1
|
Moon JE, Lee YN, Jeong S, Jun HR, Hoang MH, Jo Y, Jang J, Shim IK, Kim SC. Enhancing differentiation and functionality of insulin-producing cells derived from iPSCs using esterified collagen hydrogel for cell therapy in diabetes mellitus. Stem Cell Res Ther 2024; 15:374. [PMID: 39443977 PMCID: PMC11515471 DOI: 10.1186/s13287-024-03971-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Islet transplantation is a recommended treatment for type 1 diabetes but is limited by donor organ shortage. This study introduces an innovative approach for improving the differentiation and functionality of insulin-producing cells (IPCs) from iPSCs using 3D spheroid formation and hydrogel matrix as an alternative pancreatic islet source. The extracellular matrix (ECM) is crucial for pancreatic islet functionality, but finding the ideal matrix for β-cell differentiation has been challenging. We aimed to advance IPC differentiation and maturation through an esterified collagen hydrogel, comparing its effectiveness with conventional basement membrane extract (BME) hydrogels. METHODS iPSCs were differentiated into IPCs using a small molecule-based sequential protocol, followed by spheroid formation in concave microwells. Rheological analysis, scanning electron microscopy, and proteomic profiling were used to characterize the chemical and physical properties of each matrix. IPCs, both in single-cell form and as spheroids, were embedded in either ionized collagen or BME hydrogels, which was followed by assessments of morphological changes, pancreatic islet-related gene expression, insulin secretion, and pathway activation using comprehensive analytical techniques. RESULTS Esterified collagen hydrogels markedly improved the structural integrity, insulin expression, and cell-cell interactions in IPC spheroids, forming densely packed insulin-expressing clusters, in contrast to the dispersed cells observed in BME cultures. Collagen hydrogel significantly enhanced the mRNA expression of crucial endocrine markers and maturation factors, with IPC spheroids showing accelerated differentiation from day 5, suggesting a faster differentiation compared to single cells in hydrogel encapsulation. Insulin secretion in response to glucose in collagen environments, with a GSIS index of 2.46 ± 0.05, exceeded those in 2D and BME, demonstrating superior pancreatic islet functionality. Pathway analysis highlighted enhanced insulin secretion capabilities, evidenced by the upregulation of genes like Secretogranin III and Chromogranin A in collagen cultures. In vivo transplantation results showed that collagen hydrogel enhanced cluster integrity, tissue integration, and insulin secretion compared to non-embedded IPCs and BME groups. CONCLUSION Esterified collagen hydrogels demonstrated superior efficacy over 2D and BME in promoting IPC differentiation and maturation, possibly through upregulation of the expression of key secretion pathway genes. Our findings suggest that using collagen hydrogels presents a promising approach to enhance insulin secretion efficiency in differentiating pancreatic β-cells, advancing cell therapy in diabetes cell therapy.
Collapse
Affiliation(s)
- Ji Eun Moon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Yu Na Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Sehui Jeong
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hye Ryeong Jun
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Minh Hien Hoang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering (POSTECH), Pohang, 37673, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering (POSTECH), Pohang, 37673, Republic of Korea
- Department of Mechanical Engineering (POSTECH), Pohang, 37673, Republic of Korea
| | - In Kyong Shim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Department of Biomedical Engineering, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Song Cheol Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
- Asan Medical Center, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Brain Korea 21 Project, Seoul, Korea.
| |
Collapse
|
2
|
Ouyang S, Xiang S, Wang X, Yang X, Liu X, Zhang M, Zhou Y, Xiao Y, Zhou L, Fan G, Yang J. The downregulation of SCGN induced by lipotoxicity promotes NLRP3-mediated β-cell pyroptosis. Cell Death Discov 2024; 10:340. [PMID: 39068218 PMCID: PMC11283536 DOI: 10.1038/s41420-024-02107-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
Lipotoxicity is a well-established phenomenon that could exacerbate damage to islet β-cells and play a significant role in the development of type 2 diabetes, the underlying mechanisms of which, however, remain unclear. In lipotoxic conditions, secretagogin (SCGN), an EF-hand calcium-binding protein abundantly expressed in islets, is found to undergo downregulation. In light of this, we aim to explore the role of SCGN in lipotoxicity-induced β-cell injury. Our findings show that exposure to ox-LDL in vitro or long-term high-fat diets (HFD) in vivo decreases SCGN expression and induces pyroptosis in β-cells. Moreover, restoring SCGN partially reverses the pyroptotic cell death under ox-LDL or HFD treatments. We have observed that the downregulation of SCGN facilitates the translocation of ChREBP from the cytosol to the nucleus, thereby promoting TXNIP transcription. The upregulation of TXNIP activates the NLRP3/Caspase-1 pathway, leading to pyroptotic cell death. In summary, our study demonstrates that lipotoxicity leads to the downregulation of SCGN expression in islet β-cells, resulting in ChREBP accumulation in the nucleus and subsequent activation of the NLRP3/Caspase-1 pyroptotic pathway. Thus, administering SCGN could be a potential therapeutic strategy to alleviate β-cell damage induced by lipotoxicity in type 2 diabetes.
