to restore the immune balance. The promise of current research avenues and a strong pipeline of clinical trials designed to tackle these challenges bode well for the realization of this goal.
Collapse
Affiliation(s)
- G Basile
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - M M F Qadir
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - F Mauvais-Jarvis
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - A Vetere
- Broad Institute, Cambridge, MA, USA
| | - V Shoba
- Broad Institute, Cambridge, MA, USA
| | | | - R L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H A Russ
- Barbara Davis Center for Diabetes, Colorado University Anschutz Medical Campus, Aurora, CO, USA.
| | | | - J Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
49
|
Wang Y, Qin Y, Gu H, Zhang L, Wang J, Huang Y, Shi Y, Hu Q, Chen Y, Gu Y, Shi Y, Tao Y, Zhang M. High Residual β-cell Function in Chinese Patients With Autoimmune Type 1 Diabetes. J Clin Endocrinol Metab 2022; 107:e2348-e2358. [PMID: 35218654 DOI: 10.1210/clinem/dgac077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The destruction of pancreatic β cells causes type 1 diabetes mellitus (T1D), an autoimmune disease. Studies have demonstrated that there is heterogeneity in residual β-cell function in Caucasians; therefore, we aimed to evaluate β-cell function in Chinese autoimmune T1D patients. METHODS β-cell function was determined using oral glucose tolerance testing or standardized steamed bread meal tolerance test in 446 participants with autoantibody-positive T1D. Clinical factors, such as age onset, sex, duration, body mass index, autoantibodies, other autoimmune diseases, diabetic ketoacidosis, hypoglycemia events, glycosylated hemoglobin, and insulin dose, were retrieved. We also analyzed single nucleotide polymorphism (SNP) data for C-peptides from 144 participants enrolled in the Chinese-T1D genome-wide association study. RESULTS Of 446 T1D patients, 98.5%, 97.4%, 86.9%, and 42.6% of individuals had detectable C-peptide values (≥ 0.003 nmol/L) at durations of < 1 year, 1 to 2 years, 3 to 6 years, and ≥ 7 years, respectively. A total of 60.7% of patients diagnosed at ≥ 18 years old and 15.8% of those diagnosed at < 18 years had detectable C-peptide after ≥ 7 years from the diagnosis. Furthermore, the patients diagnosed at ≥ 18 years old had higher absolute values of stimulated C-peptide (≥ 0.2 nmol/L). Diabetic ketoacidosis, hypoglycemia events, and insulin doses were shown to be associated with β-cell function. SNPs rs1770 and rs55904 were associated with C-peptide levels. CONCLUSION Our results have indicated that there are high residuals of β-cell mass in Chinese patients with autoimmune T1D. These findings may aid in the consideration of therapeutic strategies seeking prevention and reversal of β-cell function among Chinese T1D patients.
Collapse
Affiliation(s)
- Yueshu Wang
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
- Department of Pediatrics, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yao Qin
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Huilan Gu
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Linyu Zhang
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jing Wang
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yiting Huang
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yuwen Shi
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Qizhen Hu
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yang Chen
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yong Gu
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yun Shi
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yang Tao
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Mei Zhang
- Department of Endocrinology, the First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| |
Collapse
|
50
|
de Leur K, Vollenbrock C, Dekker P, de Vries M, Birnie E, Mul D, Wolffenbuttel BHR, Groen J, Aanstoot H, Boesten L. How low is really low? Comparison of two C-peptide assays to establish residual C-peptide production in type 1 diabetes. Diabet Med 2022; 39:e14785. [PMID: 34989030 PMCID: PMC9303196 DOI: 10.1111/dme.14785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/04/2022] [Indexed: 01/12/2023]
Abstract
INTRODUCTION C-peptide is an important marker to assess residual insulin production in individuals with type 1 diabetes (T1D). The accuracy and detection limits of C-peptide assays are important to detect C-peptide microsecretion and to reliably observe changes over time in these people. We compared and verified two commercially available assays able to measure C-peptide in the picomolar range. METHODS The ultrasensitive Mercodia enzyme-linked immunosorbent C-peptide assay (ELISA) was compared with the Beckman immunoradiometric assay (IRMA) for C-peptide, assessing reproducibility (coefficient of variation [CV]), limit of blank (LoB), limit of detection (LoD) and limit of quantitation (LoQ). RESULTS For both assays within-run and between-run variation were high at the low (around the detection limit) C-peptide concentration range, with CVs of around 40%. LoB values for the ultrasensitive ELISA and the IRMA were 1.3 and 0.16 pmol/L respectively. LoD values were 2.4 and 0.54 pmol/L respectively. LoQ values were 9.7 and 3.8 pmol/L respectively. Only the IRMA met the specifications claimed by the manufacturer. CONCLUSIONS The IRMA provided the lowest threshold for quantification of serum C-peptide. LoQ of commercially available assays should be established in-house before applying them in research studies and clinical trials in which low C-peptide levels have clinical or scientific relevance.
Collapse
Affiliation(s)
- Kitty de Leur
- Department of Clinical ChemistryIJsselland HospitalCapelle aan den IJsselThe Netherlands
| | - Charlotte Vollenbrock
- Department of EndocrinologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Pim Dekker
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Martine de Vries
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Erwin Birnie
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Dick Mul
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Bruce H. R. Wolffenbuttel
- Department of EndocrinologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Joost Groen
- Department of Clinical ChemistryIJsselland HospitalCapelle aan den IJsselThe Netherlands
| | - Henk‐Jan Aanstoot
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Lianne Boesten
- Department of Clinical ChemistryIJsselland HospitalCapelle aan den IJsselThe Netherlands
| |
Collapse
|