1
|
Tagawa K, Matsui K, Tsukamura A, Shibata M, Tsutsui H, Nagai S, Maruo Y. Use of a long-term continuous glucose monitor for predicting sulfonylurea dose in patients with neonatal diabetes mellitus: a case series. Clin Pediatr Endocrinol 2025; 33:131-138. [PMID: 38993723 PMCID: PMC11234181 DOI: 10.1297/cpe.2023-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/17/2024] [Indexed: 07/13/2024] Open
Abstract
Neonatal diabetes mellitus (NDM) is a monogenic form of diabetes that presents with uncontrolled hyperglycemia during the first 6 months of life. NDM is a rare disease in which gene variants mainly cause β-cell loss or dysfunction (6q24 duplication, KCNJ11, and ABCC8). Although NDM is primarily treated through insulin therapy, it is highly challenging to manage blood glucose levels using insulin therapy during infancy. In contrast, KCNJ11 and ABCC8 mutant patients received oral sulfonylureas (SU) instead of insulin injections; however, the dose and frequency differ among individuals. Continuous glucose monitoring (CGM) is useful in patients with type 1 diabetes; but reports on patients with NDM are lacking. Herein, we report two cases of NDM with the KCNJ11 variant. We used CGM not only during insulin injection therapy but also after switching to oral SU therapy. The CGM data can also be used to determine the dose and frequency of SU. Furthermore, long-term CGM may be useful for adjusting SU dose and frequency, and maintaining good glycemic control not only during insulin injection but also during oral SU therapy.
Collapse
Affiliation(s)
- Koji Tagawa
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
- Department of Pediatrics, Sapporo Tokushukai Hospital, Hokkaido, Japan
| | - Katsuyuki Matsui
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
- Division of Endocrinology, Metabolism & Diabetes, Shiga Medical Center for Children, Shiga, Japan
| | - Atsushi Tsukamura
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Masami Shibata
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
- Department of Pediatrics, Ogaki Municipal Hospital, Gifu, Japan
| | - Hidemi Tsutsui
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Shizuyo Nagai
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
2
|
Pezzotta F, Sarale N, Spacco G, Tantari G, Bertelli E, Bracciolini G, Secco A, d’Annunzio G, Maghnie M, Minuto N, Bassi M. Safety and Efficacy of Using Advanced Hybrid Closed Loop Off-Label in an Infant Diagnosed with Permanent Neonatal Diabetes Mellitus: A Case Report and a Look to the Future. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1225. [PMID: 39457190 PMCID: PMC11506781 DOI: 10.3390/children11101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
The case report shows the safety and efficacy of insulin treatment with Advanced Hybrid Closed Loop (AHCL) system in a young patient affected by permanent neonatal diabetes mellitus (PNDM) due to chromosome 8 deletion syndrome involving the GATA4 gene. In the first days of life, he presented hyperglycaemia and started an intravenous insulin infusion therapy, replaced by a continuous subcutaneous insulin infusion (CSII) with Medtronic Minimed 780G® insulin pump (Medtronic, Northridge, CA, USA). At the age of 2 years, the off-label activation of SmartGuard® automated insulin delivery mode led to a great improvement in glycaemic control, reaching all recommended targets. At the 1-month follow-up visit, Time in Range (TIR) increased from 66% to 79%, with a Time in Tight Range (TTIR) of 55% and a reduction of 11% in time in hyperglycaemia and of 2% in time in hypoglycaemia. During the entire follow-up, no episodes of ketoacidosis or severe hypoglycaemia were observed and the patient maintained the glycaemic recommended targets reached at 1 month. Maintaining optimal glycaemic control and reducing hyperglycaemia are essential for brain growth and neurocognitive development in young patients. AHCL use should be considered to ensure good glycaemic control in patients affected by neonatal diabetes.
Collapse
Affiliation(s)
- Federico Pezzotta
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| | - Nicola Sarale
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| | - Giordano Spacco
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| | - Giacomo Tantari
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
| | - Enrica Bertelli
- Pediatric and Pediatric Emergency Unit, Children Hospital, Azienda Ospedaliera Universitaria SS Antonio e Biagio e C. Arrigo, 15121 Alessandria, Italy
| | - Giulia Bracciolini
- Pediatric and Pediatric Emergency Unit, Children Hospital, Azienda Ospedaliera Universitaria SS Antonio e Biagio e C. Arrigo, 15121 Alessandria, Italy
| | - Andrea Secco
- Pediatric and Pediatric Emergency Unit, Children Hospital, Azienda Ospedaliera Universitaria SS Antonio e Biagio e C. Arrigo, 15121 Alessandria, Italy
| | - Giuseppe d’Annunzio
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
| | - Mohamad Maghnie
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| | - Nicola Minuto
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
| | - Marta Bassi
- Pediatric Clinic and Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (M.B.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| |
Collapse
|
3
|
Li C, Qiao L, Ge J, Hu S, Yang H, Hu C, Li T. PLAGL1 overexpression induces cytoplasmic DNA accumulation that triggers cGAS/STING activation. J Cell Mol Med 2024; 28:e70130. [PMID: 39365284 PMCID: PMC11451391 DOI: 10.1111/jcmm.70130] [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: 06/25/2024] [Revised: 08/12/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
Pancreatic β-cell damage mediated by apoptosis is believed to be a main trigger of type 1 diabetes mellitus (T1DM), which is proposed as an organ-specific autoimmune disease mediated by T cells. Nonetheless, the fundamental origins of T1DM remain uncertain. Here, we illustrate that an increase in PLAGL1 expression induces β-cell apoptosis, as evidenced by mitochondrial membrane impairment and nucleolar degradation. The gene expression levels from cDNA samples were determined using qRT-PCR method. Western blot and Co-immunoprecipitation were applied for protein expression and interactions, respectively. Flow cytometry and TUNEL assay were used to detect pancreatic β cell apoptosis. Female NOD/LtJ mice with recent-onset T1DM has been used in in vivo studies. Glucose-stimulated insulin secretion (GSIS) and glucose tolerance test (GTT) method is used for islet function assessment. Haematoxylin and Eosin (H&E) and Immunohistochemistry (IHC) were performed to evalute histological improvement of islet beta. Subsequent cytoplasmic DNA accumulation triggers DNA senser, the cyclic guanosine monophosphate-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. STING activation further stimulates downstream IRF3 and NF-kB pathways, thus boost type-I interferon signalling and NF-kB mediated inflammation. These findings elucidate a molecular mechanism linking PLAGL1 induced cell apoptosis to type-I interferon signalling and suggest a potential benefit for targeting cGAS/STING in T1DM treatment.
Collapse
Affiliation(s)
- Cheng Li
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| | - Lingyan Qiao
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| | - Juan Ge
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| | - Sicui Hu
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| | - Hongxiu Yang
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| | - Conghui Hu
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| | - Tang Li
- Department of Pediatric Endocrinologic and Genetic and Metabolic DiseasesQingdao Women and Children's HospitalQingdaoChina
| |
Collapse
|
4
|
Li Y, Xu M, Chen J, Huang J, Cao J, Chen H, Zhang J, Luo Y, Wang Y, Sun J. Ameliorating and refining islet organoids to illuminate treatment and pathogenesis of diabetes mellitus. Stem Cell Res Ther 2024; 15:188. [PMID: 38937834 PMCID: PMC11210168 DOI: 10.1186/s13287-024-03780-7] [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: 04/02/2024] [Accepted: 06/01/2024] [Indexed: 06/29/2024] Open
Abstract
Diabetes mellitus, a significant global public health challenge, severely impacts human health worldwide. The organoid, an innovative in vitro three-dimensional (3D) culture model, closely mimics tissues or organs in vivo. Insulin-secreting islet organoid, derived from stem cells induced in vitro with 3D structures, has emerged as a potential alternative for islet transplantation and as a possible disease model that mirrors the human body's in vivo environment, eliminating species difference. This technology has gained considerable attention for its potential in diabetes treatment. Despite advances, the process of stem cell differentiation into islet organoid and its cultivation demonstrates deficiencies, prompting ongoing efforts to develop more efficient differentiation protocols and 3D biomimetic materials. At present, the constructed islet organoid exhibit limitations in their composition, structure, and functionality when compared to natural islets. Consequently, further research is imperative to achieve a multi-tissue system composition and improved insulin secretion functionality in islet organoid, while addressing transplantation-related safety concerns, such as tumorigenicity, immune rejection, infection, and thrombosis. This review delves into the methodologies and strategies for constructing the islet organoid, its application in diabetes treatment, and the pivotal scientific challenges within organoid research, offering fresh perspectives for a deeper understanding of diabetes pathogenesis and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Yushan Li
- Department of Endocrinology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Meiqi Xu
- Department of Biomedical Engineering, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jiali Chen
- Department of Endocrinology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiansong Huang
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaying Cao
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Huajing Chen
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiayi Zhang
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yukun Luo
- Department of Endocrinology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yazhuo Wang
- Tsinghua-Peking Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
| | - Jia Sun
- Department of Endocrinology, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Du T, Xia Y, Sun C, Gong Z, Liang L, Gong Z, Wang R, Lu D, Zhang K, Yang Y, Sun Y, Sun M, Sun Y, Xiao B, Qiu W. Clinical, genetic profile and therapy evaluation of 11 Chinese pediatric patients with Fanconi-Bickel syndrome. Orphanet J Rare Dis 2024; 19:75. [PMID: 38365697 PMCID: PMC10874070 DOI: 10.1186/s13023-024-03070-8] [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: 10/11/2023] [Accepted: 02/03/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Fanconi-Bickel syndrome (FBS) is a rare autosomal recessive disorder characterized by impaired glucose and galactose utilization as well as proximal renal tubular dysfunction. METHODS Clinical, biochemical, genetic, treatment, and follow-up data for 11 pediatric patients with FBS were retrospectively analysed. RESULTS Hepatomegaly (10/11), short stature (10/11) and hypophosphataemic rickets (7/11) were the most common initial symptoms. At diagnosis, all patients had decreased fasting blood glucose (FBG), plasma bicarbonate (HCO3-) and serum phosphorus, as well as elevated liver transaminases, alkaline phosphatase (AKP) and proximal renal tubular dysfunction. Two infant patients were misdiagnosed with transient neonatal diabetes mellitus. After therapy with uncooked cornstarch and conventional rickets treatment, remission of hepatomegaly was observed in all patients, with significant improvements in pre-prandial blood glucose, liver transaminases, triglyceride, plasma HCO3- and AKP (p < 0.05). At the last follow-up, 5/7 patients with elevated AKP had nephrocalcinosis. The mean height standard deviation score (Ht SDS) of eight patients with regular treatment increased from - 4.1 to -3.5 (p = 0.02). Recombinant human growth hormone (rhGH) was administered to 4/9 patients, but their Ht SDS did not improve significantly (p = 0.13). Fourteen variants of the SLC2A2 gene were identified, with six being novel, among which one was recurrent: c.1217T > G (p.L406R) (allele frequency: 4/22, 18%). Patients with biallelic missense variants showed milder metabolic acidosis than those with null variants. Two of five patients from nonconsanguineous families with rare homozygous variations showed 5.3 Mb and 36.6 Mb of homozygosity surrounding the variants, respectively; a region of homozygosity (ROH) involving the entire chromosome 3 covering the SLC2A2 gene, suggesting uniparental disomy 3, was detected in one patient. CONCLUSIONS Early diagnosis of FBS is difficult due to the heterogeneity of initial symptoms. Although short stature is a major issue of treatment for FBS, rhGH is not recommended in FBS patients who have normal GH stimulation tests. Patients with biallelic null variants may require alkali supplementation since urine bicarbonate loss is genetically related. ROH is a mechanism for rare homozygous variants of FBS in nonconsanguineous families.
Collapse
Affiliation(s)
- Taozi Du
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Yu Xia
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Chengkai Sun
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Zhuwen Gong
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Lili Liang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Zizhen Gong
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Ruifang Wang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Deyun Lu
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Kaichuang Zhang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Yi Yang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Yuning Sun
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Manqing Sun
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Yu Sun
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
- Department of Clinical Genetics Centre, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China
| | - Bing Xiao
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China.