Collapse
Affiliation(s)
- Shuhui Ouyang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Sunmin Xiang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Department of Hospital Infection Control, Xingsha District of Hunan Provincial People's Hospital (Changsha County People's Hospital), Changsha, 410100, Hunan, China
| | - Xin Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xin Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xuan Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Meilin Zhang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yiting Zhou
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yang Xiao
- The School of Humanities and Social Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Lingzhi Zhou
- Department of pediatrics, Huazhong University of Science and Technology Union Shenzhen Hospital (Shenzhen Nanshan people's hospital), Shenzhen, 518052, Guangdong, China
| | - Gang Fan
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital (Shenzhen Nanshan people's hospital), Shenzhen, 518052, Guangdong, China.
| | - Jing Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- Department of Metabolism and Endocrinology, Huazhong University of Science and Technology Union Shenzhen Hospital (Shenzhen Nanshan people's hospital), Shenzhen, 518052, Guangdong, China.
| |
Collapse
|
3
|
Mendonça V, Soares-Lima SC, Moreira MAM. Exploring cross-tissue DNA methylation patterns: blood-brain CpGs as potential neurodegenerative disease biomarkers. Commun Biol 2024; 7:904. [PMID: 39060467 PMCID: PMC11282059 DOI: 10.1038/s42003-024-06591-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The difficulty of obtaining samples from certain human tissues has led to efforts to find accessible sources to analyze molecular markers derived from DNA. In this study, we look for DNA methylation patterns in blood samples and its association with the brain methylation pattern in neurodegenerative disorders. Using data from methylation databases, we selected 18,293 CpGs presenting correlated methylation levels between blood and brain (bb-CpGs) and compare their methylation level between blood samples from patients with neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Multiple Sclerosis, and X Fragile Syndrome) and healthy controls. Sixty-four bb-CpGs presented significant distinct methylation levels in patients, being: nine for Alzheimer's disease, nine for Parkinson's disease, 28 for Multiple Sclerosis, and 18 for Fragile X Syndrome. Similar differences in methylation pattern for the nine Alzheimer's bb-CpGs was also observed when comparing brain tissue from patients vs. controls. The genomic regions of some of these 64 bb-CpGs are placed close to or inside genes previously associated with the respective condition. Our findings support the rationale of using blood DNA as a surrogate of brain tissue to analyze changes in CpG methylation level in patients with neurodegenerative diseases, opening the possibility for characterizing new biomarkers.
Collapse
Affiliation(s)
- Vanessa Mendonça
- Genetic Graduation Program, Genetics Deparment, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Tumoral Genetics and Virology Program, Instituto Nacional de Cancer, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
4
|
Guo T, Wang X, Wang T, Zhang J, Liu Y, Chen S, Wang X, Yang X, Wang C, Wang X. Dynamic changes of SCGN expression imply different phases of clear cell renal cell carcinoma progression. Discov Oncol 2024; 15:205. [PMID: 38831128 PMCID: PMC11147981 DOI: 10.1007/s12672-024-01071-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 05/29/2024] [Indexed: 06/05/2024] Open
Abstract
The secretagogin (SCGN) was originally identified as a secreted calcium-binding protein present in the cytoplasm. Recent studies have found that SCGN has a close relationship with cancer. However, its role in the occurrence, progression, and prognosis of clear cell renal cell carcinoma (ccRCC) remains unclear. In this study, we utilized a mutual authentication method based on public databases and clinical samples to determine the role of SCGN in the progression and prognosis of ccRCC. Firstly, we comprehensively analyzed the expression characteristics of SCGN in ccRCC in several public databases. Subsequently, we systematically evaluated SCGN expression on 252 microarrays of ccRCC tissues from different grades. It was found that SCGN was absent in all the normal kidney tissues and significantly overexpressed in ccRCC tumor tissues. In addition, the expression level of SCGN gradually decreased with an increase in tumor grade, and the percentage of SCGN staining positivity over 50% was 86.7% (13/15) and 73.4% (58/79) in Grade1 and Grade2, respectively, while it was only 8.3% (12/144) in Grade3, and the expression of SCGN was completely absent in Grade4 (0/14) and distant metastasis group (0/4). Additionally, the expression of SCGN was strongly correlated with the patient's prognosis, with the higher the expression levels of SCGN being associated with longer overall survival and disease-free survival of patients. In conclusion, our results suggest that reduced expression of SCGN in cancer cells is correlated with the progression and prognosis of ccRCC.
