1
|
Gao Y, Chen Q, Wu Z, Yuan L. Regulation of pancreatic β cells by exosomes from different sources. Diabetes Res Clin Pract 2025; 224:112222. [PMID: 40324722 DOI: 10.1016/j.diabres.2025.112222] [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: 02/26/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Diabetes is a chronic metabolic disorder with rising global prevalence, particularly in developed and high-income regions. Central to its pathogenesis is the dysfunction of pancreatic β-cells, alongside impaired glucose and lipid metabolism in peripheral insulin-responsive tissues. Exosomes are nano-sized extracellular vesicles essential for intercellular communication and have emerged as pivotal regulators of metabolic homeostasis. Secreted by virtually all cell types, exosomes encapsulate bioactive cargo that reflects their cellular origin and physiological state, thereby exerting diverse functional effects. Recent evidence highlights the role of exosomes derived from the liver, gut, adipose tissue, skeletal muscle, and mesenchymal stem cells in modulating β-cell proliferation, insulin secretion, and survival. In peripheral tissues exosomes also influence insulin sensitivity by regulating glucose and lipid metabolism, ultimately shaping β-cell responses under hyperglycemic conditions. A more comprehensive understanding of exosome-mediated crosstalk between metabolic organs and pancreatic β-cells could pave the way for the development of exosome-based diagnostic tools and therapeutic strategies aimed at improving early detection, prevention, and treatment of the diabetes.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhuoying Wu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
2
|
Dekkers MC, Pu X, Enciso-Martinez A, Zaldumbide A. Beta-Cell-Derived Extracellular Vesicles: Mediators of Intercellular Communication in the Islet Microenvironment in Type 1 Diabetes. Cells 2024; 13:1996. [PMID: 39682744 PMCID: PMC11640590 DOI: 10.3390/cells13231996] [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: 10/19/2024] [Revised: 11/23/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder characterised by an autoimmune response specifically mounted against the insulin-producing beta cells. Within the islet, high cellular connectivity and extensive vascularisation facilitate intra-islet communication and direct crosstalk with the surrounding tissues and the immune system. During the development of T1D, cytokines and extracellular vesicles released by beta cells can contribute to the recruitment of immune cells, further amplifying autoimmunity and aggravating beta cell damage and dysfunction. In this review, we will evaluate the role of beta-cell-derived extracellular vesicles as mediators of the autoimmune response and discuss their potential for early diagnosis and new therapeutic strategies in T1D.
Collapse
Affiliation(s)
- Mette C. Dekkers
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.C.D.); (X.P.); (A.E.-M.)
| | - Xudong Pu
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.C.D.); (X.P.); (A.E.-M.)
| | - Agustin Enciso-Martinez
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.C.D.); (X.P.); (A.E.-M.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Vesicle Center, Biomedical Engineering and Physics and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.C.D.); (X.P.); (A.E.-M.)
| |
Collapse
|
3
|
Yu CC, Yang CY, Chang TY, Lan KC, Liu SH. A negative regulatory role of β-cell-derived exosomes in the glucose-stimulated insulin secretion of recipient β-cells. Arch Toxicol 2024; 98:3885-3896. [PMID: 39127846 DOI: 10.1007/s00204-024-03838-8] [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: 06/21/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Exosomes are extracellular vesicles that play a role in intercellular communication through the transportation of their cargo including mRNAs, microRNAs, proteins, and nucleic acids. Exosomes can also regulate glucose homeostasis and insulin secretion under diabetic conditions. However, the role of exosomes in insulin secretion in islet β-cells under physiological conditions remains to be clarified. The aim of this study was to investigate whether exosomes derived from pancreatic islet β-cells could affect insulin secretion in naïve β-cells. We first confirmed that exosomes derived from the RIN-m5f β-cell line interfered with the glucose-stimulated insulin secretion (GSIS) of recipient β-cells without affecting cell viability. The exosomes significantly reduced the protein expression levels of phosphorylated Akt, phosphorylated GSK3α/β, CaMKII, and GLUT2 (insulin-related signaling molecules), and they increased the protein expression levels of phosphorylated NFκB-p65 and Cox-2 (inflammation-related signaling molecules), as determined by a Western blot analysis. A bioinformatics analysis of Next-Generation Sequencing data suggested that exosome-carried microRNAs, such as miR-1224, -122-5p, -133a-3p, -10b-5p, and -423-5p, may affect GSIS in recipient β-cells. Taken together, these findings suggest that β-cell-derived exosomes may upregulate exosomal microRNA-associated signals to dysregulate glucose-stimulated insulin secretion in naïve β-cells.
Collapse
Affiliation(s)
- Chia-Ching Yu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Yao Yang
- Department of Surgery, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Ting-Yu Chang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
4
|
Guo M, Huang X, Zhang J, Huang Y, Tang Y, Wen H, Xu Y, Zhang S, Wei X, Sun S, Zhu Q. Palmitic acid induces β-cell ferroptosis by activating ceramide signaling pathway. Exp Cell Res 2024; 440:114134. [PMID: 38901790 DOI: 10.1016/j.yexcr.2024.114134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/02/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
Individuals with type 2 diabetes mellitus frequently display heightened levels of palmitic acid (PA) in their serum, which may lead to β-cell damage. The involvement of ferroptosis, a form of oxidative cell death in lipotoxic β-cell injury remains uncertain. Here, we have shown that PA induces intracellular lipid peroxidation, increases intracellular Fe2+ content and decreases intracellular glutathione peroxidase 4 (GPX4) expression. Furthermore, PA causes distinct changes in pancreatic islets and INS-1 cells, such as mitochondrial atrophy and increased membrane density. Furthermore, the presence of the ferroptosis inhibitor has a significant mitigating effect on PA-induced β-cell damage. Mechanistically, PA increased ceramide content and c-Jun N-terminal kinase (JNK) phosphorylation. The ceramide synthase inhibitor effectively attenuated PA-induced β-cell damage and GPX4/Fe2+ abnormalities, while inhibiting JNK phosphorylation. Additionally, the JNK inhibitor SP600125 improved PA-induced cell damage. In conclusion, by promoting ceramide synthesis, PA inhibited GPX4 expression and increased intracellular Fe2+ to induce β-cell ferroptosis. Moreover, JNK may be a downstream mechanism of ceramide-triggered lipotoxic ferroptosis in β-cells.