- Department of Clinical Genetics Centre, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China.
| | - Wenjuan Qiu
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, 200092, Shanghai, China.
| |
Collapse
|
6
|
de Souza RB, Cabello PH, Rosado EL, Junior MC, de Medeiros Abreu G. What Do We Know about Neonatal Diabetes caused by PDX1 Mutations? Curr Diabetes Rev 2024; 21:e290124226471. [PMID: 38299270 DOI: 10.2174/0115733998265866231204070606] [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: 07/04/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Neonatal diabetes mellitus (NDM) is characterized by severe hyperglycemia, usually diagnosed in the first few months of an individual's life. It is a genetic disease and one of the main forms of monogenic diabetes. Changes in different genes have already been associated with NDM, including changes in the gene PDX1. METHODS In this review, we intend to summarize all neonatal diabetes cases caused by PDX1 mutations reported in the literature. For this purpose, we searched keywords in the literature from PubMed and articles cited by the HGMD database. The search retrieved 84 articles, of which 41 had their full text accessed. After applying the study exclusion criteria, nine articles were included. RESULTS Of those articles, we detected thirteen cases of NDM associated with changes in PDX1; the majority in homozygous or compound heterozygous patients. Until now, variants in the PDX1 gene have been a rare cause of NDM; however, few studies have included the screening of this gene in the investigation of neonatal diabetes. CONCLUSION In this review, we reinforce the importance of the PDX1 gene inclusion in genetic NGS panels for molecular diagnosis of NDM, and systematic morphological and functional exams of the pancreas when NDM is present.
Collapse
Affiliation(s)
- Ritiele Bastos de Souza
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Pedro Hernán Cabello
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Genetics, School of Health Science, University of Grande Rio, Rio de Janeiro, Brazil
| | - Eliane Lopes Rosado
- Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mário Campos Junior
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Gabriella de Medeiros Abreu
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Lian H, Gong S, Li M, Wang X, Wang F, Cai X, Liu W, Luo Y, Zhang S, Zhang R, Zhou L, Zhu Y, Ma Y, Ren Q, Zhang X, Chen J, Chen L, Wu J, Gao L, Zhou X, Li Y, Zhong L, Han X, Ji L. Prevalence and Clinical Characteristics of PDX1 Variant Induced Diabetes in Chinese Early-Onset Type 2 Diabetes. J Clin Endocrinol Metab 2023; 108:e1686-e1694. [PMID: 37279936 DOI: 10.1210/clinem/dgad303] [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: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/08/2023]
Abstract
CONTEXT Maturity-onset diabetes of the young 4 (MODY4) is caused by mutations of PDX1; its prevalence and clinical features are not well known. OBJECTIVE This study aimed to investigate the prevalence and clinical characteristics of MODY4 in Chinese people clinically diagnosed with early-onset type 2 diabetes (EOD), and to evaluate the relationship between the PDX1 genotype and the clinical phenotype. METHOD The study cohort consisted of 679 patients with EOD. PDX1 mutations were screened by DNA sequencing, and their pathogenicity was evaluated by functional experiments and American College of Medical Genetics and Genomics guidelines. MODY4 was diagnosed in individuals with diabetes who carry a pathogenic or likely pathogenic PDX1 variant. All reported cases were reviewed for analyzing the genotype-phenotype relationship. RESULT 4 patients with MODY4 were identified, representing 0.59% of this Chinese EOD cohort. All the patients were diagnosed before 35 years old, either obese or not obese. Combined with previously reported cases, the analysis revealed that the carriers of homeodomain variants were diagnosed earlier than those with transactivation domain variants (26.10 ± 11.00 vs 41.85 ± 14.66 years old, P < .001), and the proportions of overweight and obese individuals with missense mutation were higher than those with nonsense or frameshift mutations (27/34 [79.4%] vs 3/8 [37.5%], P = .031). CONCLUSION Our study suggested that MODY4 was prevalent in 0.59% of patients with EOD in a Chinese population. It was more difficult to identify clinically than other MODY subtypes owning to its clinical similarity to EOD. Also, this study revealed that there is some relationship between genotype and phenotype.
Collapse
Affiliation(s)
- Hong Lian
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Siqian Gong
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Meng Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Xirui Wang
- Department of Endocrinology, Beijing Airport Hospital. No. 49, Beijing 101318, China
| | - Fang Wang
- Department of Endocrinology, Capital Medical University Beijing Tiantan Hospital. No. 119, Beijing 100050, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Wei Liu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Yingying Luo
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Simin Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Rui Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Lingli Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Yu Zhu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Yumin Ma
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Qian Ren
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Xiuying Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Jing Chen
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Ling Chen
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Jing Wu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Leili Gao
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Xianghai Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Yufeng Li
- Department of Endocrinology, Beijing Pinggu Hospital. No. 59, Beijing 101200, China
| | - Liyong Zhong
- Department of Endocrinology, Capital Medical University Beijing Tiantan Hospital. No. 119, Beijing 100050, China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center. No. 11, Beijing 100044, China
| |
Collapse
|
8
|
Globa E, Zelinska N, Johnson MB, Flanagan SE, De Franco E. Neonatal and early-onset diabetes in Ukraine: Atypical features and mortality. Diabet Med 2023; 40:e15013. [PMID: 36398453 PMCID: PMC10946703 DOI: 10.1111/dme.15013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 10/27/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
AIMS The aim of this study is to elucidate the aetiology and clinical features of neonatal and early-onset diabetes in a large database for pediatric diabetes patients in Ukraine. METHODS We established a Pediatric Diabetes Register to identify patients diagnosed with diabetes before 9 months of age. Genetic testing was undertaken for 66 patients from 65 unrelated families with diabetes diagnosed within the first 6 months of life (neonatal diabetes, n = 36) or between 6 and 9 months (early-onset diabetes, n = 30). RESULTS We determined the genetic aetiology in 86.1% of patients (31/36) diagnosed before 6 months and in 20% (6/30) diagnosed between 6 and 9 months. Fourteen individuals (37.8% of those with a genetic cause identified) had activating heterozygous variants in ABCC8 or KCNJ11. An additional 10 individuals had pathogenic variants in the INS or GCK genes, while 4 had 6q24 transient neonatal diabetes. Rare genetic subtypes (including pathogenic variants in EIF2AK3, GLIS3, INSR, PDX1, LRBA, RFX6 and FOXP3) were identified in nine probands (24.3% of solved cases), 6 of whom died. In total, eight individuals died between infancy and childhood, all of them were diagnosed before 6 months and had received a genetic diagnosis. CONCLUSIONS In the last decade, the increased availability of comprehensive genetic testing has resulted in increased recognition of the contribution of rare genetic subtypes within pediatric diabetes cohorts. In our study, we identified a high mortality rate among these patients.
Collapse
Affiliation(s)
- Evgenia Globa
- Ukrainian Scientific and Practical Center of Endocrine SurgeryTransplantation of Endocrine Organs and Tissues of the Ministry of Health of UkraineKyivUkraine
| | - Nataliya Zelinska
- Ukrainian Scientific and Practical Center of Endocrine SurgeryTransplantation of Endocrine Organs and Tissues of the Ministry of Health of UkraineKyivUkraine
| | - Matthew B. Johnson
- Institute of Biomedical and Clinical Science, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Sarah E. Flanagan
- Institute of Biomedical and Clinical Science, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| |
Collapse
|
9
|
Sato Y, Isojima T, Takamiya K, Motoyama K, Enkai S, Ogawa E, Kodama H, Yorifuji T, Mimaki M. Longitudinal Glycaemic Profiles during Remission in 6q24-Related Transient Neonatal Diabetes Mellitus. Horm Res Paediatr 2022; 94:229-234. [PMID: 34348302 DOI: 10.1159/000518617] [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: 04/06/2021] [Accepted: 07/20/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Transient neonatal diabetes mellitus (TNDM) is a rare condition that is characterized by the presence of diabetes mellitus during the first 6 months of life and remission by 18 months of age. It usually relapses at a median age of 14 years. Hyperinsulinaemic hypoglycaemia is a relatively common complication during remission. Although β-cell function is reported to be impaired at relapse, the clinical course of glycaemic profiles during remission in patients with TNDM remains largely unknown. CASE PRESENTATION Longitudinal glycaemic profiles were investigated annually from remission (185 days) to relapse (14.5 years) in a patient with TNDM due to paternal 6q24 duplication using the oral glucose tolerance test (glucose intake: 1.75 g/kg to a maximum of 75 g). The patient's β-cell function and insulin sensitivity were assessed by calculating the insulinogenic index, homeostasis model assessment of β-cell function (HOMA-β), homeostasis model assessment of insulin resistance (HOMA-IR), quantitative insulin sensitivity check index, and Matsuda index. Early insulin response to glucose intake was impaired throughout remission, whereas fasting insulin and β-cell function by HOMA-β gradually increased in the first few years since remission, followed by a gradual decline in function. In contrast, HOMA-IR fluctuated and peaked at 6.5 years of age. CONCLUSION This is the first report of annual longitudinal glycaemic profiles in a patient with 6q24-related TNDM during remission. We identified fluctuations in β-cell function and insulin resistance during remission.
Collapse
Affiliation(s)
- Yasuhiro Sato
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Isojima
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Kiyomi Takamiya
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Kahoko Motoyama
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigehiro Enkai
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Eishin Ogawa
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan.,Minatocho Kodomo Clinic, Kawasaki, Japan
| | - Hiroko Kodama
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan.,Department of Dietetics, Faculty of Health and Medical Science, Teikyo Heisei University, Tokyo, Japan
| | - Tohru Yorifuji
- Division of Pediatric Endocrinology and Metabolism, Children's Medical Center, Osaka City General Hospital, Osaka, Japan
| | - Masakazu Mimaki
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Dhayalan B, Glidden MD, Zaykov AN, Chen YS, Yang Y, Phillips NB, Ismail-Beigi F, Jarosinski MA, DiMarchi RD, Weiss MA. Peptide Model of the Mutant Proinsulin Syndrome. I. Design and Clinical Correlation. Front Endocrinol (Lausanne) 2022; 13:821069. [PMID: 35299972 PMCID: PMC8922534 DOI: 10.3389/fendo.2022.821069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
Abstract
The mutant proinsulin syndrome is a monogenic cause of diabetes mellitus due to toxic misfolding of insulin's biosynthetic precursor. Also designated mutant INS-gene induced diabetes of the young (MIDY), this syndrome defines molecular determinants of foldability in the endoplasmic reticulum (ER) of β-cells. Here, we describe a peptide model of a key proinsulin folding intermediate and variants containing representative clinical mutations; the latter perturb invariant core sites in native proinsulin (LeuB15→Pro, LeuA16→Pro, and PheB24→Ser). The studies exploited a 49-residue single-chain synthetic precursor (designated DesDi), previously shown to optimize in vitro efficiency of disulfide pairing. Parent and variant peptides contain a single disulfide bridge (cystine B19-A20) to provide a model of proinsulin's first oxidative folding intermediate. The peptides were characterized by circular dichroism and redox stability in relation to effects of the mutations on (a) in vitro foldability of the corresponding insulin analogs and (b) ER stress induced in cell culture on expression of the corresponding variant proinsulins. Striking correlations were observed between peptide biophysical properties, degree of ER stress and age of diabetes onset (neonatal or adolescent). Our findings suggest that age of onset reflects the extent to which nascent structure is destabilized in proinsulin's putative folding nucleus. We envisage that such peptide models will enable high-resolution structural studies of key folding determinants and in turn permit molecular dissection of phenotype-genotype relationships in this monogenic diabetes syndrome. Our companion study (next article in this issue) employs two-dimensional heteronuclear NMR spectroscopy to define site-specific perturbations in the variant peptides.
Collapse
Affiliation(s)
- Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michael D. Glidden
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | | | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yanwu Yang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nelson B. Phillips
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Mark A. Jarosinski
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Michael A. Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
11
|
Yang Y, Glidden MD, Dhayalan B, Zaykov AN, Chen YS, Wickramasinghe NP, DiMarchi RD, Weiss MA. Peptide Model of the Mutant Proinsulin Syndrome. II. Nascent Structure and Biological Implications. Front Endocrinol (Lausanne) 2022; 13:821091. [PMID: 35299958 PMCID: PMC8922542 DOI: 10.3389/fendo.2022.821091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Toxic misfolding of proinsulin variants in β-cells defines a monogenic diabetes syndrome, designated mutant INS-gene induced diabetes of the young (MIDY). In our first study (previous article in this issue), we described a one-disulfide peptide model of a proinsulin folding intermediate and its use to study such variants. The mutations (LeuB15→Pro, LeuA16→Pro, and PheB24→Ser) probe residues conserved among vertebrate insulins. In this companion study, we describe 1H and 1H-13C NMR studies of the peptides; key NMR resonance assignments were verified by synthetic 13C-labeling. Parent spectra retain nativelike features in the neighborhood of the single disulfide bridge (cystine B19-A20), including secondary NMR chemical shifts and nonlocal nuclear Overhauser effects. This partial fold engages wild-type side chains LeuB15, LeuA16 and PheB24 at the nexus of nativelike α-helices α1 and α3 (as defined in native proinsulin) and flanking β-strand (residues B24-B26). The variant peptides exhibit successive structural perturbations in order: parent (most organized) > SerB24 >> ProA16 > ProB15 (least organized). The same order pertains to (a) overall α-helix content as probed by circular dichroism, (b) synthetic yields of corresponding three-disulfide insulin analogs, and (c) ER stress induced in cell culture by corresponding mutant proinsulins. These findings suggest that this and related peptide models will provide a general platform for classification of MIDY mutations based on molecular mechanisms by which nascent disulfide pairing is impaired. We propose that the syndrome's variable phenotypic spectrum-onsets ranging from the neonatal period to later in childhood or adolescence-reflects structural features of respective folding intermediates.