Collapse
Affiliation(s)
- Tuanjie Guo
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuan Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Liu
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siteng Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Wang
- Department of Pathology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqun Yang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Pathology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
SCGN deficiency is a risk factor for autism spectrum disorder. Signal Transduct Target Ther 2023; 8:3. [PMID: 36588101 PMCID: PMC9806109 DOI: 10.1038/s41392-022-01225-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/15/2022] [Accepted: 09/30/2022] [Indexed: 01/03/2023] Open
Abstract
Autism spectrum disorder (ASD) affects 1-2% of all children and poses a great social and economic challenge for the globe. As a highly heterogeneous neurodevelopmental disorder, the development of its treatment is extremely challenging. Multiple pathways have been linked to the pathogenesis of ASD, including signaling involved in synaptic function, oxytocinergic activities, immune homeostasis, chromatin modifications, and mitochondrial functions. Here, we identify secretagogin (SCGN), a regulator of synaptic transmission, as a new risk gene for ASD. Two heterozygous loss-of-function mutations in SCGN are presented in ASD probands. Deletion of Scgn in zebrafish or mice leads to autism-like behaviors and impairs brain development. Mechanistically, Scgn deficiency disrupts the oxytocin signaling and abnormally activates inflammation in both animal models. Both ASD probands carrying Scgn mutations also show reduced oxytocin levels. Importantly, we demonstrate that the administration of oxytocin and anti-inflammatory drugs can attenuate ASD-associated defects caused by SCGN deficiency. Altogether, we identify a convergence between a potential autism genetic risk factor SCGN, and the pathological deregulation in oxytocinergic signaling and immune responses, providing potential treatment for ASD patients suffering from SCGN deficiency. Our study also indicates that it is critical to identify and stratify ASD patient populations based on their disease mechanisms, which could greatly enhance therapeutic success.
Collapse
|
6
|
ox-LDL induces autophagy-mediated apoptosis by suppressing secretagogin-regulated autophagic flux in pancreatic β-cells. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1822-1831. [PMID: 36789686 PMCID: PMC10157621 DOI: 10.3724/abbs.2022186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lipotoxicity has been shown to induce the loss of functional β-cell mass and lead to type 2 diabetes, but the mechanism remains unknown. In this study, we aim to explore the role of secretagogin (SCGN) in lipotoxicity-induced β-cell injury. Our results indicate that ox-LDL treatment leads to autophagic cell death, as evidenced by decreased cell viability, aggravated cell apoptosis, and the accumulation of the p62 protein in MIN6 cells. LysoTracker Red staining, TEM and mRFP-GFP-LC3 assays demonstrate that autophagic flux is blocked in ox-LDL-treated MIN6 cells. Intriguingly, SCGN is significantly decreased in MIN6 cells under lipotoxic conditions. Additionally, siRNA-guided SCGN knockdown blocks autophagic flux triggered by rapamycin, while SCGN restoration alleviates autophagic flux retardation and mitigates cell apoptosis. The physical interaction between SCGN and SNAP29 is validated by bioinformatics analysis, coimmunoprecipitation assay and SCGN knockdown test. Downregulation of SCGN expression reduces the interaction of these two proteins. Taken together, our results indicate that ox-LDL treatment induces apoptotic β-cell death by blocking autophagic flux dependent on SCGN downregulation. SCGN administration prevents lipotoxic β-cell injury and may be a potential therapeutic strategy to promote β-cell expansion in type 2 diabetes.