Collapse
Affiliation(s)
- Maojun Guo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Xiaolong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Junhan Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Ying Huang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Ying Tang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Honghua Wen
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Yanan Xu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China; Department of Endocrinology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, 222002, China
| | - Shaokun Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China; Department of Infectious Diseases, Taizhou Second People's Hospital, Taizhou, Jiangsu, 225500, China
| | - Xiao Wei
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Shuoshuo Sun
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qun Zhu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China.
| |
Collapse
|
5
|
Zhang Z, Tang L, Guo Y, Guo X, Pan Z, Ji X, Gao C. Development of Biomarkers and Prognosis Model of Mortality Risk in Patients with COVID-19. J Inflamm Res 2024; 17:2445-2457. [PMID: 38681069 PMCID: PMC11048291 DOI: 10.2147/jir.s449497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024] Open
Abstract
Background As of 30 April 2023, the COVID-19 pandemic has resulted in over 6.9 million deaths worldwide. The virus continues to spread and mutate, leading to continuously evolving pathological and physiological processes. It is imperative to reevaluate predictive factors for identifying the risk of early disease progression. Methods A retrospective study was conducted on a cohort of 1379 COVID-19 patients who were discharged from Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine between 15 December 2022 and 15 February 2023. Patient symptoms, comorbidities, demographics, vital signs, and laboratory test results were systematically documented. The dataset was split into testing and training sets, and 15 different machine learning algorithms were employed to construct prediction models. These models were assessed for accuracy and area under the receiver operating characteristic curve (AUROC), and the best-performing model was selected for further analysis. Results AUROC for models generated by 15 machine learning algorithms all exceeded 90%, and the accuracy of 10 of them also surpassed 90%. Light Gradient Boosting model emerged as the optimal choice, with accuracy of 0.928 ± 0.0006 and an AUROC of 0.976 ± 0.0028. Notably, the factors with the greatest impact on in-hospital mortality were growth stimulation expressed gene 2 (ST2,19.3%), interleukin-8 (IL-8,17.2%), interleukin-6 (IL-6,6.4%), age (6.1%), NT-proBNP (5.1%), interleukin-2 receptor (IL-2R, 5%), troponin I (TNI,4.6%), congestive heart failure (3.3%) in Light Gradient Boosting model. Conclusion ST-2, IL-8, IL-6, NT-proBNP, IL-2R, TNI, age and congestive heart failure were significant predictors of in-hospital mortality among COVID-19 patients.
Collapse
Affiliation(s)
- Zhishuo Zhang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lujia Tang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yiran Guo
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xin Guo
- School of Information Science and Technology, Sanda University, Shanghai, Pudong District, 201209, China
| | - Zhiying Pan
- School of Information Science and Technology, Sanda University, Shanghai, Pudong District, 201209, China
| | - Xiaojing Ji
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
6
|
Xu J, Harris-Kawano A, Enriquez JR, Mirmira RG, Sims EK. Proinflammatory stress activates neutral sphingomyelinase 2 based generation of a ceramide-enriched β cell EV subpopulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589943. [PMID: 38659945 PMCID: PMC11042299 DOI: 10.1101/2024.04.17.589943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
β cell extracellular vesicles (EVs) play a role as paracrine effectors in islet health, yet mechanisms connecting β cell stress to changes in EV cargo and potential impacts on diabetes remain poorly defined. We hypothesized that β cell inflammatory stress engages neutral sphingomyelinase 2 (nSMase2)-dependent EV formation pathways, generating ceramide-enriched EVs that could impact surrounding β cells. Consistent with this, proinflammatory cytokine treatment of INS-1 β cells and human islets concurrently increased β cell nSMase2 and ceramide expression, as well as EV ceramide staining. Direct chemical activation or genetic knockdown of nSMase2, or treatment with a GLP-1 receptor agonist also modulated cellular and EV ceramide. Small RNA sequencing of ceramide-enriched EVs identified a distinct set of miRNAs linked to β cell function and identity. Coculture experiments using CD9-GFP tagged INS-1 cell EVs demonstrated that either cytokine treatment or chemical nSMase2 activation increased EV transfer to recipient cells. Children with recent-onset T1D showed no abnormalities in circulating ceramide-enriched EVs, suggesting a localized, rather than systemic phenomenon. These findings highlight nSMase2 as a regulator of β cell EV cargo and identify ceramide-enriched EV populations as a contributor to EV-related paracrine signaling under conditions of β cell inflammatory stress. Article Highlights a. Why did we undertake this study?: Mechanisms connecting β cell stress to changes in extracellular vesicle (EV) cargo and potential impacts on diabetes are poorly defined.b. What is the specific question we wanted to answer?: Does β cell inflammatory stress engage neutral sphingomyelinase 2 (nSMase2)-dependent EV formation pathways to generate ceramide-enriched EVs.c. What did we find?: Proinflammatory cytokine treatment of β cells increased β cell ceramide expression, along with EV ceramide in part via increases in nSMase2. Ceramide-enriched EVs housed a distinct set of miRNAs linked to insulin signaling. Both cytokine treatment and nSMase2 activation increase EV transfer to other β cells.d. What are the implications of our findings?: Our findings highlight nSMase2 as a regulator of β cell EV cargo and identify ceramide-enriched EV populations as a contributor to EV-related paracrine signaling under conditions of β cell inflammatory stress.
Collapse
|
7
|
Lakey JRT, Wang Y, Alexander M, Chan MKS, Wong MBF, Casazza K, Jenkins I. Exosomes; a Potential Source of Biomarkers, Therapy, and Cure for Type-1 Diabetes. Int J Mol Sci 2023; 24:15713. [PMID: 37958696 PMCID: PMC10647572 DOI: 10.3390/ijms242115713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
The scourge of type-1 diabetes (T1D) is the morbidity and mortality it and its complications cause at a younger age. This propels the constant search for better diagnostic, treatment, and management strategies, with the ultimate quest being a cure for T1D. Recently, the therapeutic potential of exosomes has generated a lot of interest. Among the characteristics of exosomes of particular interest are (a) their regenerative capacity, which depends on their "origin", and (b) their "content", which determines the cell communication and crosstalk they influence. Other functional capacities, including paracrine and endocrine homeostatic regulation, pathogenic response ability resulting in insulin secretory defects or β-cell death under normal metabolic conditions, immunomodulation, and promotion of regeneration, have also garnered significant interest. Exosome "specificity" makes them suitable as biomarkers or predictors, and their "mobility" and "content" lend credence to drug delivery and therapeutic suitability. This review aims to highlight the functional capacities of exosomes and their established as well as novel contributions at various pathways in the onset and progression of T1D. The pathogenesis of T1D involves a complex crosstalk between insulin-secreting pancreatic β-cells and immune cells, which is partially mediated by exosomes. We also examine the potential implications for type 2 diabetes (T2D), as the link in T2D has guided T1D exploration. The collective landscape presented is expected to help identify how a deeper understanding of exosomes (and their cargo) can provide a framework for actionable solutions to prevent, halt, or change the very course of T1D and its complications.