Collapse
Affiliation(s)
- Yanwu Yang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michael D. Glidden
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Yen-Shan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nalinda P. Wickramasinghe
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | | | - Michael A. Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
12
|
Lal RA, Moeller HP, Thomson EA, Horton TM, Lee S, Freeman R, Prahalad P, Poon ASY, Annes JP. Novel Pathogenic De Novo INS p.T97P Variant Presenting With Severe Neonatal DKA. Endocrinology 2022; 163:bqab246. [PMID: 34888628 PMCID: PMC9017997 DOI: 10.1210/endocr/bqab246] [Citation(s) in RCA: 2] [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: 08/12/2021] [Indexed: 11/19/2022]
Abstract
Pathogenic INS gene mutations are causative for mutant INS-gene-induced diabetes of youth (MIDY). We characterize a novel de novo heterozygous INS gene mutation (c.289A>C, p.T97P) that presented in an autoantibody-negative 5-month-old male infant with severe diabetic ketoacidosis. In silico pathogenicity prediction tools provided contradictory interpretations, while structural modeling indicated a deleterious effect on proinsulin folding. Transfection of wildtype and INS p.T97P expression and luciferase reporter constructs demonstrated elevated intracellular mutant proinsulin levels and dramatically impaired proinsulin/insulin and luciferase secretion. Notably, proteasome inhibition partially and selectively rescued INS p.T97P-derived luciferase secretion. Additionally, expression of INS p.T97P caused increased intracellular proinsulin aggregate formation and XBP-1s protein levels, consistent with induction of endoplasmic reticulum stress. We conclude that INS p.T97P is a newly identified pathogenic A-chain variant that is causative for MIDY via disruption of proinsulin folding and processing with induction of the endoplasmic reticulum stress response.
Collapse
Affiliation(s)
- Rayhan A Lal
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA, USA
- Division of Endocrinology, Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford, CA, USA
| | - Hannah P Moeller
- Stanford Diabetes Research Center, Stanford, CA, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Ella A Thomson
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Timothy M Horton
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Sooyeon Lee
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Raquel Freeman
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Priya Prahalad
- Division of Endocrinology, Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford, CA, USA
| | - Ada S Y Poon
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Justin P Annes
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford, CA, USA
| |
Collapse
|
13
|
Ikegwuonu C, Nweke S, Nnaji T, Oduneye O, Nwankwo G, Ogugua C, Orji ML, Ibekwe M. Sequence analysis of common gene mutation for neonatal diabetes: Three case series report of neonates with severe neonatal hyperglycaemia. NIGERIAN JOURNAL OF MEDICINE 2022. [DOI: 10.4103/njm.njm_29_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
14
|
Timmers M, Dirinck E, Lauwers P, Wuyts W, De Block C. ABCC8 variants in MODY12: Review of the literature and report of a case with severe complications. Diabetes Metab Res Rev 2021; 37:e3459. [PMID: 34014594 DOI: 10.1002/dmrr.3459] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 11/10/2022]
Abstract
More than 1000 variants of the ATP-binding cassette transporter subfamily C member 8 (ABCC8) gene have been reported in neonatal diabetes mellitus. Up to now only 55 ABCC8 variants were associated with Maturity-Onset Diabetes of the Young 12 (MODY12). We present a c.3544C>T p.(Arg1182Trp) ABCC8 variant in a 35-year-old women who had pronounced microvascular diabetic complications and a charcot arthropathy necessitating a lower limb amputation. The unusual severity of the disease course prompted us to perform a systematic review of all genetic variants in MODY12. The present mutation has mostly been associated with neonatal diabetes and in only three papers reporting a MODY12. The 55 MODY12 variants show a large clinical heterogeneity, even in relatives with the same mutation, ranging from mild impaired glucose tolerance to severe insulin-dependent diabetes mellitus. HbA1c at diagnosis ranged from 5% to 14% and age at diagnosis ranged from 2 to 53 years. However, several case reports lack documentation of diabetic complications. Hence, more detailed reports remain necessary to improve insight in MODY12 pathophysiology and outcome. In this article current data regarding therapeutic management are provided, and key points to consider for the individual patient affected by MODY12 are presented.
Collapse
Affiliation(s)
- Marijke Timmers
- Department of Endocrinology, Diabetology, and Metabolism, Antwerp University Hospital, Edegem, Belgium
| | - Eveline Dirinck
- Department of Endocrinology, Diabetology, and Metabolism, Antwerp University Hospital, Edegem, Belgium
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Patrick Lauwers
- Department of Vascular Surgery, Antwerp University Hospital, Edegem, Belgium
| | - Wim Wuyts
- Department of Medical Genetics, Antwerp University Hospital, Edegem, Belgium
| | - Christophe De Block
- Department of Endocrinology, Diabetology, and Metabolism, Antwerp University Hospital, Edegem, Belgium
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
15
|
Amirruddin NS, Tan WX, Tan YS, Gardner DSL, Bee YM, Verma CS, Hoon S, Lee KO, Teo AKK. Progressive endoplasmic reticulum stress over time due to human insulin gene mutation contributes to pancreatic beta cell dysfunction. Diabetologia 2021; 64:2534-2549. [PMID: 34448879 DOI: 10.1007/s00125-021-05530-3] [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: 12/11/2020] [Accepted: 05/14/2021] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS We studied the effects of heterozygous human INS gene mutations on insulin secretion, endoplasmic reticulum (ER) stress and other mechanisms in both MIN6 and human induced pluripotent stem cells (hiPSC)-derived beta-like cells, as well as the effects of prolonged overexpression of mutant human INS in MIN6 cells. METHODS We modelled the structure of mutant C109Y and G32V proinsulin computationally to examine the in silico effects. We then overexpressed either wild-type (WT), mutant (C109Y or G32V), or both WT and mutant human preproinsulin in MIN6 cells, both transiently and stably over several weeks. We measured the levels of human and rodent insulin secreted, and examined the transcript and protein levels of several ER stress and apoptotic markers. We also reprogrammed human donor fibroblasts heterozygous for the C109Y mutation into hiPSCs and differentiated these into pancreatic beta-like cells, which were subjected to single-cell RNA-sequencing and transcript and protein analyses for ER stress and apoptotic markers. RESULTS The computational modelling studies, and short-term and long-term expression studies in beta cells, revealed the presence of ER stress, organelle changes and insulin processing defects, resulting in a decreased amount of insulin secreted but not the ability to secrete insulin. By 9 weeks of expression of mutant human INS, dominant-negative effects of mutant INS were evident and beta cell insulin secretory capacity declined. INS+/C109Y patient-derived beta-like cells and single-cell RNA-sequencing analyses then revealed compensatory upregulation in genes involved in insulin secretion, processing and inflammatory response. CONCLUSIONS/INTERPRETATION The results provide deeper insights into the mechanisms of beta cell failure during INS mutation-mediated diabetes disease progression. Decreasing spliced X-box binding protein 1 (sXBP1) or inflammatory response could be avenues to restore the function of the remaining WT INS allele.
Collapse
Affiliation(s)
- Nur Shabrina Amirruddin
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Wei Xuan Tan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Yaw Sing Tan
- Bioinformatics Institute, A*STAR, Singapore, Republic of Singapore
| | - Daphne Su-Lyn Gardner
- Department of Endocrinology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Yong Mong Bee
- Department of Endocrinology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Chandra Shekhar Verma
- Bioinformatics Institute, A*STAR, Singapore, Republic of Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Republic of Singapore
| | - Shawn Hoon
- Molecular Engineering Laboratory, IMCB, A*STAR, Singapore, Republic of Singapore
| | - Kok Onn Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
16
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Ali Khan I. Do second generation sequencing techniques identify documented genetic markers for neonatal diabetes mellitus? Heliyon 2021; 7:e07903. [PMID: 34584998 PMCID: PMC8455689 DOI: 10.1016/j.heliyon.2021.e07903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 01/15/2021] [Accepted: 08/27/2021] [Indexed: 12/24/2022] Open
Abstract
Neonatal diabetes mellitus (NDM) is noted as a genetic, heterogeneous, and rare disease in infants. NDM occurs due to a single-gene mutation in neonates. A common source for developing NDM in an infant is the existence of mutations/variants in the KCNJ11 and ABCC8 genes, encoding the subunits of the voltage-dependent potassium channel. Both KCNJ11 and ABCC8 genes are useful in diagnosing monogenic diabetes during infancy. Genetic analysis was previously performed using first-generation sequencing techniques, such as DNA-Sanger sequencing, which uses chain-terminating inhibitors. Sanger sequencing has certain limitations; it can screen a limited region of exons in one gene, but it cannot screen large regions of the human genome. In the last decade, first generation sequencing techniques have been replaced with second-generation sequencing techniques, such as next-generation sequencing (NGS), which sequences nucleic-acids more rapidly and economically than Sanger sequencing. NGS applications are involved in whole exome sequencing (WES), whole genome sequencing (WGS), and targeted gene panels. WES characterizes a substantial breakthrough in human genetics. Genetic testing for custom genes allows the screening of the complete gene, including introns and exons. The aim of this review was to confirm if the 22 genetic variations previously documented to cause NDM by Sanger sequencing could be detected using second generation sequencing techniques. The author has cross-checked global studies performed in NDM using NGS, ES/WES, WGS, and targeted gene panels as second-generation sequencing techniques; WES confirmed the similar variants, which have been previously documented with Sanger sequencing. WES is documented as a powerful tool and WGS as the most comprehensive test for verified the documented variants, as well as novel enhancers. This review recommends for the future studies should be performed with second generation sequencing techniques to identify the verified 22 genetic and novel variants by screening in NDM (PNDM or TNMD) children.
Collapse
Affiliation(s)
- Imran Ali Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, PO Box-10219, Riyadh, 11433, Saudi Arabia
| |
Collapse
|
18
|
Abstract
OBJECTIVE To describe the spectrum of neonatal diabetes mellitus (NDM), document new mutations, and review published Indian literature on the etiology of NDM. METHODS Retrospective analysis of the clinical and genetic profile of 12 NDM patients. RESULTS Eight patients presented with NDM before the age of 6 mo. Three other patients, including 2 siblings presented in later part of infancy. An additional patient was diagnosed at age 5 y with the same etiology as her infant sibling. Four patients had transient diabetes [TNDM:1 each with a mutation in KCNJ11 and INS gene, 2 with ABCC8 mutation], 7 had permanent diabetes [PNDM: 2 siblings with complete glucokinase deficiency, 2 siblings with thiamine responsive megaloblastic anemia (TRMA), 1 with Immune dysregulation, Polyendocrinopathy, Enteropathy, X-linked (IPEX) syndrome and 2 with Wolcott Rallison syndrome, (WRS)]. Four patients had 5 novel mutations. Genetic etiology could not be established in 1 patient with features of insulin resistance. Poorly controlled blood glucose in the TRMA patient led to hyperglycemia-induced hemichorea-hemiballismus, a rare manifestation in children. CONCLUSIONS The authors describe 5 novel mutations, in the EIF2AK3, ABCC8, and GCK genes, a homozygous mutation at the ABCC8 locus presenting as TNDM, an obscure phenotype of the GCK gene mutation, and hyperglycemia-induced hemichorea-hemiballismus in a patient with TRMA. In India, PNDM is most commonly due to WRS similar to Middle Eastern countries with high consanguinity rates.