Collapse
|
7
|
Biancolin AD, Srikrishnaraj A, Jeong H, Martchenko A, Brubaker PL. The Cytoskeletal Transport Protein, Secretagogin, Is Essential for Diurnal Glucagon-like Peptide-1 Secretion in Mice. Endocrinology 2022; 163:6678475. [PMID: 36036556 DOI: 10.1210/endocr/bqac142] [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: 07/15/2022] [Indexed: 11/19/2022]
Abstract
The intestinal L-cell incretin, glucagon-like peptide-1 (GLP-1), exhibits a circadian pattern of secretion, thereby entraining diurnal insulin release. Secretagogin (Scgn), an actin-binding regulatory protein, is essential for the temporal peak of GLP-1 secretion in vitro. To interrogate the role of Scgn in diurnal GLP-1 secretion in vivo, peak and trough GLP-1 release were evaluated in knockout mice (Scgn-/-, Gcg-CreERT2/+; Scgnfl/fl and Vil-CreERT2/+; Scgnfl/fl), and RNA sequencing (RNA-Seq) was conducted in Scgn knockdown L-cells. All 3 knockout models demonstrated loss of the diurnal rhythm of GLP-1 secretion in response to oral glucose. Gcg-CreERT2/+; Scgnfl/fl mice also lost the normal pattern in glucagon secretion, while Scgn-/- and Vil-CreERT2/+; Scgnfl/fl animals demonstrated impaired diurnal secretion of the related incretin, glucose-dependent insulinotrophic polypeptide. RNA-Seq of mGLUTag L-cells showed decreased pathways regulating vesicle transport, transport and binding, and protein-protein interaction at synapse, as well as pathways related to proteasome-mediated degradation including chaperone-mediated protein complex assembly following Scgn knockdown. Scgn is therefore essential for diurnal L-cell GLP-1 secretion in vivo, likely mediated through effects on secretory granule dynamics.
Collapse
Affiliation(s)
| | - Arjuna Srikrishnaraj
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hyerin Jeong
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Alexandre Martchenko
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Patricia Lee Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
8
|
A set of common buccal CpGs that predict epigenetic age and associate with lifespan-regulating genes. iScience 2022; 25:105304. [PMID: 36304118 PMCID: PMC9593711 DOI: 10.1016/j.isci.2022.105304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 10/02/2022] [Indexed: 11/23/2022] Open
Abstract
Epigenetic aging clocks are computational models that use DNA methylation sites to predict age. Since cheek swabs are non-invasive and painless, collecting DNA from buccal tissue is highly desirable. Here, we review 11 existing clocks that have been applied to buccal tissue. Two of these were exclusively trained on adults and, while moderately accurate, have not been used to capture health-relevant differences in epigenetic age. Using 130 common CpGs utilized by two or more existing buccal clocks, we generate a proof-of-concept predictor in an adult methylomic dataset. In addition to accurately estimating age (r = 0.95 and mean absolute error = 3.88 years), this clock predicted that Down syndrome subjects were significantly older relative to controls. A literature and database review of CpG-associated genes identified numerous genes (e.g., CLOCK, ELOVL2, and VGF) and molecules (e.g., alpha-linolenic acid, glycine, and spermidine) reported to influence lifespan and/or age-related disease in model organisms. 130 CpGs have been used by two or more aging clocks applied to human buccal tissue Common CpG genes are linked to the adaptive immune system and telomere maintenance Common CpGs can be used to build a novel, proof-of-concept epigenetic aging clock Several compounds associated with common CpG genes regulate lifespan in animals
Collapse
|
9
|
Chen CW, Guan BJ, Alzahrani MR, Gao Z, Gao L, Bracey S, Wu J, Mbow CA, Jobava R, Haataja L, Zalavadia AH, Schaffer AE, Lee H, LaFramboise T, Bederman I, Arvan P, Mathews CE, Gerling IC, Kaestner KH, Tirosh B, Engin F, Hatzoglou M. Adaptation to chronic ER stress enforces pancreatic β-cell plasticity. Nat Commun 2022; 13:4621. [PMID: 35941159 PMCID: PMC9360004 DOI: 10.1038/s41467-022-32425-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic β-cells are prone to endoplasmic reticulum (ER) stress due to their role in insulin secretion. They require sustainable and efficient adaptive stress responses to cope with this stress. Whether episodes of chronic stress directly compromise β-cell identity is unknown. We show here under reversible, chronic stress conditions β-cells undergo transcriptional and translational reprogramming associated with impaired expression of regulators of β-cell function and identity. Upon recovery from stress, β-cells regain their identity and function, indicating a high degree of adaptive plasticity. Remarkably, while β-cells show resilience to episodic ER stress, when episodes exceed a threshold, β-cell identity is gradually lost. Single cell RNA-sequencing analysis of islets from type 1 diabetes patients indicates severe deregulation of the chronic stress-adaptation program and reveals novel biomarkers of diabetes progression. Our results suggest β-cell adaptive exhaustion contributes to diabetes pathogenesis.