Collapse
Affiliation(s)
- Jonathan R. T. Lakey
- Department of Surgery, University of California Irvine, Irvine, CA 92617, USA;
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - Yanmin Wang
- California Medical Innovations Institute, 11107 Roselle Street, San Diego, CA 92121, USA;
| | - Michael Alexander
- Department of Surgery, University of California Irvine, Irvine, CA 92617, USA;
| | - Mike K. S. Chan
- Uropean Wellness Group, Klosterstrasse 205ID, 67480 Edenkoben, Germany; (M.K.S.C.); (M.B.F.W.)
- Baden R&D Laboratories GmbH, z Hd.v. Sabine Conrad, Ferdinand-Lassalle-Strasse 40, 72770 Reutlingen, Germany
| | - Michelle B. F. Wong
- Uropean Wellness Group, Klosterstrasse 205ID, 67480 Edenkoben, Germany; (M.K.S.C.); (M.B.F.W.)
- Baden R&D Laboratories GmbH, z Hd.v. Sabine Conrad, Ferdinand-Lassalle-Strasse 40, 72770 Reutlingen, Germany
| | - Krista Casazza
- GATC Health Inc., Suite 600, 2030 Main Street, Irvine, CA 92718, USA; (K.C.); (I.J.)
| | - Ian Jenkins
- GATC Health Inc., Suite 600, 2030 Main Street, Irvine, CA 92718, USA; (K.C.); (I.J.)
| |
Collapse
|
8
|
Wei J, Wang Z, Han T, Chen J, Ou Y, Wei L, Zhu X, Wang K, Yan Z, Han YP, Zheng X. Extracellular vesicle-mediated intercellular and interorgan crosstalk of pancreatic islet in health and diabetes. Front Endocrinol (Lausanne) 2023; 14:1170237. [PMID: 37305058 PMCID: PMC10248434 DOI: 10.3389/fendo.2023.1170237] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Diabetes mellitus (DM) is a systemic metabolic disease with high mortality and morbidity. Extracellular vesicles (EVs) have emerged as a novel class of signaling molecules, biomarkers and therapeutic agents. EVs-mediated intercellular and interorgan crosstalk of pancreatic islets plays a crucial role in the regulation of insulin secretion of β-cells and insulin action in peripheral insulin target tissues, maintaining glucose homeostasis under physiological conditions, and it's also involved in pathological changes including autoimmune response, insulin resistance and β-cell failure associated with DM. In addition, EVs may serve as biomarkers and therapeutic agents that respectively reflect the status and improve function and viability of pancreatic islets. In this review, we provide an overview of EVs, discuss EVs-mediated intercellular and interorgan crosstalk of pancreatic islet under physiological and diabetic conditions, and summarize the emerging applications of EVs in the diagnosis and treatment of DM. A better understanding of EVs-mediated intercellular and interorgan communication of pancreatic islets will broaden and enrich our knowledge of physiological homeostasis maintenance as well as the development, diagnosis and treatment of DM.
Collapse
Affiliation(s)
- Junlun Wei
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenghao Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institute, Stockholm, Sweden
| | - Tingrui Han
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaoting Chen
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yiran Ou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Wei
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyue Zhu
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Ke Wang
- Department of Vascular Surgery, University Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhe Yan
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, The College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Kim H, Lichtenstein AH, Ganz P, Du S, Tang O, Yu B, Chatterjee N, Appel LJ, Coresh J, Rebholz CM. Identification of Protein Biomarkers of the Dietary Approaches to Stop Hypertension Diet in Randomized Feeding Studies and Validation in an Observational Study. J Am Heart Assoc 2023; 12:e028821. [PMID: 36974735 PMCID: PMC10122905 DOI: 10.1161/jaha.122.028821] [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: 12/20/2022] [Accepted: 02/15/2023] [Indexed: 03/29/2023]
Abstract
Background The Dietary Approaches to Stop Hypertension (DASH) diet is recommended for cardiovascular disease prevention. We aimed to identify protein biomarkers of the DASH diet using data from 2 randomized feeding studies and validate them in an observational study, the ARIC (Atherosclerosis Risk in Communities) study. Methods and Results Large-scale proteomic profiling was conducted in serum specimens (SomaLogic) collected at the end of 8-week and 4-week DASH diet interventions in multicenter, randomized controlled feeding studies of the DASH trial (N=215) and the DASH-Sodium trial (N=396), respectively. Multivariable linear regression models were used to compare the relative abundance of 7241 proteins between the DASH and control diet interventions. Estimates from the 2 trials were meta-analyzed using fixed-effects models. We validated significant proteins in the ARIC study (N=10 490) using the DASH diet score. At a false discovery rate <0.05, there were 71 proteins that were different between the DASH diet and control diet in the DASH and DASH-Sodium trials. Nineteen proteins were validated in the ARIC study. The 19 proteins collectively improved the prediction of the DASH diet intervention in the feeding studies (range of difference in C statistics, 0.267-0.313; P<0.001 for both tests) and the DASH diet score in the ARIC study (difference in C statistics, 0.017; P<0.001) beyond participant characteristics. Conclusions We identified 19 proteins robustly associated with the DASH diet in 3 studies, which may serve as biomarkers of the DASH diet. These results suggest potential pathways that are impacted by consumption of the DASH diet. Registration URL: https://www.clinicaltrials.gov; Unique identifiers: NCT03403166, NCT00000608.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology, and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
| | | | - Peter Ganz
- Cardiovascular Division, Zuckerberg San Francisco General HospitalUniversity of California, San FranciscoSan FranciscoCA
| | - Shutong Du
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology, and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
| | - Olive Tang
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology, and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
| | - Bing Yu
- Department of Epidemiology, Human Genetics & Environmental SciencesUniversity of Texas Health Sciences Center at Houston School of Public HealthHoustonTX
| | - Nilanjan Chatterjee
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | - Lawrence J. Appel
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology, and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
- Division of Nephrology, Department of MedicineJohns Hopkins School of MedicineBaltimoreMD
| | - Josef Coresh
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology, and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
- Division of Nephrology, Department of MedicineJohns Hopkins School of MedicineBaltimoreMD
| | - Casey M. Rebholz
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
- Welch Center for Prevention, Epidemiology, and Clinical ResearchJohns Hopkins UniversityBaltimoreMD
- Division of Nephrology, Department of MedicineJohns Hopkins School of MedicineBaltimoreMD
| |
Collapse
|
10
|
Shen S. Prospective Advances of Extracellular Vesicles Investigation in Cardiovascular and Metabolic Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:231-240. [PMID: 37603283 DOI: 10.1007/978-981-99-1443-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Extracellular vesicles (EVs) play an important role in cardiovascular and metabolic diseases through intercellular communication. Although there has been extensive research on EVs, there are still some unsolved problems in the technologies of investigation of EVs. In this chapter, we reviewed the current knowledge of EVs functions in cardiovascular and metabolic pathophysiology and EVs as biomarkers and therapeutic agents in cardiovascular and metabolic diseases. We also addressed the challenges in isolation and identification of EVs as well as challenges in visualization and tracking of EVs. By addressing these challenges, we hope to have a more in-depth understanding of the biological functions of EVs.