Collapse
|
19
|
Adler A, Bennett P, Colagiuri Chair S, Gregg E, Narayan KMV, Inês Schmidt M, Sobngwi E, Tajima N, Tandon N, Unwin N, Wild S, Yudkin J, Levitt N, Mohan V, Montgomery S, Nyirenda MJ, Tuomilehto J, Den Boon S, Hocking S. REPRINT OF: CLASSIFICATION OF DIABETES MELLITUS. Diabetes Res Clin Pract 2021:108972. [PMID: 34343595 DOI: 10.1016/j.diabres.2021.108972] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Executive Summary This document updates the 1999 World Health Organization (WHO) classification of diabetes. It prioritizes clinical care and guides health professionals in choosing appropriate treatments at the time of diabetes diagnosis, and provides practical guidance to clinicians in assigning a type of diabetes to individuals at the time of diagnosis. It is a compromise between clinical and aetiological classification because there remain gaps in knowledge of the aetiology and pathophysiology of diabetes. While acknowledging the progress that is being made towards a more precise categorization of diabetes subtypes, the aim of this document is to recommend a classification that is feasible to implement in different settings throughout the world. The revised classification is presented in Table 1. Unlike the previous classification, this classification does not recognize subtypes of type 1 diabetes and type 2 diabetes and includes new types of diabetes ("hybrid types of diabetes" and "unclassified diabetes").
Collapse
Affiliation(s)
| | - Peter Bennett
- Phoenix Epidemiology & Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, USA
| | | | - Edward Gregg
- Centers for Disease Control and Prevention, Atlanta, USA
| | | | | | - Eugene Sobngwi
- Faculté de Medecine et des Sciences Biomedicales et Centre de Biotechnologie, Université de Yaounde 1, Cameroon
| | - Naoko Tajima
- Jikei University School of Medicine, Tokyo, Japan
| | - Nikhil Tandon
- All India Institute of Medical Sciences, New Delhi, India
| | - Nigel Unwin
- Chronic Disease Research Centre, The University of the West Indies, Bridgetown, Barbados, and MRC Epidemiology Unit, University of Cambridge, UK
| | | | | | - Naomi Levitt
- Diabetic Medicine and Endocrinology, Department of Medicine at Groote Schuur Hospital and University of Cape Town, South Africa
| | | | | | - Moffat J Nyirenda
- Medical Research Council/Uganda Virus Research Institute/London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | | | | | | |
Collapse
|
20
|
Eksi YE, Sanlioglu AD, Akkaya B, Ozturk BE, Sanlioglu S. Genome engineering and disease modeling via programmable nucleases for insulin gene therapy: Promises of CRISPR/Cas9 technology. World J Stem Cells 2021; 13:485-502. [PMID: 34249224 PMCID: PMC8246254 DOI: 10.4252/wjsc.v13.i6.485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/02/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Targeted genome editing is a continually evolving technology employing programmable nucleases to specifically change, insert, or remove a genomic sequence of interest. These advanced molecular tools include meganucleases, zinc finger nucleases, transcription activator-like effector nucleases and RNA-guided engineered nucleases (RGENs), which create double-strand breaks at specific target sites in the genome, and repair DNA either by homologous recombination in the presence of donor DNA or via the error-prone non-homologous end-joining mechanism. A recently discovered group of RGENs known as CRISPR/Cas9 gene-editing systems allowed precise genome manipulation revealing a causal association between disease genotype and phenotype, without the need for the reengineering of the specific enzyme when targeting different sequences. CRISPR/Cas9 has been successfully employed as an ex vivo gene-editing tool in embryonic stem cells and patient-derived stem cells to understand pancreatic beta-cell development and function. RNA-guided nucleases also open the way for the generation of novel animal models for diabetes and allow testing the efficiency of various therapeutic approaches in diabetes, as summarized and exemplified in this manuscript.
Collapse
Affiliation(s)
- Yunus E Eksi
- Department of Gene and Cell Therapy, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Ahter D Sanlioglu
- Department of Gene and Cell Therapy, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Bahar Akkaya
- Department of Gene and Cell Therapy, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Bilge Esin Ozturk
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Salih Sanlioglu
- Department of Gene and Cell Therapy, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| |
Collapse
|
21
|
Kochar IS, Ramachandran S. Empirical sulphonylurea in neonatal diabetes: results from a tertiary care center. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-021-00959-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
22
|
Abdelalim EM. Modeling different types of diabetes using human pluripotent stem cells. Cell Mol Life Sci 2021; 78:2459-2483. [PMID: 33242105 PMCID: PMC11072720 DOI: 10.1007/s00018-020-03710-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/19/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022]
Abstract
Diabetes mellitus (DM) is a metabolic disease characterized by chronic hyperglycemia as a result of progressive loss of pancreatic β cells, which could lead to several debilitating complications. Different paths, triggered by several genetic and environmental factors, lead to the loss of pancreatic β cells and/or function. Understanding these many paths to β cell damage or dysfunction could help in identifying therapeutic approaches specific for each path. Most of our knowledge about diabetes pathophysiology has been obtained from studies on animal models, which do not fully recapitulate human diabetes phenotypes. Currently, human pluripotent stem cell (hPSC) technology is a powerful tool for generating in vitro human models, which could provide key information about the disease pathogenesis and provide cells for personalized therapies. The recent progress in generating functional hPSC-derived β cells in combination with the rapid development in genomic and genome-editing technologies offer multiple options to understand the cellular and molecular mechanisms underlying the development of different types of diabetes. Recently, several in vitro hPSC-based strategies have been used for studying monogenic and polygenic forms of diabetes. This review summarizes the current knowledge about different hPSC-based diabetes models and how these models improved our current understanding of the pathophysiology of distinct forms of diabetes. Also, it highlights the progress in generating functional β cells in vitro, and discusses the current challenges and future perspectives related to the use of the in vitro hPSC-based strategies.
Collapse
Affiliation(s)
- Essam M Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha, Qatar.
| |
Collapse
|
23
|
Öngen YD, Eren E, Demirbaş Ö, Sobu E, Ellard S, De Franco E, Tarım Ö. Genotype and Phenotype Heterogeneity in Neonatal Diabetes: A Single Centre Experience in Turkey. J Clin Res Pediatr Endocrinol 2021; 13:80-87. [PMID: 32820876 PMCID: PMC7947723 DOI: 10.4274/jcrpe.galenos.2020.2020.0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Neonatal diabetes mellitus (NDM) may be transient or permanent, and the majority is caused by genetic mutations. Early diagnosis is essential to select the patients who will respond to oral treatment. In this investigation, we aimed to present the phenotype and genotype of our patients with NDM and share our experience in a single tertiary center METHODS A total of 16 NDM patients from 12 unrelated families are included in the study. The clinical presentation, age at diagnosis, perinatal and family history, consanguinity, gender, hemoglobin A1c, C-peptide, insulin, insulin autoantibodies, genetic mutations, and response to treatment are retrospectively evaluated. RESULTS The median age at diagnosis of diabetes was five months (4 days-18 months) although six patients with a confirmed genetic diagnosis were diagnosed >6 months. Three patients had KCNJ11 mutations, six had ABCC8 mutations, three had EIF2AK3 mutations, and one had a de novo INS mutation. All the permanent NDM patients with KCNJ11 and ABCC8 mutations were started on sulfonylurea treatment resulting in a significant increase in C-peptide level, better glycemic control, and discontinuation of insulin. CONCLUSION Although NDM is defined as diabetes diagnosed during the first six months of life, and a diagnosis of type 1 diabetes is more common between the ages of 6 and 24 months, in rare cases NDM may present as late as 12 or even 24 months of age. Molecular diagnosis in NDM is important for planning treatment and predicting prognosis. Therefore, genetic testing is essential in these patients.
Collapse
Affiliation(s)
- Yasemin Denkboy Öngen
- Bursa Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology, Bursa, Turkey,* Address for Correspondence: Bursa Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology, Bursa, Turkey Phone: +90 224 295 05 33 E-mail:
| | - Erdal Eren
- Bursa Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology, Bursa, Turkey
| | - Özgecan Demirbaş
- Bursa Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology, Bursa, Turkey
| | - Elif Sobu
- Bursa Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology, Bursa, Turkey
| | - Sian Ellard
- University of Exeter Medical School, Institute of Biomedical and Clinical Science, Exeter, United Kingdom,Royal Devon and Exeter NHS Foundation Trust, Genomics Laboratory, Exeter, United Kingdom
| | - Elisa De Franco
- University of Exeter Medical School, Institute of Biomedical and Clinical Science, Exeter, United Kingdom
| | - Ömer Tarım
- Bursa Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology, Bursa, Turkey
| |
Collapse
|
24
|
Kim MS, Kim SE, Lee NY, Kim SK, Kim SH, Cho WK, Cho KS, Jung MH, Suh BK, Ahn MB. Transient Neonatal Diabetes Mellitus Managed with Continuous Subcutaneous Insulin Infusion (CSII) and Continuous Glucose Monitoring. NEONATAL MEDICINE 2021. [DOI: 10.5385/nm.2021.28.1.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
25
|
Zhang H, Colclough K, Gloyn AL, Pollin TI. Monogenic diabetes: a gateway to precision medicine in diabetes. J Clin Invest 2021; 131:142244. [PMID: 33529164 PMCID: PMC7843214 DOI: 10.1172/jci142244] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Monogenic diabetes refers to diabetes mellitus (DM) caused by a mutation in a single gene and accounts for approximately 1%-5% of diabetes. Correct diagnosis is clinically critical for certain types of monogenic diabetes, since the appropriate treatment is determined by the etiology of the disease (e.g., oral sulfonylurea treatment of HNF1A/HNF4A-diabetes vs. insulin injections in type 1 diabetes). However, achieving a correct diagnosis requires genetic testing, and the overlapping of the clinical features of monogenic diabetes with those of type 1 and type 2 diabetes has frequently led to misdiagnosis. Improvements in sequencing technology are increasing opportunities to diagnose monogenic diabetes, but challenges remain. In this Review, we describe the types of monogenic diabetes, including common and uncommon types of maturity-onset diabetes of the young, multiple causes of neonatal DM, and syndromic diabetes such as Wolfram syndrome and lipodystrophy. We also review methods of prioritizing patients undergoing genetic testing, and highlight existing challenges facing sequence data interpretation that can be addressed by forming collaborations of expertise and by pooling cases.
Collapse
Affiliation(s)
- Haichen Zhang
- University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| | - Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Anna L. Gloyn
- Department of Pediatrics, Division of Endocrinology, and,Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, California, USA
| | - Toni I. Pollin
- University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Gopi S, Kavitha B, Kanthimathi S, Kannan A, Kumar R, Joshi R, Kanodia S, Arya AD, Pendsey S, Pendsey S, Raghupathy P, Mohan V, Radha V. Genotype-phenotype correlation of K ATP channel gene defects causing permanent neonatal diabetes in Indian patients. Pediatr Diabetes 2021; 22:82-92. [PMID: 32893419 DOI: 10.1111/pedi.13109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 08/06/2020] [Accepted: 08/23/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND There are very few reports pertaining to Indian patients with neonatal diabetes mellitus (NDM). Activating or gain of function mutations of KATP channel genes namely KCNJ11 and ABCC8 are most predominant cause of permanent neonatal diabetes mellitus (PNDM). OBJECTIVES To identify the genotype-phenotype correlation of KATP channel gene defects in a large series of (n = 181) Indian PNDM patients. METHODS Direct sequencing of all exons of KCNJ11 and ABCC8 genes in all 181 patients with PNDM were performed. Clinical and biochemical data were collected. RESULTS We have identified the molecular basis of KATP -NDM in 39 out of 181 patients (22%). Of these, 20 had KCNJ11 mutations and 19 had ABCC8 mutations, thus comprising 51% of KCNJ11 and 49% of ABCC8. There were four novel mutations (D1128Tfs*16, Y1287C, S1422T, and H1537R) in ABCC8 gene. Three patients with KCNJ11 mutations had developmental delay with DEND syndrome. In patients with ABCC8 mutations developmental delay was seen in seven out of 19 (36.8%). Of this, three patients (15.7%) had DEND phenotype and four (21%) had iDEND. Of the 39 patients, 33 (84%) patients were shifted to sulfonylurea therapy (glibenclamide). Of this, 19(57.5%) patients harbored KCNJ11 mutations and 14(42.1%) ABCC8 mutations. CONCLUSIONS This is the first largest study in NDM patients in India demonstrating the importance of KATP channel gene mutation screening in PNDM and efficacy of glibenclamide for Indian patients with KATP -PNDM. The success rate of transfer is more in patients with KCNJ11 mutations compared with those with ABCC8 mutations.