Collapse
Affiliation(s)
- Chien-Wen Chen
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Bo-Jhih Guan
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mohammed R Alzahrani
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Zhaofeng Gao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Long Gao
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Syrena Bracey
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jing Wu
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cheikh A Mbow
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Raul Jobava
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Leena Haataja
- The Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI, 48105, USA
| | - Ajay H Zalavadia
- Lerner Research Institute, Cleveland Clinic, 9620 Carnegie Ave N Bldg, Cleveland, OH, 44106, US
| | - Ashleigh E Schaffer
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peter Arvan
- The Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI, 48105, USA
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, US
| | - Ivan C Gerling
- Department of Medicine, University of Tennessee, Memphis, TN, US
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Boaz Tirosh
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA.
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
10
|
Chidananda AH, Khandelwal R, Jhamkhindikar A, Pawar AD, Sharma AK, Sharma Y. Secretagogin is a Ca 2+-dependent stress-responsive chaperone that may also play a role in aggregation-based proteinopathies. J Biol Chem 2022; 298:102285. [PMID: 35870554 PMCID: PMC9425029 DOI: 10.1016/j.jbc.2022.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Secretagogin (SCGN) is a three-domain hexa-EF-hand Ca2+-binding protein that plays a regulatory role in the release of several hormones. SCGN is expressed largely in pancreatic β-cells, certain parts of the brain, and also in neuroendocrine tissues. The expression of SCGN is altered in several diseases, such as diabetes, cancers, and neurodegenerative disorders; however, the precise associations that closely link SCGN expression to such pathophysiologies are not known. In this work, we report that SCGN is an early responder to cellular stress, and SCGN expression is temporally upregulated by oxidative stress and heat shock. We show the overexpression of SCGN efficiently prevents cells from heat shock and oxidative damage. We further demonstrate that in the presence of Ca2+, SCGN efficiently prevents the aggregation of a broad range of model proteins in vitro. Small-angle X-ray scattering (BioSAXS) studies further reveal that Ca2+ induces the conversion of a closed compact apo-SCGN conformation into an open extended holo-SCGN conformation via multistate intermediates, consistent with the augmentation of chaperone activity of SCGN. Furthermore, isothermal titration calorimetry establishes that Ca2+ enables SCGN to bind α-synuclein and insulin, two target proteins of SCGN. Altogether, our data not only demonstrate that SCGN is a Ca2+-dependent generic molecular chaperone involved in protein homeostasis with broad substrate specificity but also elucidate the origin of its altered expression in several cancers. We describe a plausible mechanism of how perturbations in Ca2+ homeostasis and/or deregulated SCGN expression would hasten the process of protein misfolding, which is a feature of many aggregation-based proteinopathies.
Collapse
Affiliation(s)
- Amrutha H Chidananda
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad-500 007, India
| | - Radhika Khandelwal
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad-500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Aditya Jhamkhindikar
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad-500 007, India
| | - Asmita D Pawar
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad-500 007, India; Indian Institute of Scientific and Education Research (IISER), Berhampur-760010, India
| | - Anand K Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad-500 007, India.
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad-500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India; Indian Institute of Scientific and Education Research (IISER), Berhampur-760010, India.