Collapse
Affiliation(s)
- Shutong Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Xu YN, Wang Z, Zhang SK, Xu JR, Pan ZX, Wei X, Wen HH, Luo YS, Guo MJ, Zhu Q. Low-grade elevation of palmitate and lipopolysaccharide synergistically induced β-cell damage via inhibition of neutral ceramidase. Mol Cell Endocrinol 2022; 539:111473. [PMID: 34610358 DOI: 10.1016/j.mce.2021.111473] [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: 04/01/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 02/08/2023]
Abstract
High concentrations of free fatty acids (FFAs) or lipopolysaccharide (LPS) could lead to β-cell apoptosis and dysfunction, while low-grade elevation of FFAs or LPS, which are more common in people with type 2 diabetes mellitus (T2DM) or obesity, have no obvious toxic effect on β-cells. Palmitate is a component closely related to metabolic disorders in FFAs. Recent studies have found that low-grade elevation of palmitate and LPS synergistically affects the sphingolipid signaling pathway by activating Toll-like receptor 4 (TLR4) and further enhances the expression of inflammatory cytokines in immune cells. Previous studies demonstrated that sphingolipids also played an important role in the occurrence and development of T2DM. This study aimed to investigate the synergistic effects of low-grade elevation of palmitate and LPS on viability, apoptosis and insulin secretion in the rat pancreatic β-cell line INS-1 or islets and the role of sphingolipids in this process. We showed that low-grade elevation of palmitate or LPS alone did not affect the viability, apoptosis, glucose-stimulated insulin secretion (GSIS) or intracellular insulin content of INS-1 cells or islets, while the combination of the two synergistically inhibited cell viability, induced apoptosis and decreased basal insulin secretion in INS-1 cells or islets. Treatment with palmitate and LPS markedly upregulated TLR4 protein expression and downregulated neutral ceramidase (NCDase) activity and protein expression. Additionally, low-grade elevation of palmitate and LPS synergistically induced a significant increase in ceramide and a decrease in sphingosine-1-phosphate. Blocking TLR4 signaling or overexpressing NCDase remarkably attenuated INS-1 cell injury induced by the combination of palmitate and LPS. However, inhibition of ceramide synthase did not ameliorate injury induced by palmitate and LPS. Overall, we show for the first time that low-grade elevation of palmitate and LPS synergistically induced β-cell damage by activating TLR4 signaling, inhibiting NCDase activity, and further modulating sphingolipid metabolism, which was different from a high concentration of palmitate-induced β-cell injury by promoting ceramide synthesis.
Collapse
Affiliation(s)
- Ya-Nan Xu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Zheng Wang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China; Department of Nephrology, Jiangsu University Affiliated People's Hospital, Zhenjiang, 212002, China
| | - Shao-Kun Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Jia-Rong Xu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Zhi-Xiong Pan
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Xiao Wei
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Hong-Hua Wen
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Yan-Shi Luo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Mao-Jun Guo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Qun Zhu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
12
|
Wang X, Yang G. Bone marrow mesenchymal stem cells-derived exosomes reduce Aβ deposition and improve cognitive function recovery in mice with Alzheimer's disease by activating sphingosine kinase/sphingosine-1-phosphate signaling pathway. Cell Biol Int 2021; 45:775-784. [PMID: 33300254 DOI: 10.1002/cbin.11522] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 10/09/2020] [Accepted: 12/06/2020] [Indexed: 12/21/2022]
Abstract
Exosomes are associated with the development and progression of Alzheimer's disease (AD), although the impact of these extracellular vesicles in brain pathological condition remains incompletely understood. Therefore, this study aimed to investigate the role and mechanism of exosomes signaling in AD. Double transgenic APP/PS1 mice were injected with bone marrow mesenchymal stem cells (BM-MSCs)-derived exosomes or combined with SKI-Ⅱ (sphingosine kinase [SphK] inhibitor) or VPC23019 (sphingosine-1-phosphate [S1P] 1 receptor blocker). We observed the spatial learning and memory ability of mice, and assessed the levels of amyloid and proteins. We found that exosomes improved spatial learning and memory ability of APP/PS1 mice, and enhanced the expression of SphK1 and S1P1. Moreover, exosomes inhibited the levels of amyloid and enhanced the expression of NeuN in cortex and hippocampus of APP/PS1 mice. Exosomes repressed the levels of Aβ1-40, Aβ1-42, BACE1, and PS1, and promoted the expression of neprilysin in APP/PS1 mice. The influence conferred by exosomes was abolished by SKI-Ⅱ or VPC23019. In conclusion, our article confirms that BM-MSCs-derived exosomes reduce Aβ deposition and promote cognitive function recovery in AD mice by activating SphK/S1P signaling pathway. Thus, our data suggest that S1P/SphK-containing exosomes should be explored as potential AD cure.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Geriatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guojie Yang
- Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Pang H, Luo S, Xiao Y, Xia Y, Li X, Huang G, Xie Z, Zhou Z. Emerging Roles of Exosomes in T1DM. Front Immunol 2020; 11:593348. [PMID: 33324409 PMCID: PMC7725901 DOI: 10.3389/fimmu.2020.593348] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a complex autoimmune disorder that mainly affects children and adolescents. The elevated blood glucose level of patients with T1DM results from absolute insulin deficiency and leads to hyperglycemia and the development of life-threatening diabetic complications. Although great efforts have been made to elucidate the pathogenesis of this disease, the precise underlying mechanisms are still obscure. Emerging evidence indicates that small extracellular vesicles, namely, exosomes, take part in intercellular communication and regulate interorgan crosstalk. More importantly, many findings suggest that exosomes and their cargo are associated with the development of T1DM. Therefore, a deeper understanding of exosomes is beneficial for further elucidating the pathogenic process of T1DM. Exosomes are promising biomarkers for evaluating the risk of developingty T1DM, monitoring the disease state and predicting related complications because their number and composition can reflect the status of their parent cells. Additionally, since exosomes are natural carriers of functional proteins, RNA and DNA, they can be used as therapeutic tools to deliver these molecules and drugs. In this review, we briefly introduce the current understanding of exosomes. Next, we focus on the relationship between exosomes and T1DM from three perspectives, i.e., the pathogenic role of exosomes in T1DM, exosomes as novel biomarkers of T1DM and exosomes as therapeutic tools for T1DM.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Xia
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
14
|
Wu WC, Song SJ, Zhang Y, Li X. Role of Extracellular Vesicles in Autoimmune Pathogenesis. Front Immunol 2020; 11:579043. [PMID: 33072123 PMCID: PMC7538611 DOI: 10.3389/fimmu.2020.579043] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases are conditions that emerge from abnormal immune responses to natural parts of the body. Extracellular vesicles (EVs) are membranous structures found in almost all types of cells. Because EVs often transport “cargo” between cells, their ability to crosstalk may be an important communication pathway within the body. The pathophysiological role of EVs is increasingly recognized in autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Sjogren's syndrome, Type 1 diabetes, and autoimmune thyroid disease. EVs are considered as biomarkers of these diseases. This article outlines existing knowledge on the biogenesis of EVs, their role as messegers in cellular communication and the function in T/B cell differentiation and maturation, and focusing on their potential application in autoimmune diseases.