Collapse
Affiliation(s)
- Sundaramoorthy Gopi
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes, Affiliated to University of Madras, Chennai, India
| | - Babu Kavitha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes, Affiliated to University of Madras, Chennai, India
| | - Sekar Kanthimathi
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes, Affiliated to University of Madras, Chennai, India
| | - Alagarsamy Kannan
- Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - Rakesh Kumar
- Advanced Paediatrics Centre, Post-Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Rajesh Joshi
- Division of Paediatric Endocrinology, B.J. Wadia Hospital for Children, Mumbai, India
| | | | - Archana Dayal Arya
- Institute of Child Health, Sir Ganga Ram Hospital Marg, New Delhi, India
| | | | | | | | - Viswanathan Mohan
- Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India.,Department of Diabetology, Madras Diabetes Research Foundation, Chennai, India
| | - Venkatesan Radha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes, Affiliated to University of Madras, Chennai, India
| |
Collapse
|
27
|
Ngoc CTB, Dien TM, De Franco E, Ellard S, Houghton JAL, Lan NN, Thao BP, Khanh NN, Flanagan SE, Craig ME, Dung VC. Molecular Genetics, Clinical Characteristics, and Treatment Outcomes of K ATP-Channel Neonatal Diabetes Mellitus in Vietnam National Children's Hospital. Front Endocrinol (Lausanne) 2021; 12:727083. [PMID: 34566892 PMCID: PMC8458931 DOI: 10.3389/fendo.2021.727083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Neonatal diabetes mellitus (NDM) is defined as insulin-requiring persistent hyperglycemia occurring within the first 6 months of life, which can result from mutations in at least 25 different genes. Activating heterozygous mutations in genes encoding either of the subunits of the ATP-sensitive K+ channel (KATP channel; KCNJ11 or ABCC8) of the pancreatic beta cell are the most common cause of permanent NDM and the second most common cause of transient NDM. Patients with NDM caused by KATP channel mutations are sensitive to sulfonylurea (SU) treatment; therefore, their clinical management can be improved by replacing insulin with oral agents. PATIENTS AND METHODS Seventy patients were diagnosed with NDM between May 2008 and May 2021 at Vietnam National Children's Hospital, and molecular genetic testing for all genes known to cause NDM was performed at the Exeter Genomic Laboratory, UK. Patients with ABCC8 or KCNJ11 mutations were transferred from insulin to oral SU. Clinical characteristics, molecular genetics, and annual data relating to glycemic control, SU dose, severe hypoglycemia, and side effects were collected. The main outcomes of interest were SU dose, SU failure (defined as permanent reintroduction of daily insulin), and glycemic control (HbA1c). RESULTS Fifty-four of 70 patients (77%) with NDM harbored a genetic mutation and of these; 27 (50%) had activating heterozygous mutations in ABCC8 or KCNJ11. A total of 21 pathogenic mutations were identified in the 27 patients, including 13 mutations in ABCC8 and 8 mutations in KCNJ11. Overall, 51% had low birth weight (below 3rd percentile), 23 (85%) were diagnosed before 3 months of age, and 23 (85%) presented with diabetic ketoacidosis. At diagnosis, clinical and biochemical findings (mean ± SD) were pH 7.16 ± 0.16; HCO3- , 7.9 ± 7.4 mmol/L; BE, -17.9 ± 9.1 mmol/L; HbA1C, 7.98% ± 2.93%; blood glucose, 36.2 ± 12.3 mmol/L; and C-peptide median, 0.09 (range, 0-1.61 nmol/l). Twenty-six patients were successfully transferred from insulin to SU therapy. In the remaining case, remission of diabetes occurred prior to transfer. Glycemic control on SU treatment was better than on insulin treatment: HbA1c and blood glucose level decreased from 7.58% ± 4.63% and 19.04 ± 14.09 mmol/L when treated with insulin to 5.8 ± 0.94% and 6.87 ± 3.46 mmol/L when treated with SU, respectively. CONCLUSIONS This is the first case series of NDM patients with ABCC8/KCNJ11 mutations reported in Vietnam. SU is safe in the short term for these patients and more effective than insulin therapy, consistent with all studies to date. This is relevant for populations where access to and cost of insulin are problematic, reinforcing the importance of genetic testing for NDM.
Collapse
Affiliation(s)
- Can Thi Bich Ngoc
- Department of Endocrinology, Metabolism and Genetics, Vietnam National Children’s Hospital, Hanoi, Vietnam
| | - Tran Minh Dien
- Department of Endocrinology, Metabolism and Genetics, Vietnam National Children’s Hospital, Hanoi, Vietnam
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Sian Ellard
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Jayne A. L. Houghton
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Nguyen Ngoc Lan
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Bui Phuong Thao
- Department of Endocrinology, Metabolism and Genetics, Vietnam National Children’s Hospital, Hanoi, Vietnam
| | - Nguyen Ngoc Khanh
- Department of Endocrinology, Metabolism and Genetics, Vietnam National Children’s Hospital, Hanoi, Vietnam
| | - Sarah E. Flanagan
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Maria E. Craig
- Institute of Endocrinology and Diabetes, The Children’s Hospital at Westmead/Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW, Australia
- School of Women’s and Children’s Health, University of New South Wales Medicine, Sydney, NSW, Australia
| | - Vu Chi Dung
- Department of Endocrinology, Metabolism and Genetics, Vietnam National Children’s Hospital, Hanoi, Vietnam
- *Correspondence: Vu Chi Dung,
| |
Collapse
|
28
|
Abstract
Neonatal diabetes (ND) appears during the first months of life and is caused by a single gene mutation. It is heterogenous and very different compared to other forms of multi-factorial or polygenic diabetes. Clinically, this form is extremely severe, however, early genetic diagnosis is pivotal for successful therapy. A large palette of genes is demonstrated to be a cause of ND, however, the mechanisms of permanent hyperglycemia are different. This review will give an overview of more frequent genetic mutations causing ND, including the function of the mutated genes and the specific therapy for certain sub-forms.
Collapse
Affiliation(s)
- M Kocova
- Medical Faculty, University Cyril and Methodius, Skopje, Republic of Macedonia
| |
Collapse
|
29
|
Abstract
Diabetes mellitus is a chronic heterogeneous metabolic disorder with complex pathogenesis. It is characterized by elevated blood glucose levels or hyperglycemia, which results from abnormalities in either insulin secretion or insulin action or both. Hyperglycemia manifests in various forms with a varied presentation and results in carbohydrate, fat, and protein metabolic dysfunctions. Long-term hyperglycemia often leads to various microvascular and macrovascular diabetic complications, which are mainly responsible for diabetes-associated morbidity and mortality. Hyperglycemia serves as the primary biomarker for the diagnosis of diabetes as well. In this review, we would be focusing on the classification of diabetes and its pathophysiology including that of its various types.
Collapse
Affiliation(s)
- Mujeeb Z Banday
- Department of Biochemistry, Government Medical College and Associated Shri Maharaja Hari Singh Hospital, Srinagar, Kashmir, India
| | - Aga S Sameer
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdul Aziz University for Health Sciences, King Abdullah International Medical Research Centre, National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Saniya Nissar
- Department of Biochemistry, Government Medical College and Associated Shri Maharaja Hari Singh Hospital, Srinagar, Kashmir, India
| |
Collapse
|
30
|
Jennings RE, Scharfmann R, Staels W. Transcription factors that shape the mammalian pancreas. Diabetologia 2020; 63:1974-1980. [PMID: 32894307 PMCID: PMC7476910 DOI: 10.1007/s00125-020-05161-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022]
Abstract
Improving our understanding of mammalian pancreas development is crucial for the development of more effective cellular therapies for diabetes. Most of what we know about mammalian pancreas development stems from mouse genetics. We have learnt that a unique set of transcription factors controls endocrine and exocrine cell differentiation. Transgenic mouse models have been instrumental in studying the function of these transcription factors. Mouse and human pancreas development are very similar in many respects, but the devil is in the detail. To unravel human pancreas development in greater detail, in vitro cellular models (including directed differentiation of stem cells, human beta cell lines and human pancreatic organoids) are used; however, in vivo validation of these results is still needed. The current best 'model' for studying human pancreas development are individuals with monogenic forms of diabetes. In this review, we discuss mammalian pancreas development, highlight some discrepancies between mouse and human, and discuss selected transcription factors that, when mutated, cause permanent neonatal diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Rachel E Jennings
- Division of Diabetes, Endocrinology & Gastroenterology, Faculty of Biology, Medicine & Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK.
- Endocrinology Department, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Raphael Scharfmann
- Institut Cochin, INSERM, U1016, CNRS, UMR8104, Université de Paris, 75014, Paris, France.
| | - Willem Staels
- Institut Cochin, INSERM, U1016, CNRS, UMR8104, Université de Paris, 75014, Paris, France.
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Department of Pediatrics, Division of Pediatric Endocrinology, University Hospital of Brussels, Jette, Belgium.
| |
Collapse
|
31
|
Athanassaki I, Lyons S, DeSalvo D, Hendrix K, Ebenezer S, McCann-Crosby B, Redondo M, Chen MJ, Uysal S, Sisley S. Diabetes Curriculum for Pediatric Endocrine Fellowship Utilizing Modified Team-Based Learning. MEDEDPORTAL : THE JOURNAL OF TEACHING AND LEARNING RESOURCES 2020; 16:10948. [PMID: 32821810 PMCID: PMC7431187 DOI: 10.15766/mep_2374-8265.10948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 01/03/2020] [Indexed: 06/11/2023]
Abstract
INTRODUCTION While type 1 diabetes is frequently encountered clinically in pediatric endocrinology fellowship training, other types of diabetes may only be encountered in educational settings. Adult learners learn best through knowledge application, but to date there are no published curricula utilizing application educational strategies for all forms of diabetes. METHODS We utilized a team-based learning (TBL) approach to create four modules on different types of diabetes: type 1 diabetes, type 2 diabetes, neonatal diabetes, and maturity-onset diabetes of the young. We divided our fellows (all training years, n = 11) into two teams and delivered four separate, 90-minute sessions. To emphasize the application of knowledge, we modified the format to combine the readiness assurance test (RAT) with application problem (APP) questions. The combined RAT/APP questions were answered by individuals and teams. We analyzed scores from individual and team tests and evaluated each module. Additionally, we acquired subjective data from the fellows regarding their experiences. RESULTS Teams outperformed individuals on the tests, as expected (94% vs. 76% correct questions, respectively). All the fellows agreed that the sessions should be included permanently. Additionally, all agreed the sessions helped them apply knowledge. Subjectively, the fellows were very engaged and lively during the sessions and felt the sessions were feasible as implemented. DISCUSSION TBL can be a valuable educational strategy to increase the application of knowledge for diabetes in pediatric endocrinology fellows. Future studies examining the use of this strategy to increase critical thinking skills and knowledge retention in the long-term would be useful.
Collapse
Affiliation(s)
- Ioanna Athanassaki
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| | - Sarah Lyons
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| | - Daniel DeSalvo
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| | | | - Sophia Ebenezer
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| | | | - Maria Redondo
- Associate Professor, Department of Pediatrics, Baylor College of Medicine
| | - Min-Jye Chen
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| | - Serife Uysal
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| | - Stephanie Sisley
- Assistant Professor, Department of Pediatrics, Baylor College of Medicine
| |
Collapse
|
32
|
Sakano D, Inoue A, Enomoto T, Imasaka M, Okada S, Yokota M, Koike M, Araki K, Kume S. Insulin2 Q104del (Kuma) mutant mice develop diabetes with dominant inheritance. Sci Rep 2020; 10:12187. [PMID: 32699230 PMCID: PMC7376009 DOI: 10.1038/s41598-020-68987-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/06/2020] [Indexed: 11/18/2022] Open
Abstract
Insulin gene mutations have been identified to cause monogenic diabetes, and most of which developed permanent neonatal diabetes at young ages before 6 months of age in humans. To establish an animal model of permanent diabetes, we performed genome editing using the CRISPR/Cas9 system. We generated a novel Kuma mutant mice with p.Q104del in the Insulin2 (Ins2) gene in a BRJ background that exhibits a severe immune deficiency. Kuma mutant mice are non-obese and developed hyperglycemia from 3 weeks after birth in both males and females, which are inherited in a dominant mode. Kuma mutant mice displayed reduced insulin protein levels from 3-weeks-old, which seem to be caused by the low stability of the mutant insulin protein. Kuma mutant showed a reduction in islet size and islet mass. Electron microscopic analysis revealed a marked decrease in the number and size of insulin granules in the beta-cells of the mutant mice. Hyperglycemia of the mutant can be rescued by insulin administration. Our results present a novel insulin mutation that causes permanent early-onset diabetes, which provides a model useful for islet transplantation studies.