| |
Collapse
|
11
|
Basu S, Mitra S, Singh O, Chandramohan B, Singru PS. Secretagogin in the brain and pituitary of the catfish, Clarias batrachus: Molecular characterization and regulation by insulin. J Comp Neurol 2022; 530:1743-1772. [PMID: 35322425 DOI: 10.1002/cne.25311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/12/2022]
Abstract
Secretagogin (scgn), is a novel hexa EF-hand, phylogenetically conserved calcium-binding protein. It serves as Ca2+ sensor and participates in Ca2+ -signaling and neuroendocrine regulation in mammals. However, its relevance in the brain of non-mammalian vertebrates has largely remained unexplored. To address this issue, we studied the cDNA encoding scgn, scgn mRNA expression, and distribution of scgn-equipped elements in the brain and pituitary of a teleost, Clarias batrachus (cb). The cbscgn cDNA consists of three transcripts (T) variants: T1 (2185 bp), T2 (2151 bp) and T3 (2060 bp). While 816 bp ORF in T1 and T2 encodes highly conserved six EF-hand 272 aa protein fully capable of Ca2+ -binding, 726-bp ORF in T3 encodes 242 aa protein. The T1 showed >90% and >70% identity with scgn of catfishes, and other teleosts and mammals, respectively. The T1-mRNA was widely expressed in the brain and pituitary, while the expression of T3 was restricted to the telencephalon. Application of the anti-scgn antiserum revealed a ∼32 kDa scgn-immunoreactive (scgn-i) band (known molecular weight of scgn) in the forebrain tissue, and immunohistochemically labeled neurons in the olfactory epithelium and bulb, telencephalon, preoptic area, hypothalamus, thalamus, and hindbrain. In the pituitary, scgn-i cells were seen in the pars distalis and intermedia. Insulin is reported to regulate scgn mRNA in the mammalian hippocampus, and feeding-related neuropeptides in the telencephalon of teleost. Intracranial injection of insulin significantly increased T1-mRNA expression and scgn-immunoreactivity in the telencephalon. We suggest that scgn may be an important player in the regulation of olfactory, neuroendocrine system, and energy balance functions in C. batrachus.
Collapse
Affiliation(s)
- Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Bathrachalam Chandramohan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
12
|
Shahal T, Segev E, Konstantinovsky T, Marcus Y, Shefer G, Pasmanik-Chor M, Buch A, Ebenstein Y, Zimmet P, Stern N. Deconvolution of the epigenetic age discloses distinct inter-personal variability in epigenetic aging patterns. Epigenetics Chromatin 2022; 15:9. [PMID: 35255955 PMCID: PMC8900303 DOI: 10.1186/s13072-022-00441-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The epigenetic age can now be extrapolated from one of several epigenetic clocks, which are based on age-related changes in DNA methylation levels at specific multiple CpG sites. Accelerated aging, calculated from the discrepancy between the chronological age and the epigenetic age, has shown to predict morbidity and mortality rate. We assumed that deconvolution of epigenetic age to its components could shed light on the diversity of epigenetic, and by inference, on inter-individual variability in the causes of biological aging. RESULTS Using the Horvath original epigenetic clock, we identified several CpG sites linked to distinct genes that quantitatively explain much of the inter-personal variability in epigenetic aging, with CpG sites related to secretagogin and malin being the most variable. We show that equal epigenetic age in different subjects can result from variable contribution size of the same CpG sites to the total epigenetic age. In a healthy cohort, the most variable CpG sites are responsible for accelerated and decelerated epigenetic aging, relative to chronological age. CONCLUSIONS Of the 353 CpG sites that form the basis for the Horvath epigenetic age, we have found the CpG sites that are responsible for accelerated and decelerated epigenetic aging in healthy subjects. However, the relative contribution of each site to aging varies between individuals, leading to variable personal aging patterns. Our findings pave the way to form personalized aging cards allowing the identification of specific genes related to CpG sites, as aging markers, and perhaps treatment of these targets in order to hinder undesirable age drifting.
Collapse
Affiliation(s)
- Tamar Shahal
- The Sagol Center for Epigenetics of Aging and Metabolism, Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Elad Segev
- Department of Applied Mathematics, Holon Institute of Technology, Holon, Israel
| | - Thomas Konstantinovsky
- The Sagol Center for Epigenetics of Aging and Metabolism, Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Applied Mathematics, Holon Institute of Technology, Holon, Israel
| | - Yonit Marcus
- The Sagol Center for Epigenetics of Aging and Metabolism, Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Gabi Shefer
- The Sagol Center for Epigenetics of Aging and Metabolism, Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, The George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Assaf Buch
- The Sagol Center for Epigenetics of Aging and Metabolism, Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Ebenstein
- Department of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Paul Zimmet
- Department of Diabetes, Monash University School of Medicine, Melbourne, Australia
| | - Naftali Stern
- The Sagol Center for Epigenetics of Aging and Metabolism, Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,The Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| |
Collapse
|
13
|
Khandelwal R, Sharma AK, Biswa BB, Sharma Y. Extracellular Secretagogin is internalized into the cells through endocytosis. FEBS J 2021; 289:3183-3204. [PMID: 34967502 DOI: 10.1111/febs.16338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 11/29/2022]
Abstract
Secretagogin (SCGN) is a calcium-sensor protein with a regulatory role in glucose metabolism and the secretion of several peptide hormones. Many, but not all, functions of SCGN can be explained by its intracellular manifestation. Despite early data on SCGN secretion, the secretory mechanism, biological fate, physiological implications, and trans-cellular signaling of extracellular SCGN remain unknown. We here report that extracellular SCGN is readily internalized into the C2C12 cells in an energy-dependent manner. Using endocytosis inhibitors, we demonstrate that SCGN internalizes via clathrin-mediated endocytosis, following which, SCGN localizes largely in the cytosol. Exogenous SCGN treatment induces a global proteomic reprogramming in C2C12 cells. Gene ontology search suggests that SCGN-induced proteomic reprogramming favors protein synthesis and cellular growth. We thus validated the cell proliferative action of SCGN using C2C12, HepG2, and NIH-3T3 cell lines. Based on the data, we propose that circulatory SCGN is internalized into the target cells and modulates the expression of cell growth-related proteins. The work suggests that extracellular SCGN is a functional protein, which communicates with specific cell types and directly modulates cell proliferation.