Collapse
Affiliation(s)
- Wen-Cheng Wu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Sheng-Jiao Song
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
15
|
Sphingolipids in Type 1 Diabetes: Focus on Beta-Cells. Cells 2020; 9:cells9081835. [PMID: 32759843 PMCID: PMC7465050 DOI: 10.3390/cells9081835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/28/2022] Open
Abstract
Type 1 diabetes (T1DM) is a chronic autoimmune disease, with a strong genetic background, leading to a gradual loss of pancreatic beta-cells, which secrete insulin and control glucose homeostasis. Patients with T1DM require life-long substitution with insulin and are at high risk for development of severe secondary complications. The incidence of T1DM has been continuously growing in the last decades, indicating an important contribution of environmental factors. Accumulating data indicates that sphingolipids may be crucially involved in T1DM development. The serum lipidome of T1DM patients is characterized by significantly altered sphingolipid composition compared to nondiabetic, healthy probands. Recently, several polymorphisms in the genes encoding the enzymatic machinery for sphingolipid production have been identified in T1DM individuals. Evidence gained from studies in rodent islets and beta-cells exposed to cytokines indicates dysregulation of the sphingolipid biosynthetic pathway and impaired function of several sphingolipids. Moreover, a number of glycosphingolipids have been suggested to act as beta-cell autoantigens. Studies in animal models of autoimmune diabetes, such as the Non Obese Diabetic (NOD) mouse and the LEW.1AR1-iddm (IDDM) rat, indicate a crucial role of sphingolipids in immune cell trafficking, islet infiltration and diabetes development. In this review, the up-to-date status on the findings about sphingolipids in T1DM will be provided, the under-investigated research areas will be identified and perspectives for future studies will be given.
Collapse
|
16
|
Chidester S, Livinski AA, Fish AF, Joseph PV. The Role of Extracellular Vesicles in β-Cell Function and Viability: A Scoping Review. Front Endocrinol (Lausanne) 2020; 11:375. [PMID: 32595604 PMCID: PMC7300279 DOI: 10.3389/fendo.2020.00375] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) released by cells throughout the body have been implicated in diabetes pathogenesis. Understanding the role of EVs in regulation of β-cell function and viability may provide insights into diabetes etiology and may lead to the development of more effective screening and diagnostic tools to detect diabetes earlier and prevent disease progression. This review was conducted to determine what is known from the literature about the effect of EV crosstalk on pancreatic β-cell function and viability in the pathogenesis of diabetes mellitus, to perform a gap analysis for future research directions, and to discuss implications of available evidence for diabetes care. The literature search yielded 380 studies from which 31 studies were determined to meet eligibility criteria. The majority of studies had the disease context of autoimmunity in T1DM. The most commonly studied EV crosstalk dynamics involved localized EV-mediated communication between β-cells and other islet cells, or between β-cells and immune cells. Other organs and tissues secreting EVs that affect β-cells include skeletal muscle, hepatocytes, adipocytes, immune cells, bone marrow, vascular endothelium, and mesenchymal stem cells. Characterization of EV cargo molecules with regulatory effects in β-cells was conducted in 24 studies, with primary focus on microRNA cargo. Gaps identified included scarcity of evidence for the effect on β-cell function and viability of EVs from major metabolic organs/tissues such as muscle, liver, and adipose depots. Future research should address these gaps as well as characterize a broader range of EV cargo molecules and their activity in β-cells.
Collapse
Affiliation(s)
- Stephanie Chidester
- Sensory Science & Metabolism Unit, Biobehavioral Branch, National Institute of Nursing Research, Division of Intramural Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, United States
- College of Nursing, University of Missouri-St. Louis, St. Louis, MO, United States
| | - Alicia A. Livinski
- National Institutes of Health Library, Office of Research Services, OD, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, United States
| | - Anne F. Fish
- College of Nursing, University of Missouri-St. Louis, St. Louis, MO, United States
| | - Paule V. Joseph
- Sensory Science & Metabolism Unit, Biobehavioral Branch, National Institute of Nursing Research, Division of Intramural Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, United States
| |
Collapse
|
17
|
Li G, Liu H, Ma C, Chen Y, Wang J, Yang Y. Exosomes are the novel players involved in the beneficial effects of exercise on type 2 diabetes. J Cell Physiol 2019; 234:14896-14905. [PMID: 30756380 DOI: 10.1002/jcp.28319] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
Exosomes contain regulatory signals such as lipids, proteins, and nucleic acids which can be transferred to adjacent or remote cells to mediate cell-to-cell communication. Exercise is a positive lifestyle for metabolic health and a nonpharmacological treatment of insulin resistance and metabolic diseases. Moreover, exercise is a stressor that induces cellular responses including gene expression and exosome release in various types of cells. Exosomes can carry the characters of parent cells by their modified cargoes, representing novel mechanisms for the effects of exercise. Here, we present a review of exosomes as the perspective players in mediating exercise's beneficial impacts on type 2 diabetes (T2D).
Collapse
Affiliation(s)
- Gaohua Li
- School of Physical Education, Henan Agricultural University, Zhengzhou, China.,Hubei Key Laboratory of Exercise Training and Monitoring,College of Health Science, Wuhan Sports University, Wuhan, China
| | - Hua Liu
- Hubei Key Laboratory of Exercise Training and Monitoring,College of Health Science, Wuhan Sports University, Wuhan, China
| | - Chunlian Ma
- Hubei Key Laboratory of Exercise Training and Monitoring,College of Health Science, Wuhan Sports University, Wuhan, China
| | - Yanfang Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Jinju Wang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Yi Yang
- Hubei Key Laboratory of Exercise Training and Monitoring,College of Health Science, Wuhan Sports University, Wuhan, China
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Theories about the pathogenesis of type 1 diabetes (T1D) refer to the potential of primary islet inflammatory signaling as a trigger for the loss of self-tolerance leading to disease onset. Emerging evidence suggests that extracellular vesicles (EV) may represent the missing link between inflammation and autoimmunity. Here, we review the evidence for a role of EV in the pathogenesis of T1D, as well as discuss their potential value in the clinical sphere, as biomarkers and therapeutic agents. RECENT FINDINGS EV derived from β cells are enriched in diabetogenic autoantigens and miRNAs that are selectively sorted and packaged. These EV play a pivotal role in antigen presentation and cell to cell communication leading to activation of autoimmune responses. Furthermore, recent evidence suggests the potential of EV as novel tools in clinical diagnostics and therapeutic interventions. In-depth analysis of EV cargo using modern multi-parametric technologies may be useful in enhancing our understanding of EV-mediated immune mechanisms and in identifying robust biomarkers and therapeutic strategies for T1D.