Collapse
Affiliation(s)
- Daisuke Sakano
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Airi Inoue
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Takayuki Enomoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Mai Imasaka
- Laboratory of Developmental Genetics, Institute of Resource Development and Analysis, Chuo-Ku, Honjo 2-2-1, Kumamoto, 860-0811, Japan.,Department of Genetics, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Retroviral Infection, Kumamoto University, Honjo 2-2-1, Kumamoto, 860-0811, Japan
| | - Mutsumi Yokota
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kimi Araki
- Laboratory of Developmental Genetics, Institute of Resource Development and Analysis, Chuo-Ku, Honjo 2-2-1, Kumamoto, 860-0811, Japan.
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan.
| |
Collapse
|
33
|
Law NC, Marinelli I, Bertram R, Corbin KL, Schildmeyer C, Nunemaker CS. Chronic stimulation induces adaptive potassium channel activity that restores calcium oscillations in pancreatic islets in vitro. Am J Physiol Endocrinol Metab 2020; 318:E554-E563. [PMID: 32069073 PMCID: PMC7191410 DOI: 10.1152/ajpendo.00482.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin pulsatility is important to hepatic response in regulating blood glucose. Growing evidence suggests that insulin-secreting pancreatic β-cells can adapt to chronic disruptions of pulsatility to rescue this physiologically important behavior. We determined the time scale for adaptation and examined potential ion channels underlying it. We induced the adaptation both by chronic application of the ATP-sensitive K+ [K(ATP)] channel blocker tolbutamide and by application of the depolarizing agent potassium chloride (KCl). Acute application of tolbutamide without pretreatment results in elevated Ca2+ as measured by fura-2AM and the loss of endogenous pulsatility. We show that after chronic exposure to tolbutamide (12-24 h), Ca2+ oscillations occur with subsequent acute tolbutamide application. The same experiment was conducted with potassium chloride (KCl) to directly depolarize the β-cells. Once again, following chronic exposure to the cell stimulator, the islets produced Ca2+ oscillations when subsequently exposed to tolbutamide. These experiments suggest that it is the chronic stimulation, and not tolbutamide desensitization, that is responsible for the adaptation that rescues oscillatory β-cell activity. This compensatory response also causes islet glucose sensitivity to shift rightward following chronic tolbutamide treatment. Mathematical modeling shows that a small increase in the number of K(ATP) channels in the membrane is one adaptation mechanism that is compatible with the data. To examine other compensatory mechanisms, pharmacological studies provide support that Kir2.1 and TEA-sensitive channels play some role. Overall, this investigation demonstrates β-cell adaptability to overstimulation, which is likely an important mechanism for maintaining glucose homeostasis in the face of chronic stimulation.
Collapse
Affiliation(s)
- Nathan C Law
- Department of Biomedical Sciences Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | | | - Richard Bertram
- Department of Mathematics and Programs in Neuroscience and Molecular Biophysics, Florida State University, Tallahassee, Florida
| | - Kathryn L Corbin
- Department of Biomedical Sciences Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Cara Schildmeyer
- Department of Biomedical Sciences Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Craig S Nunemaker
- Department of Biomedical Sciences Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| |
Collapse
|
34
|
Otto WR, Orr S, Boyd JW. Case 4: Vomiting and Tachypnea in a 9-week-old Girl. Pediatr Rev 2019; 40:599-601. [PMID: 31676535 DOI: 10.1542/pir.2017-0254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
| | - Sarah Orr
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS
| | - Jennifer W Boyd
- Department of Pediatrics and.,Division of Endocrinology, Children's Mercy Hospital, Kansas City, MO
| |
Collapse
|
35
|
García-Carmona L, Martín A, Sempionatto JR, Moreto JR, González MC, Wang J, Escarpa A. Pacifier Biosensor: Toward Noninvasive Saliva Biomarker Monitoring. Anal Chem 2019; 91:13883-13891. [DOI: 10.1021/acs.analchem.9b03379] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Laura García-Carmona
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Faculty of Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain
| | - Aida Martín
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Juliane R. Sempionatto
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Jose R. Moreto
- Department of Aerospace Engineering, San Diego State University, San Diego, San Diego, California 92182, United States
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California 92093, United States
| | - María Cristina González
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Faculty of Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain
- Chemical Research Institute “Andrés M. del Río”, University of Alcalá, Alcalá de Henares, E-28871 Madrid, Spain
| | - Joseph Wang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Alberto Escarpa
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Faculty of Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain
- Chemical Research Institute “Andrés M. del Río”, University of Alcalá, Alcalá de Henares, E-28871 Madrid, Spain
| |
Collapse
|
36
|
Gaisl O, Konrad D, Joset P, Lang-Muritano M. A novel GATA6 variant in a boy with neonatal diabetes and diaphragmatic hernia: a familial case with a review of the literature. J Pediatr Endocrinol Metab 2019; 32:1027-1030. [PMID: 31271559 DOI: 10.1515/jpem-2019-0057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/18/2019] [Indexed: 11/15/2022]
Abstract
GATA6 gene variants come along with possible features such as pancreas agenesis/hypoplasia, neonatal diabetes and congenital heart defect. Congenital hypothyroidism, and hepatobiliary and gut abnormalities are also detectable. Children with congenital heart defects and neonatal diabetes were already described in 1970. GATA6 variants can be due to de novo variants or due to inherited variants. To date, 11 cases due to an inherited variant have been described. Herein we present a novel heterozygous GATA6 variant (c.1291C > T p.[Gln431*]) in a boy with transient neonatal diabetes, diaphragmatic hernia, congenital heart defect and early-onset scoliosis. The same variant was also present in the mother. At the age of 3 years, a random evaluation revealed a hemoglobin A1c (HbA1c) level of 7.8% (62 mmol/mol) without any diabetes-related symptoms. He was started on insulin therapy and HbA1c normalized. A short review of the literature of hereditary cases of the GATA6 variant revealed the variable phenotypic spectrum and showed that patients with a mild phenotype are likely to have children with a more severe phenotype.
Collapse
Affiliation(s)
- Odile Gaisl
- Department of Pediatric Endocrinology and Diabetology, and Children's Research Centre, University Children's Hospital, Steinwiesstrasse 75, 8032Zürich, Switzerland, Phone: +0041 44 266 84 23, Fax: +0041 44 266 79 83
| | - Daniel Konrad
- Department of Pediatric Endocrinology and Diabetology, and Children's Research Centre, University Children's Hospital, Zürich, Switzerland
| | - Pascal Joset
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Mariarosaria Lang-Muritano
- Department of Pediatric Endocrinology and Diabetology, and Children's Research Centre, University Children's Hospital, Zürich, Switzerland
| |
Collapse
|
37
|
Al-Khawaga S, Mohammed I, Saraswathi S, Haris B, Hasnah R, Saeed A, Almabrazi H, Syed N, Jithesh P, El Awwa A, Khalifa A, AlKhalaf F, Petrovski G, Abdelalim EM, Hussain K. The clinical and genetic characteristics of permanent neonatal diabetes (PNDM) in the state of Qatar. Mol Genet Genomic Med 2019; 7:e00753. [PMID: 31441606 PMCID: PMC6785445 DOI: 10.1002/mgg3.753] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/04/2019] [Accepted: 04/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal diabetes mellitus (NDM) is a rare condition that occurs within the first six months of life. Permanent NDM (PNDM) is caused by mutations in specific genes that are known for their expression at early and/or late stages of pancreatic beta‐ cell development, and are either involved in beta‐cell survival, insulin processing, regulation, and release. The native population in Qatar continues to practice consanguineous marriages that lead to a high level of homozygosity. To our knowledge, there is no previous report on the genomics of NDM among the Qatari population. The aims of the current study are to identify patients with NDM diagnosed between 2001 and 2016, and examine their clinical and genetic characteristics. Methods To calculate the incidence of PNDM, all patients with PNDM diagnosed between 2001 and 2016 were compared to the total number of live births over the 16‐year‐period. Whole Genome Sequencing (WGS) was used to investigate the genetic etiology in the PNDM cohort. Results PNDM was diagnosed in nine (n = 9) patients with an estimated incidence rate of 1:22,938 live births among the indigenous Qatari. Seven different mutations in six genes (PTF1A, GCK, SLC2A2, EIF2AK3, INS, and HNF1B) were identified. In the majority of cases, the genetic etiology was part of a previously identified autosomal recessive disorder. Two novel de novo mutations were identified in INS and HNF1B. Conclusion Qatar has the second highest reported incidence of PNDM worldwide. A majority of PNDM cases present as rare familial autosomal recessive disorders. Pancreas associated transcription factor 1a (PTF1A) enhancer deletions are the most common cause of PNDM in Qatar, with only a few previous cases reported in the literature.
Collapse
Affiliation(s)
- Sara Al-Khawaga
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar.,Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Idris Mohammed
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Saras Saraswathi
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Basma Haris
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Reem Hasnah
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Amira Saeed
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | | | - Najeeb Syed
- Biomedical Informatics Division, Sidra Medicine, Doha, Qatar
| | - Puthen Jithesh
- Biomedical Informatics Division, Sidra Medicine, Doha, Qatar
| | - Ahmed El Awwa
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar.,Faculty of medicine, Alexandria University, Alexandria, Egypt
| | - Amal Khalifa
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Fawziya AlKhalaf
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Goran Petrovski
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Essam M Abdelalim
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
38
|
Balamurugan K, Kavitha B, Yang Z, Mohan V, Radha V, Shyng SL. Functional characterization of activating mutations in the sulfonylurea receptor 1 (ABCC8) causing neonatal diabetes mellitus in Asian Indian children. Pediatr Diabetes 2019; 20:397-407. [PMID: 30861254 PMCID: PMC11423867 DOI: 10.1111/pedi.12843] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/28/2019] [Accepted: 02/24/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Gain-of-function of ATP-sensitive K+ (KATP ) channels because of mutations in the genes encoding SUR1 (ABCC8) or Kir6.2 (KCNJ11) is a major cause of neonatal diabetes mellitus (NDM). Our aim is to determine molecular defects in KATP channels caused by ABCC8 mutations in Asian Indian children with NDM by in vitro functional studies. METHODS Wild-type (WT; NM_000352.4) or mutant sulfonylurea receptor 1 (SUR1) and Kir6.2 were co-expressed in COSm6 cells. Biogenesis efficiency and surface expression of mutant channels were assessed by immunoblotting and immunostaining. The response of mutant channels to cytoplasmic ATP and ADP was assessed by inside-out patch-clamp recordings. The response of mutant channels to known KATP inhibitors in intact cells were determined by 86 Rb efflux assays. RESULTS Five SUR1 missense mutations, D212Y, P254S, R653Q, R992C, and Q1224H, were studied and showed increased activity in MgATP/MgADP. Two of the mutants, D212Y and P254S, also showed reduced response to ATP4- inhibition, as well as markedly reduced surface expression. Moreover, all five mutants were inhibited by the KATP channel inhibitors glibenclamide and carbamazepine. CONCLUSIONS The study shows the mechanisms by which five SUR1 mutations identified in Asian Indian NDM patients affect KATP channel function to cause the disease. The reduced ATP4- sensitivity caused by the D212Y and P254S mutations in the L0 of SUR1 provides novel insight into the role of L0 in channel inhibition by ATP. The results also explain why sulfonylurea therapy is effective in two patients and inform how it should be effective for the other three patients.
Collapse
Affiliation(s)
- Kandasamy Balamurugan
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialties Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon
| | - Babu Kavitha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialties Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - Zhongying Yang
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon
| | - Viswanathan Mohan
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialties Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - Venkatesan Radha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialties Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
39
|
Saraswathi S, Al-Khawaga S, Elkum N, Hussain K. A Systematic Review of Childhood Diabetes Research in the Middle East Region. Front Endocrinol (Lausanne) 2019; 10:805. [PMID: 31824422 PMCID: PMC6882272 DOI: 10.3389/fendo.2019.00805] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Diabetes mellitus (DM) is a common chronic disorder in children and is caused by absolute or relative insulin deficiency, with or without insulin resistance. There are several different forms of childhood DM. Children can suffer from neonatal diabetes mellitus (NDM), type 1 diabetes (T1DM), type 2 diabetes (T2DM), Maturity Onset Diabetes of the Young (MODY), autoimmune monogenic, mitochondrial, syndromic and as yet unclassified forms of DM. The Middle East has one of the highest incidences of several types of DM in children; however, it is unclear whether pediatric diabetes is an active area of research in the Middle East and if ongoing, which research areas are of priority for DM in children. Objectives: To review the literature on childhood DM related to research in the Middle East, summarize results, identify opportunities for research and make observations and recommendations for collaborative studies in pediatric DM. Methods: We conducted a thorough and systematic literature review by adhering to a list recommended by PRISMA. We retrieved original papers written in English that focus on childhood DM research, using electronic bibliographic databases containing publications from the year 2000 until October 2018. For our final assessment, we retrieved 429 full-text articles and selected 95 articles, based on our inclusion and exclusion criteria. Results: Our literature review suggests that childhood DM research undertaken in the Middle East has focused mainly on reporting retrospective review of case notes, a few prospective case studies, systemic reviews, questionnaire-based studies, and case reports. These reported studies have focused mostly on the incidence/prevalence of different types of DM in childhood. No studies report on the establishment of National Childhood Diabetes Registries. There is a lack of consolidated studies focusing on national epidemiology data of different types of childhood DM (such as NDM, T1DM, T2DM, MODY, and syndromic forms) and no studies reporting on clinical trials in children with DM. Conclusions: Investing in and funding basic and translational childhood diabetes research and encouraging collaborative studies, will bring enormous benefits financially, economically, and socially for the whole of the Middle East region.