Collapse
Affiliation(s)
- Radhika Khandelwal
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Anand Kumar Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, 500 007, India
| | - Bhim B Biswa
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, 500 007, India
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.,Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, 760010, India
| |
Collapse
|
14
|
Wu L, Lv Y, Lv Y, Xiang S, Zhao Z, Tang Z, Ou L, Yan B, Xiao X, Wen G, Cao R, Yang J. A novel secretagogin/ATF4 pathway is involved in oxidized LDL-induced endoplasmic reticulum stress and islet β-cell apoptosis. Acta Biochim Biophys Sin (Shanghai) 2021; 53:54-62. [PMID: 33289795 DOI: 10.1093/abbs/gmaa142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Indexed: 12/16/2022] Open
Abstract
Excessive accumulation of cholesterol in β cells initiates endoplasmic reticulum (ER) stress and associated apoptosis. We have reported that excessive uptake of cholesterol by MIN6 cells decreases the expression of secretagogin (SCGN) and then attenuates insulin secretion. Here, we aimed to determine whether cholesterol-induced SCGN decrease is involved in the modulation of ER stress and apoptosis in pancreatic β cells. In this study, MIN6 cells were treated with oxidized low-density lipoprotein (ox-LDL) for 24 h, and then intracellular lipid droplets and cell apoptosis were quantified, and SCGN and ER stress markers were identified by western blot analysis. Furthermore, small interfer RNA (siRNA)-mediated SCGN knockdown and recombinant plasmid-mediated SCGN restoration experiments were performed to confirm the role of SCGN in ER stress and associated cell apoptosis. Finally, the interaction of SCGN with ATF4 was computationally predicted and then validated by a co-immunoprecipitation assay. We found that ox-LDL treatment increased the levels of ER stress markers, such as phosphorylated protein kinase-like endoplasmic reticulum kinase, phosphorylated eukaryotic initiation factor 2 alpha, activating transcription factor 4 (ATF4), and transcription factor CCAAT-enhancer-binding protein homologous protein, and promoted MIN6 cell apoptosis; in addition, the expression of SCGN was downregulated. siRNA-mediated SCGN knockdown exacerbated β-cell ER stress by increasing ATF4 expression. Pretreatment of MIN6 cells with the recombinant SCGN partly antagonized ox-LDL-induced ER stress and apoptosis. Furthermore, a co-immunoprecipitation assay revealed an interaction between SCGN and ATF4 in MIN6 cells. Taken together, these results demonstrated that pancreatic β-cell apoptosis induced by ox-LDL treatment can be attributed, in part, to an SCGN/ATF4-dependent ER stress response.