Collapse
Affiliation(s)
- Sarita Negi
- Human Islet Transplant Laboratory, Department of Surgery, D5.5736, Royal Victoria Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, T6G 2E1, Canada
| | - Alissa K Rutman
- Human Islet Transplant Laboratory, Department of Surgery, D5.5736, Royal Victoria Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, T6G 2E1, Canada
| | - Steven Paraskevas
- Human Islet Transplant Laboratory, Department of Surgery, D5.5736, Royal Victoria Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada.
- Canadian Donation and Transplantation Research Program, Edmonton, Alberta, T6G 2E1, Canada.
| |
Collapse
|
19
|
Castaño C, Novials A, Párrizas M. Exosomes and diabetes. Diabetes Metab Res Rev 2019; 35:e3107. [PMID: 30513130 DOI: 10.1002/dmrr.3107] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Diabetes is a group of metabolic diseases characterized by elevated blood glucose levels that drive the development of life-threatening complications. Diabetes results from a situation of insufficient insulin action, either by deficient production of the hormone by the pancreas, or by the development of insulin resistance in peripheral tissues such as liver, muscle, or the adipose depots. Communication between organs is thus central to the maintenance of glucose homoeostasis. Recently, several studies are evidencing that small vesicles called exosomes released by, amongst other, the adipose tissue can regulate gene expression in other tissues, hence modulating interorgan crosstalk. Therefore, exosomes participate in the development of diabetes and its associated complications. Their study holds the potential of providing us with novel biomarkers for the early diagnosis and stratification of patients at risk of developing diabetes, hence allowing the timely implementation of more personalized therapies. On the other hand, the molecular dissection of the pathways initiated by exosomes under situations of metabolic stress could help to gain a deeper knowledge of the pathophysiology of diabetes and its associated metabolic diseases.
Collapse
Affiliation(s)
- Carlos Castaño
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Marcelina Párrizas
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| |
Collapse
|
20
|
Javeed N. Shedding Perspective on Extracellular Vesicle Biology in Diabetes and Associated Metabolic Syndromes. Endocrinology 2019; 160:399-408. [PMID: 30624638 PMCID: PMC6349001 DOI: 10.1210/en.2018-01010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Abstract
The etiology of diabetes and associated metabolic derailments is a complex process that relies on crosstalk between metabolically active tissues. Dysregulation of secreted factors and metabolites from islets, adipose tissue, liver, and skeletal muscle contributes to the overall progression of diabetes and metabolic syndrome. Extracellular vesicles (EVs) are circulating nanovesicles secreted by most cell types and are comprised of bioactive cargoes that are horizontally transferred to targeted cells/tissues. Accumulating evidence from the past decade implicates the role of EVs as mediators of islet cell dysfunction, inflammation, insulin resistance, and other metabolic consequences associated with diabetes. This review covers a broad spectrum of basic EV biology (i.e., biogenesis, secretion, and uptake), including a comprehensive investigation of the emerging role of EVs in β-cell autocrine/paracrine interactions and the multidirectional crosstalk in metabolically active tissues. Understanding the utility of this novel means of intercellular communication could impart insight into the development of new treatment regimens and biomarker detection to treat diabetes.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Correspondence: Naureen Javeed, PhD, Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905. E-mail:
| |
Collapse
|
21
|
Coant N, Hannun YA. Neutral ceramidase: Advances in mechanisms, cell regulation, and roles in cancer. Adv Biol Regul 2018; 71:141-146. [PMID: 30389354 DOI: 10.1016/j.jbior.2018.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 01/11/2023]
Abstract
Extensive research conducted in the last three decades has identified the roles for the main bioactive sphingolipids, namely ceramide, sphingosine, and sphingosine 1-phosphate (S1P) as key regulators of cellular homeostasis, growth and death. One of the major groups of enzymes in the ceramide pathway, ceramidases, converts ceramide into sphingosine and fatty acids, with sphingosine being further metabolized to S1P. Thus, these enzymes play important roles in the network controlling the functions associated with these bioactive sphingolipids. Among the family of ceramidases, neutral ceramidase (nCDase), which is named according to its optimal pH for catalytic activity, has received increased attention in the last decade. The goal of this review is to provide a brief background on bioactive sphingolipids and the ceramidases. We then describe more recent advances on nCDase, specifically the resolution of its crystal structure and understanding its roles in cell biology and physiology.
Collapse
Affiliation(s)
- Nicolas Coant
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 182, 11794, Stony Brook, NY, USA.
| |
Collapse
|
22
|
Lin M, Liao W, Dong M, Zhu R, Xiao J, Sun T, Chen Z, Wu B, Jin J. Exosomal neutral sphingomyelinase 1 suppresses hepatocellular carcinoma via decreasing the ratio of sphingomyelin/ceramide. FEBS J 2018; 285:3835-3848. [PMID: 30106227 DOI: 10.1111/febs.14635] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/19/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022]
Abstract
Neutral sphingomyelinase 1 (NSMase1) mediates caspase-3 activation and apoptosis. However, the role of NSMase1, especially exosome-borne NSMase1 in hepatocellular carcinoma (HCC), remains unclear. We report that NSMase1, which converts sphingomyelin (SM) to ceramide, was significantly downregulated in HCC tissues. Low NSMase1 expression predicted poor long-term survival of HCC patients. NSMase1 downregulation in HCC resulted in increased SM and reduced ceramide (Cer) that led to an increased SM/Cer ratio. Interestingly, NSMase1 and NSMase activity were also decreased in exosomes isolated from HCC tissues and cell lines. Furthermore, NSMase activity increased in exosomes isolated from the culture medium of L02 cells transfected with pEGFP-C3-NSMase1 (NSMase1-Exo). NSMase1-Exo suppressed HCC cell growth and induced apoptosis via reduction of the SM/Cer ratio. Thus, NSMase1 in exosomes inhibits HCC growth by decreasing the SM/Cer ratio.