Collapse
Affiliation(s)
- Saras Saraswathi
- Division of Endocrinology, Department of Pediatrics, Sidra Medicine, Doha, Qatar
| | - Sara Al-Khawaga
- Division of Endocrinology, Department of Pediatrics, Sidra Medicine, Doha, Qatar
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Education City, Doha, Qatar
| | - Naser Elkum
- Biostatistics Section, Clinical Research Center, Research Services, Sidra Medicine, Doha, Qatar
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatrics, Sidra Medicine, Doha, Qatar
- *Correspondence: Khalid Hussain
| |
Collapse
|
40
|
Sikimic J, McMillen TS, Bleile C, Dastvan F, Quast U, Krippeit-Drews P, Drews G, Bryan J. ATP binding without hydrolysis switches sulfonylurea receptor 1 (SUR1) to outward-facing conformations that activate K ATP channels. J Biol Chem 2018; 294:3707-3719. [PMID: 30587573 DOI: 10.1074/jbc.ra118.005236] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/19/2018] [Indexed: 11/06/2022] Open
Abstract
Neuroendocrine-type ATP-sensitive K+ (KATP) channels are metabolite sensors coupling membrane potential with metabolism, thereby linking insulin secretion to plasma glucose levels. They are octameric complexes, (SUR1/Kir6.2)4, comprising sulfonylurea receptor 1 (SUR1 or ABCC8) and a K+-selective inward rectifier (Kir6.2 or KCNJ11). Interactions between nucleotide-, agonist-, and antagonist-binding sites affect channel activity allosterically. Although it is hypothesized that opening these channels requires SUR1-mediated MgATP hydrolysis, we show here that ATP binding to SUR1, without hydrolysis, opens channels when nucleotide antagonism on Kir6.2 is minimized and SUR1 mutants with increased ATP affinities are used. We found that ATP binding is sufficient to switch SUR1 alone between inward- or outward-facing conformations with low or high dissociation constant, KD , values for the conformation-sensitive channel antagonist [3H]glibenclamide ([3H]GBM), indicating that ATP can act as a pure agonist. Assembly with Kir6.2 reduced SUR1's KD for [3H]GBM. This reduction required the Kir N terminus (KNtp), consistent with KNtp occupying a "transport cavity," thus positioning it to link ATP-induced SUR1 conformational changes to channel gating. Moreover, ATP/GBM site coupling was constrained in WT SUR1/WT Kir6.2 channels; ATP-bound channels had a lower KD for [3H]GBM than ATP-bound SUR1. This constraint was largely eliminated by the Q1179R neonatal diabetes-associated mutation in helix 15, suggesting that a "swapped" helix pair, 15 and 16, is part of a structural pathway connecting the ATP/GBM sites. Our results suggest that ATP binding to SUR1 biases KATP channels toward open states, consistent with SUR1 variants with lower KD values causing neonatal diabetes, whereas increased KD values cause congenital hyperinsulinism.
Collapse
Affiliation(s)
- Jelena Sikimic
- From the Institute of Pharmacy, Department of Pharmacology, University of Tübingen, D-72076 Tübingen, Germany and
| | - Timothy S McMillen
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122, and
| | - Cita Bleile
- From the Institute of Pharmacy, Department of Pharmacology, University of Tübingen, D-72076 Tübingen, Germany and
| | - Frank Dastvan
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122, and
| | - Ulrich Quast
- Department of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, D-72074 Tübingen, Germany
| | - Peter Krippeit-Drews
- From the Institute of Pharmacy, Department of Pharmacology, University of Tübingen, D-72076 Tübingen, Germany and
| | - Gisela Drews
- From the Institute of Pharmacy, Department of Pharmacology, University of Tübingen, D-72076 Tübingen, Germany and
| | - Joseph Bryan
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122, and
| |
Collapse
|
41
|
Balboa D, Saarimäki-Vire J, Otonkoski T. Concise Review: Human Pluripotent Stem Cells for the Modeling of Pancreatic β-Cell Pathology. Stem Cells 2018; 37:33-41. [PMID: 30270471 PMCID: PMC7379656 DOI: 10.1002/stem.2913] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/21/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic β‐cells are the only source of insulin. Disturbances in β‐cell development or function may thus result in insulin deficiency or excess, presenting as hyper‐ or hypoglycemia. It is increasingly evident that common forms of diabetes (types 1 and 2) are pathogenically heterogeneous. Development of efficient therapies is dependent on reliable disease models. Although animal models are remarkably useful research tools, they present limitations because of species differences. As an alternative, human pluripotent stem cell technologies offer multiple possibilities for the study of human diseases in vitro. In the last decade, advances in the derivation of induced pluripotent stem cells from diabetic patients, combined with β‐cell differentiation protocols, have resulted in the generation of useful disease models for diabetes. First disease models have been focusing on monogenic diabetes. The development of genome editing technologies, more advanced differentiation protocols and humanized mouse models based on transplanted cells have opened new horizons for the modeling of more complex forms of β‐cell dysfunction. We present here the incremental progress made in the modeling of diabetes using pluripotent stem cells. We discuss the current challenges and opportunities of these approaches to dissect β‐cell pathology and devise new pharmacological and cell replacement therapies. stem cells2019;37:33–41
Collapse
Affiliation(s)
- Diego Balboa
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jonna Saarimäki-Vire
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
42
|
Chen T, Zhang D, Bai Z, Wu S, Wu H, Xie R, Li Y, Wang F, Chen X, Sun H, Wang X, Chen L. Successful Treatment of Diabetic Ketoacidosis and Hyperglycemic Hyperosmolar Status in an Infant with KCNJ11-Related Neonatal Diabetes Mellitus via Continuous Renal Replacement Therapy. Diabetes Ther 2018; 9:2179-2184. [PMID: 30094785 PMCID: PMC6167281 DOI: 10.1007/s13300-018-0484-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
Neonatal diabetes mellitus (NDM) is a rare monogenic disorder presenting as uncontrolled hyperglycemia during the first 6 months of life. Hyperglycemic hyperosmolar state (HHS) is quite rare in NDM patients, and reported experience with this condition is limited. Continuous renal replacement therapy (CRRT) is frequently used as a mode of dialytic treatment in critically ill patients with acute renal failure, but has seldom been used in patients with diabetic ketoacidosis (DKA) and HHS. We report the case of a 2-month-old infant admitted to our hospital presenting with dyspnea and lethargy. Blood gas showed severe hyperosmotic DKA. After 21 h of fluid and insulin therapy, the baby presented with increased drowsiness and irregular respiration, which suggested cerebral edema. Moreover, the DKA and HHS were exacerbated. After 18 h of CRRT, the patient gradually recovered from DKA and HHS. The gene analysis revealed a de novo mutation (c.602G > A (p.R201H)) of the KCNJ11 gene, and oral glibenclamide successfully replaced insulin treatment in the patient.
Collapse
Affiliation(s)
- Ting Chen
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dandan Zhang
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhenjiang Bai
- Department of Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shuiyan Wu
- Department of Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haiying Wu
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rongrong Xie
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying Li
- Department of Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fengyun Wang
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiuli Chen
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hui Sun
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoyan Wang
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Linqi Chen
- Department of Endocrinology, Metabolism and Genetic Disorders, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
43
|
Li N, Yang Z, Li Q, Yu Z, Chen X, Li JC, Li B, Ning SL, Cui M, Sun JP, Yu X. Ablation of somatostatin cells leads to impaired pancreatic islet function and neonatal death in rodents. Cell Death Dis 2018; 9:682. [PMID: 29880854 PMCID: PMC5992210 DOI: 10.1038/s41419-018-0741-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 02/08/2023]
Abstract
The somatostatin (SST)-secreting cells were mainly distributed in the pancreatic islets, brain, stomach and intestine in mammals and have many physiological functions. In particular, the SST-secreting δ cell is the third most common cell type in the islets of Langerhans. Recent studies have suggested that dysregulation of paracrine interaction between the pancreatic δ cells and β cells results in impaired glucose homeostasis and contributes to diabetes development. However, direct evidence of the functional importance of SST cells in glucose homeostasis control is still lacking. In the present study, we specifically ablated SST-secreting cells by crossing Sst-cre transgenic mice with R26 DTA mice (Sst Cre R26 DTA ). The Sst Cre R26 DTA mice exhibited neonatal death. The life spans of these mice with severe hypoglycemia were extended by glucose supplementation. Moreover, we observed that SST cells deficiency led to increased insulin content and excessive insulin release, which might contribute to the observed hypoglycemia. Unexpectedly, although SST is critical for the regulation of insulin content, factors other than SST that are produced by pancreatic δ cells via their endogenous corticotropin-releasing hormone receptor 2 (CRHR2) activity play the main roles in maintaining normal insulin release, as well as neonatal glucose homeostasis in the resting state. Taken together, our results identified that the SST cells in neonatal mouse played critical role in control of insulin release and normal islet function. Moreover, we provided direct in vivo evidence of the functional importance of the SST cells, which are essential for neonatal survival and the maintenance of glucose homeostasis.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Zhao Yang
- Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Qing Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Zhen Yu
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Xu Chen
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Jia-Cheng Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Bo Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Shang-Lei Ning
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong, 250012, China
| | - Min Cui
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Jin-Peng Sun
- Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China.,School of Medicine, Duke University, Durham, North Carolina, 27705, USA
| | - Xiao Yu
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China.
| |
Collapse
|
44
|
Neonatal Diabetes Mellitus. MEDICAL BULLETIN OF SISLI ETFAL HOSPITAL 2018; 52:71-78. [PMID: 32595377 PMCID: PMC7315067 DOI: 10.14744/semb.2017.51422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 06/07/2017] [Indexed: 11/23/2022]
Abstract
Neonatal diabetes is a rare cause of hyperglycemia in the neonatal period. It is caused by mutations in genes that encode proteins playing critical roles in normal functions of pancreatic beta cells. Neonatal diabetes is divided into temporary and permanent subtypes. Treatment is based on the correction of fluid-electrolyte disturbances and hyperglycemia. Patients respond to insulin or sulfonylurea treatment according to the mutation type. Close glucose monitoring and education of caregivers about diabetes are vital.
Collapse
|
45
|
Abbasi F, Habibi M, Enayati S, Bitarafan F, Razzaghy-Azar M, Sotodeh A, Omran SP, Maroofian R, Amoli MM. A Genotype-First Approach for Clinical and Genetic Evaluation of Wolcott-Rallison Syndrome in a Large Cohort of Iranian Children With Neonatal Diabetes. Can J Diabetes 2018; 42:272-275. [DOI: 10.1016/j.jcjd.2017.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/12/2017] [Accepted: 06/21/2017] [Indexed: 11/15/2022]
|
46
|
Rohm M, Savic D, Ball V, Curtis MK, Bonham S, Fischer R, Legrave N, MacRae JI, Tyler DJ, Ashcroft FM. Cardiac Dysfunction and Metabolic Inflexibility in a Mouse Model of Diabetes Without Dyslipidemia. Diabetes 2018; 67:1057-1067. [PMID: 29610263 DOI: 10.2337/db17-1195] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/12/2018] [Indexed: 11/13/2022]
Abstract
Diabetes is a well-established risk factor for heart disease, leading to impaired cardiac function and a metabolic switch toward fatty acid usage. In this study, we investigated if hyperglycemia/hypoinsulinemia in the absence of dyslipidemia is sufficient to drive these changes and if they can be reversed by restoring euglycemia. Using the βV59M mouse model, in which diabetes can be rapidly induced and reversed, we show that stroke volume and cardiac output were reduced within 2 weeks of diabetes induction. Flux through pyruvate dehydrogenase was decreased, as measured in vivo by hyperpolarized [1-13C]pyruvate MRS. Metabolomics showed accumulation of pyruvate, lactate, alanine, tricarboxyclic acid cycle metabolites, and branched-chain amino acids. Myristic and palmitoleic acid were decreased. Proteomics revealed proteins involved in fatty acid metabolism were increased, whereas those involved in glucose metabolism decreased. Western blotting showed enhanced pyruvate dehydrogenase kinase 4 (PDK4) and uncoupling protein 3 (UCP3) expression. Elevated PDK4 and UCP3 and reduced pyruvate usage were present 24 h after diabetes induction. The observed effects were independent of dyslipidemia, as mice showed no evidence of elevated serum triglycerides or lipid accumulation in peripheral organs (including the heart). The effects of diabetes were reversible, as glibenclamide therapy restored euglycemia, cardiac metabolism and function, and PDK4/UCP3 levels.