Collapse
Affiliation(s)
- Li Wu
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Yuncheng Lv
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541199, China
| | - Ying Lv
- Department of Metabolism & Endocrinology, The Second Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Sunmin Xiang
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Zhibo Zhao
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Ziqing Tang
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Linling Ou
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Bin Yan
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Xinhua Xiao
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Gebo Wen
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Renxian Cao
- Department of Clinical Research, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| | - Jing Yang
- Department of Metabolism & Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang 421001, China
| |
Collapse
|
15
|
Lv Y, Xiang S, Cao R, Wu L, Yang J. SCGN-regulated Stage-wise SNARE Assembly: Novel Insight into Synaptic Exocytosis. Neurosci Bull 2020; 36:1576-1578. [PMID: 33025412 DOI: 10.1007/s12264-020-00590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 07/19/2020] [Indexed: 10/23/2022] Open
Affiliation(s)
- Ying Lv
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of the University of South China, Hengyang, 421001, China
| | - Sunmin Xiang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang, 421001, China
| | - Renxian Cao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang, 421001, China
| | - Li Wu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang, 421001, China
| | - Jing Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of the University of South China, Hengyang, 421001, China.
| |
Collapse
|
16
|
Daryabor G, Atashzar MR, Kabelitz D, Meri S, Kalantar K. The Effects of Type 2 Diabetes Mellitus on Organ Metabolism and the Immune System. Front Immunol 2020; 11:1582. [PMID: 32793223 PMCID: PMC7387426 DOI: 10.3389/fimmu.2020.01582] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic abnormalities such as dyslipidemia, hyperinsulinemia, or insulin resistance and obesity play key roles in the induction and progression of type 2 diabetes mellitus (T2DM). The field of immunometabolism implies a bidirectional link between the immune system and metabolism, in which inflammation plays an essential role in the promotion of metabolic abnormalities (e.g., obesity and T2DM), and metabolic factors, in turn, regulate immune cell functions. Obesity as the main inducer of a systemic low-level inflammation is a main susceptibility factor for T2DM. Obesity-related immune cell infiltration, inflammation, and increased oxidative stress promote metabolic impairments in the insulin-sensitive tissues and finally, insulin resistance, organ failure, and premature aging occur. Hyperglycemia and the subsequent inflammation are the main causes of micro- and macroangiopathies in the circulatory system. They also promote the gut microbiota dysbiosis, increased intestinal permeability, and fatty liver disease. The impaired immune system together with metabolic imbalance also increases the susceptibility of patients to several pathogenic agents such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Thus, the need for a proper immunization protocol among such patients is granted. The focus of the current review is to explore metabolic and immunological abnormalities affecting several organs of T2DM patients and explain the mechanisms, whereby diabetic patients become more susceptible to infectious diseases.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohamad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Seppo Meri
- Department of Bacteriology and Immunology and the Translational Immunology Research Program (TRIMM), The University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Secretagogin is Related to Insulin Secretion but Unrelated to Gestational Diabetes Mellitus Status in Pregnancy. J Clin Med 2020; 9:jcm9072277. [PMID: 32708966 PMCID: PMC7408624 DOI: 10.3390/jcm9072277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
Secretagogin (SCGN) is a calcium binding protein related to insulin release in the pancreas. Although SCGN is not co-released with insulin, plasma concentrations have been found to be increased in type 2 diabetes mellitus patients. Until now, no study on SCGN levels in pregnancy or patients with gestational diabetes mellitus (GDM) has been published. In 93 women of a high-risk population for GDM at the Medical University of Vienna, secretagogin levels of 45 GDM patients were compared to 48 women with a normal glucose tolerance (NGT). Glucose tolerance, insulin resistance and secretion were assessed with oral glucose tolerance tests (OGTT) between the 10th and 28th week of gestation (GW) and postpartum. In all women, however, predominantly in women with NGT, there was a significant positive correlation between SCGN levels and Stumvoll first (rp = 0.220, p = 0.032) and second phase index (rp = 0.224, p = 0.028). SCGN levels were not significantly different in women with NGT and GDM. However, SCGN was higher postpartum than during pregnancy (postpartum: 88.07 ± 35.63 pg/mL; pregnancy: 75.24 ± 37.90 pg/mL, p = 0.004). SCGN was directly correlated with week of gestation (rp = 0.308; p = 0.021) and triglycerides (rp = 0.276; p = 0.038) in women with GDM. Therefore, SCGN is related to insulin secretion and hyperinsulinemia during pregnancy; however, it does not display differences between women with NGT and GDM.
Collapse
|
18
|
Chidananda AH, Sharma AK, Khandelwal R, Sharma Y. Secretagogin Binding Prevents α-Synuclein Fibrillation. Biochemistry 2019; 58:4585-4589. [DOI: 10.1021/acs.biochem.9b00656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Amrutha H. Chidananda
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
| | - Anand Kumar Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
| | - Radhika Khandelwal
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Indian Institute of Science Education and Research (IISER), Berhampur 760010, India
| |
Collapse
|