Collapse
Affiliation(s)
- Minglin Lin
- Department of Surgery, Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Mingjun Dong
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Rongping Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Juan Xiao
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Tian Sun
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Zhiyong Chen
- Light of Life Biotechnology, Ltd., Changsha, China
| | - Bin Wu
- Light of Life Biotechnology, Ltd., Changsha, China
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China.,China-USA Lipids in Health and Disease Research Center, Guilin Medical University, China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, China
| |
Collapse
|
23
|
Verderio C, Gabrielli M, Giussani P. Role of sphingolipids in the biogenesis and biological activity of extracellular vesicles. J Lipid Res 2018; 59:1325-1340. [PMID: 29853528 DOI: 10.1194/jlr.r083915] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/06/2018] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles released by both eukaryotic and prokaryotic cells; they not only serve physiological functions, such as disposal of cellular components, but also play pathophysiologic roles in inflammatory and degenerative diseases. Common molecular mechanisms for EV biogenesis are evident in different cell biological contexts across eukaryotic phyla, and inhibition of this biogenesis may provide an avenue for therapeutic research. The involvement of sphingolipids (SLs) and their enzymes on EV biogenesis and release has not received much attention in current research. Here, we review how SLs participate in EV biogenesis by shaping membrane curvature and how they contribute to EV action in target cells. First, we describe how acid and neutral SMases, by generating the constitutive SL, ceramide, facilitate biogenesis of EVs at the plasma membrane and inside the endocytic compartment. We then discuss the involvement of other SLs, such as sphingosine-1-phosphate and galactosyl-sphingosine, in EV formation and cargo sorting. Last, we look ahead at some biological effects of EVs mediated by changes in SL levels in recipient cells.
Collapse
Affiliation(s)
- Claudia Verderio
- Consiglio Nazionale delle Ricerche (CNR) Institute of Neuroscience, 20129 Milano, Italy .,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas, 20089 Rozzano, Italy
| | - Martina Gabrielli
- Consiglio Nazionale delle Ricerche (CNR) Institute of Neuroscience, 20129 Milano, Italy
| | - Paola Giussani
- Department of Biotechnology and Translational Medicine, University of Milano, 20090 Segrate, Italy
| |
Collapse
|
24
|
Lakhter AJ, Pratt RE, Moore RE, Doucette KK, Maier BF, DiMeglio LA, Sims EK. Beta cell extracellular vesicle miR-21-5p cargo is increased in response to inflammatory cytokines and serves as a biomarker of type 1 diabetes. Diabetologia 2018; 61:1124-1134. [PMID: 29445851 PMCID: PMC5878132 DOI: 10.1007/s00125-018-4559-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/09/2018] [Indexed: 02/08/2023]
Abstract
AIMS/HYPOTHESIS Improved biomarkers are acutely needed for the detection of developing type 1 diabetes, prior to critical loss of beta cell mass. We previously demonstrated that elevated beta cell microRNA 21-5p (miR-21-5p) in rodent and human models of type 1 diabetes increased beta cell apoptosis. We hypothesised that the inflammatory milieu of developing diabetes may also increase miR-21-5p in beta cell extracellular vesicle (EV) cargo and that circulating EV miR-21-5p would be increased during type 1 diabetes development. METHODS MIN6 and EndoC-βH1 beta cell lines and human islets were treated with IL-1β, IFN-γ and TNF-α to mimic the inflammatory milieu of early type 1 diabetes. Serum was collected weekly from 8-week-old female NOD mice until diabetes onset. Sera from a cross-section of 19 children at the time of type 1 diabetes diagnosis and 16 healthy children were also analysed. EVs were isolated from cell culture media or serum using sequential ultracentrifugation or ExoQuick precipitation and EV miRNAs were assayed. RESULTS Cytokine treatment in beta cell lines and human islets resulted in a 1.5- to threefold increase in miR-21-5p. However, corresponding EVs were further enriched for this miRNA, with a three- to sixfold EV miR-21-5p increase in response to cytokine treatment. This difference was only partially reduced by pre-treatment of beta cells with Z-VAD-FMK to inhibit cytokine-induced caspase activity. Nanoparticle tracking analysis showed cytokines to have no effect on the number of EVs, implicating specific changes within EV cargo as being responsible for the increase in beta cell EV miR-21-5p. Sequential ultracentrifugation to separate EVs by size suggested that this effect was mostly due to cytokine-induced increases in exosome miR-21-5p. Longitudinal serum collections from NOD mice showed that EVs displayed progressive increases in miR-21-5p beginning 3 weeks prior to diabetes onset. To validate the relevance to human diabetes, we assayed serum from children with new-onset type 1 diabetes compared with healthy children. While total serum miR-21-5p and total serum EVs were reduced in diabetic participants, serum EV miR-21-5p was increased threefold compared with non-diabetic individuals. By contrast, both serum and EV miR-375-5p were increased in parallel among diabetic participants. CONCLUSIONS/INTERPRETATION We propose that circulating EV miR-21-5p may be a promising marker of developing type 1 diabetes. Additionally, our findings highlight that, for certain miRNAs, total circulating miRNA levels are distinct from circulating EV miRNA content.
Collapse
Affiliation(s)
- Alexander J Lakhter
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachel E Pratt
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachel E Moore
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kaitlin K Doucette
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bernhard F Maier
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA
| | - Linda A DiMeglio
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Emily K Sims
- Department of Pediatrics, Section of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, 635 Barnhill Dr., Rm 2031, Indianapolis, IN, 46202, USA.
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
25
|
Console L, Scalise M, Tonazzi A, Giangregorio N, Indiveri C. Characterization of Exosomal SLC22A5 (OCTN2) carnitine transporter. Sci Rep 2018; 8:3758. [PMID: 29491466 PMCID: PMC5830701 DOI: 10.1038/s41598-018-22170-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022] Open
Abstract
Exosomes are extracellular vesicles involved in cell-to-cell communication. Previous large scale proteomics revealed that they contain SLC proteins. However, no data on the function of exosomal SLCs is available, so far. An SLC localized in exosomes was here characterized for the first time: the carnitine transporter OCTN2 (SLC22A5). The protein was detected by Western Blot analysis in HEK293 exosomes. To investigate the functional properties of the exosomal OCTN2, the proteins extracted from vesicles were reconstituted into proteolipsomes and the transport function was measured as uptake of 3H-carnitine. Transport was stimulated by sodium and was dependent on pH. 3H-carnitine uptake was inhibited by Acetyl-carnitine, but not by Asn, Gln and Arg thus excluding interference by ATB0,+, an amino acid transporter which also recognizes carnitine. Cardiolipin failed to stimulate transport, excluding the activity of the mitochondrial Carnitine/acylcarnitine transporter. Increased level of exosomal OCTN2 was induced by treatment of HEK293 with the pro-inflammatory cytokine INFγ. All data concurred to demonstrate that OCTN2 present in exosomes is fully functional and is in its native conformation. Functional OCTN2 was detected also in human urinary exosomes, thus suggesting the OCTN2 exosomal protein as a candidate biomarker for inflammation related pathologies.