Collapse
Affiliation(s)
- Maria Rohm
- Department of Physiology, Anatomy and Genetics and OXION, University of Oxford, Oxford, U.K
| | - Dragana Savic
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Vicky Ball
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - M Kate Curtis
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Sarah Bonham
- Discovery Proteomics Facility, Target Discovery Institute, University of Oxford, Oxford, U.K
| | - Roman Fischer
- Discovery Proteomics Facility, Target Discovery Institute, University of Oxford, Oxford, U.K
| | | | | | - Damian J Tyler
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Frances M Ashcroft
- Department of Physiology, Anatomy and Genetics and OXION, University of Oxford, Oxford, U.K.
| |
Collapse
|
47
|
Evliyaoğlu O, Ercan O, Ataloğlu E, Zübarioğlu Ü, Özcabı B, Dağdeviren A, Erdoğan H, De Franco E, Ellard S. Neonatal Diabetes: Two Cases with Isolated Pancreas Agenesis due to Homozygous PTF1A Enhancer Mutations and One with Developmental Delay, Epilepsy, and Neonatal Diabetes Syndrome due to KCNJ11 Mutation. J Clin Res Pediatr Endocrinol 2018; 10:168-174. [PMID: 28943513 PMCID: PMC5985387 DOI: 10.4274/jcrpe.5162] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/23/2017] [Indexed: 12/01/2022] Open
Abstract
Neonatal diabetes mellitus is a rare form of monogenic diabetes which is diagnosed in the first six months of life. Here we report three patients with neonatal diabetes; two with isolated pancreas agenesis due to mutations in the pancreas-specific transcription factor 1A (PTF1A) enhancer and one with developmental delay, epilepsy, and neonatal diabetes (DEND) syndrome, due to a KCNJ11 mutation. The two cases with mutations in the distal enhancer of PTF1A had a homozygous g.23508363A>G and a homozygous g.23508437A>G mutation respectively. Previous functional analyses showed that these mutations can decrease expression of PTF1A which is involved in pancreas development. Both patients were born small for gestational age to consanguineous parents. Both were treated with insulin and pancreatic enzymes. One of these patients’ fathers was also homozygous for the PTF1A mutation, whilst his partner and the parents of the other patient were heterozygous carriers. In the case with DEND sydrome, a previosly reported heterozygous KCNJ11 mutation, p.Cys166Tyr (c.497G>A), was identified. This patient was born to nonconsanguineous parents with normal birth weight. The majority of neonatal diabetes patients with KCNJ11 mutations will respond to sulphonylurea treatment. Therefore Glibenclamide, an oral antidiabetic of the sulphonylurea group, was started. This treatment regimen relatively improved blood glucose levels and neurological symptoms in the short term. Because we could not follow the patient in the long term, we are not able to draw conclusions about the efficacy of the treatment. Although neonatal diabetes mellitus can be diagnosed clinically, genetic analysis is important since it is a guide for the treatment and for prognosis.
Collapse
Affiliation(s)
- Olcay Evliyaoğlu
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
| | - Oya Ercan
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
| | - Emel Ataloğlu
- University of Health Science, Haseki Training and Research Hospital, Newborn Intensive Unit, İstanbul, Turkey
| | - Ümit Zübarioğlu
- Şişli Hamidiye Etfal Training and Research Hospital, Newborn Intensive Unit, İstanbul, Turkey
| | - Bahar Özcabı
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
| | - Aydilek Dağdeviren
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
| | - Hande Erdoğan
- İstanbul University Cerrahpaşa Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
| | - Elisa De Franco
- University of Exeter Medical School, Institute of Biomedical and Clinical Science, Exeter, United Kingdom
| | - Sian Ellard
- University of Exeter Medical School, Institute of Biomedical and Clinical Science, Exeter, United Kingdom
| |
Collapse
|
48
|
Dahl AR, Dhamija R, Nofal AA, Pittock ST, Schwenk WF, Kumar S. Transient Neonatal Diabetes due to a Mutation in KCNJ11 in a Child with Klinefelter Syndrome. J Clin Res Pediatr Endocrinol 2018; 10:79-82. [PMID: 28766502 PMCID: PMC5838377 DOI: 10.4274/jcrpe.4807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Klinefelter syndrome is the most frequent chromosomal aneuploidy in males occurring in about 1 in 660 males. Epidemiological studies have demonstrated increased risk of type 1 diabetes and type 2 diabetes in adults with Klinefelter syndrome. There is only one previous report of neonatal diabetes in a patient with Klinefelter syndrome. We report transient neonatal diabetes due to a pathogenic heterozygous variant in KCNJ11 in a male infant with Klinefelter syndrome. A 78-day old male infant was noted to have sustained hyperglycemia with serum glucose ranging between 148 mg/dL (8.2 mmol/L) and 381 mg/dL (21.2 mmol/L) three days after undergoing a complete repair of an atrioventricular defect. Hemoglobin A1c was 6.6%. The patient was born at term with a birth weight of 2.16 kg following a pregnancy complicated by gestational diabetes that was controlled with diet. The patient was initially started on a continuous intravenous insulin drip and subsequently placed on subcutaneous insulin (glargine, human isophane and regular insulin). Insulin was gradually decreased and eventually discontinued at seven months of age. Chromosomal microarray at 11 weeks of age showed XXY and a panel-based, molecular test for neonatal diabetes revealed a pathogenic heterozygous variant c.685G>A (p.Glu229Lys) in KCNJ11. The patient is now 34 months old and continues to have normal fasting and post-prandial glucose and HbA1C levels. The patient will need prospective follow up for assessment of his glycemic status. To our knowledge this is the second reported case of neonatal diabetes in an infant with Klinefelter syndrome and the first due to a mutation in the KCNJ11 in a patient with Klinefelter syndrome.
Collapse
Affiliation(s)
- Amanda R. Dahl
- Mayo Clinic, Department of Pediatric and Adolescent Medicine, Rochester, Minnesota, USA
| | - Radhika Dhamija
- Mayo Clinic, Department of Clinical Genomics, Phoenix, Arizona, USA
| | - Alaa Al Nofal
- University of South Dakota, Sanford Children Specialty Clinic, Division of Pediatric Endocrinology, Sioux Falls, South Dakota, USA
| | - Siobhan T. Pittock
- Mayo Clinic, Department of Pediatric and Adolescent Medicine, Division of Pediatric Endocrinology, Rochester, Minnesota, USA
| | - W. Frederick Schwenk
- Mayo Clinic, Department of Pediatric and Adolescent Medicine, Division of Pediatric Endocrinology, Rochester, Minnesota, USA
| | - Seema Kumar
- Mayo Clinic, Department of Pediatric and Adolescent Medicine, Division of Pediatric Endocrinology, Rochester, Minnesota, USA,* Address for Correspondence: Department of Pediatric and Adolescent Medicine, Division of Pediatric Endocrinology, Rochester, Minnesota, USA GSM: +507-284-3300 E-mail:
| |
Collapse
|
49
|
Li X, Xu A, Sheng H, Ting TH, Mao X, Huang X, Jiang M, Cheng J, Liu L. Early transition from insulin to sulfonylureas in neonatal diabetes and follow-up: Experience from China. Pediatr Diabetes 2018; 19:251-258. [PMID: 28791793 DOI: 10.1111/pedi.12560] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/22/2017] [Accepted: 06/20/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Sulfonylurea therapy can improve glycemic control and ameliorate neurodevelopmental outcomes in patients suffering from neonatal diabetes mellitus (NDM) with KCNJ11 or ABCC8 mutations. As genetic testing results are often delayed, it remains controversial whether sulfonylurea treatment should be attempted immediately at diagnosis or doctors should await genetic confirmation. OBJECTIVE This study aimed to investigate the effectiveness and safety of sulfonylurea therapy in Chinese NDM patients during infancy before genetic testing results were available. METHODS The medical records of NDM patients with their follow-up details were reviewed and molecular genetic analysis was performed. Sulfonylurea transfer regimens were applied in patients diagnosed after May 2010, and glycemic status and side effects were evaluated in each patient. RESULTS There were 23 NDM patients from 22 unrelated families, 10 had KCNJ11 mutations, 3 harbored ABCC8 mutations, 1 had INS mutations, 4 had chromosome 6q24 abnormalities, 1 had a deletion at chromosome 1p36.23p36.12, and 4 had no genetic abnormality identified. Sixteen NDM infants were treated with glyburide at an average age of 49 days (range 14-120 days) before genetic confirmation. A total of 11 of 16 (69%) were able to successfully switch to glyburide with a more stable glucose profile. The responsive glyburide dose was 0.51 ± 0.16 mg/kg/d (0.3-0.8 mg/kg/d), while the maintenance dose was 0.30 ± 0.07 mg/kg/d (0.2-0.4 mg/kg/d). No serious adverse events were reported. CONCLUSIONS Molecular genetic diagnosis is recommended in all patients with NDM. However, if genetic testing results are delayed, sulfonylurea therapy should be considered before such results are received, even in infants with newly diagnosed NDM.
Collapse
Affiliation(s)
- Xiuzhen Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Aijing Xu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Huiying Sheng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Tzer Hwu Ting
- Department of Paediatrics, Faculty of Medicine and Health Sciences, Univeristy Putra Malaysia, Serdang, Malaysia
| | - Xiaojian Mao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xinjiang Huang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Minyan Jiang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Jing Cheng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| |
Collapse
|
50
|
Rosa AP, Mescka CP, Catarino FM, de Castro AL, Teixeira RB, Campos C, Baldo G, Graf DD, de Mattos-Dutra A, Dutra-Filho CS, da Rosa Araujo AS. Neonatal hyperglycemia induces cell death in the rat brain. Metab Brain Dis 2018; 33:333-342. [PMID: 29260360 DOI: 10.1007/s11011-017-0170-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 12/11/2017] [Indexed: 02/06/2023]
Abstract
Several studies have examined neonatal diabetes, a rare disease characterized by hyperglycemia and low insulin levels that is usually diagnosed in the first 6 month of life. Recently, the effects of diabetes on the brain have received considerable attention. In addition, hyperglycemia may perturb brain function and might be associated with neuronal death in adult rats. However, few studies have investigated the damaging effects of neonatal hyperglycemia on the rat brain during central nervous system (CNS) development, particularly the mechanisms involved in the disease. Thus, in the present work, we investigated whether neonatal hyperglycemia induced by streptozotocin (STZ) promoted cell death and altered the levels of proteins involved in survival/death pathways in the rat brain. Cell death was assessed using FluoroJade C (FJC) staining and the expression of the p38 mitogen-activated protein kinase (p38), phosphorylated-c-Jun amino-terminal kinase (p-JNK), c-Jun amino-terminal kinase (JNK), protein kinase B (Akt), phosphorylated-protein kinase B (p-Akt), glycogen synthase kinase-3β (Gsk3β), B-cell lymphoma 2 (Bcl2) and Bcl2-associated X protein (Bax) protein were measured by Western blotting. The main results of this study showed that the metabolic alterations observed in diabetic rats (hyperglycemia and hypoinsulinemia) increased p38 expression and decreased p-Akt expression, suggesting that cell survival was altered and cell death was induced, which was confirmed by FJC staining. Therefore, the metabolic conditions observed during neonatal hyperglycemia may contribute to the harmful effect of diabetes on the CNS in a crucial phase of postnatal neuronal development.
Collapse
Affiliation(s)
- Andrea Pereira Rosa
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 (Anexo), Porto Alegre, RS, 90035-003, Brazil.
| | - Caroline Paula Mescka
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Maciel Catarino
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 (Anexo), Porto Alegre, RS, 90035-003, Brazil
| | - Alexandre Luz de Castro
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rayane Brinck Teixeira
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cristina Campos
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Guilherme Baldo
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Débora Dalmas Graf
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Angela de Mattos-Dutra
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carlos Severo Dutra-Filho
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 (Anexo), Porto Alegre, RS, 90035-003, Brazil
| | - Alex Sander da Rosa Araujo
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|