Collapse
Affiliation(s)
- Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Annamaria Tonazzi
- CNR Institute of Biomembranes and Bioenergetics, via Amendola 165/A, 70126, Bari, Italy
| | - Nicola Giangregorio
- CNR Institute of Biomembranes and Bioenergetics, via Amendola 165/A, 70126, Bari, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy. .,CNR Institute of Biomembranes and Bioenergetics, via Amendola 165/A, 70126, Bari, Italy.
| |
Collapse
|
26
|
Zhu Q, Yang J, Zhu R, Jiang X, Li W, He S, Jin J. Dihydroceramide-desaturase-1-mediated caspase 9 activation through ceramide plays a pivotal role in palmitic acid-induced HepG2 cell apoptosis. Apoptosis 2018; 21:1033-44. [PMID: 27364952 DOI: 10.1007/s10495-016-1267-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, results showed that the inhibition of PA-induced HepG2 cell growth takes place in a time- and concentration-dependent manner, that activation of caspase 9 is necessary for PA-induced HepG2 cell apoptosis, that dihydroceramide desaturase 1 (DES1) plays a key role in PA-mediated caspase 9 and caspase 3 activation, and that palmitoleic acid (POA), an omega-7 monounsaturated fatty acid, reverses PA-induced apoptosis through DES1 → Ceramide → Caspase 9 → Caspase 3 signaling.
Collapse
Affiliation(s)
- Qun Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 210011, Nanjing, Jiangsu, People's Republic of China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Jianjun Yang
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Rongping Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Xin Jiang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Wanlian Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Songqing He
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
| |
Collapse
|
27
|
Guay C, Regazzi R. Exosomes as new players in metabolic organ cross-talk. Diabetes Obes Metab 2017; 19 Suppl 1:137-146. [PMID: 28880477 DOI: 10.1111/dom.13027] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/22/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022]
Abstract
Blood glucose homeostasis requires a constant communication between insulin-secreting and insulin-sensitive cells. A wide variety of circulating factors, including hormones, cytokines and chemokines work together to orchestrate the systemic response of metabolic organs to changes in the nutritional state. Failure in the coordination between these organs can lead to a rise in blood glucose levels and to the appearance of metabolic disorders such as diabetes mellitus. Exosomes are small extracellular vesicles (EVs) that are produced via the endosomal pathway and are released from the cells upon fusion of multivesicular bodies with the plasma membrane. There is emerging evidence indicating that these EVs play a central role in cell-to-cell communication. The interest in exosomes exploded when they were found to transport bioactive proteins, messenger RNA (mRNAs) and microRNA (miRNAs) that can be transferred in active form to adjacent cells or to distant organs. In this review, we will first outline the mechanisms governing the biogenesis, the cargo upload and the release of exosomes by donor cells as well as the uptake by recipient cells. We will then summarize the studies that support the novel concept that miRNAs and other exosomal cargo components are new important vehicles for metabolic organ cross-talk.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
28
|
Luo F, Feng Y, Ma H, Liu C, Chen G, Wei X, Mao X, Li X, Xu Y, Tang S, Wen H, Jin J, Zhu Q. Neutral ceramidase activity inhibition is involved in palmitate-induced apoptosis in INS-1 cells. Endocr J 2017; 64:767-776. [PMID: 28674283 DOI: 10.1507/endocrj.ej16-0512] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Neutral ceramidase (NCDase) is a class of ceramidases, a key enzyme in ceramide degradation. Recently, it was observed that NCDase activity was suppressed by saturated fatty acids to increase ceramide content in rat muscle. However, little is known about its changes in activity and roles in palmitate (Palm)-induced lipotoxicity in pancreatic β cells. Here, we demonstrated that Palm treatment significantly down-regulated NCDase activity, mRNA and protein levels in rat INS-1 cells. In addition, Palm caused a significant accumulation of ceramide, while SPH level remained unchanged, suggesting that inhibition of NCDase activity led to no change of SPH level after treatment with Palm for 24 h. Furthermore, NCDase overexpression significantly reduced Palm-induced apoptosis in INS-1 cells. Conversely, NCDase siRNA knockdown markedly exacerbated Palm-induced apoptosis. In conclusion, Palm treatment suppressed the activity of NCDase and down-regulated its mRNA and protein expression. Furthermore, NCDase inhibition was involved in Palm-induced apoptosis by blocking ceramide degradation in INS-1 cells.
Collapse
Affiliation(s)
- Fen Luo
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Yamin Feng
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Huimin Ma
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Chao Liu
- Endocrine and Diabetes Center, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Branch of China Academy of Chinese Medical Science, Nanjing 210028, China
| | - Guofang Chen
- Endocrine and Diabetes Center, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Branch of China Academy of Chinese Medical Science, Nanjing 210028, China
| | - Xiao Wei
- Endocrine and Diabetes Center, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Branch of China Academy of Chinese Medical Science, Nanjing 210028, China
| | - Xiaodong Mao
- Endocrine and Diabetes Center, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Branch of China Academy of Chinese Medical Science, Nanjing 210028, China
| | - Xingjia Li
- Endocrine and Diabetes Center, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Branch of China Academy of Chinese Medical Science, Nanjing 210028, China
| | - Yijiao Xu
- Endocrine and Diabetes Center, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Branch of China Academy of Chinese Medical Science, Nanjing 210028, China
| | - Shan Tang
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Honghua Wen
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Junfei Jin
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Gulin 541001, China
| | - Qun Zhu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| |
Collapse
|
29
|
Guay C, Regazzi R. New emerging tasks for microRNAs in the control of β-cell activities. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2121-2129. [DOI: 10.1016/j.bbalip.2016.05.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/22/2016] [Accepted: 05/02/2016] [Indexed: 12/26/2022]
|
30
|
Zhu Q, Zhu R, Jin J. Neutral ceramidase-enriched exosomes prevent palmitic acid-induced insulin resistance in H4IIEC3 hepatocytes. FEBS Open Bio 2016; 6:1078-1084. [PMID: 27833848 PMCID: PMC5095145 DOI: 10.1002/2211-5463.12125] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 01/11/2023] Open
Abstract
Oversupply of free fatty acids such as palmitic acid (PA) from the portal vein may cause liver insulin resistance. Production of reactive oxygen species plays a pivotal role in PA‐induced insulin resistance in H4IIEC3 hepatocytes. Recently, we found that exosomes secreted from INS‐1 cells that were transfected with neutral ceramidase (NCDase) plasmids had raised NCDase activity; these NCDase‐enriched exosomes could inhibit PA‐induced INS‐1 cell apoptosis. Here, we showed that PA reduced insulin‐stimulated tyrosine phosphorylation of insulin receptor substrate 2 and decreased insulin‐stimulated uptake of the fluorescent glucose analog 2‐NBDG, confirming that insulin resistance occurred in PA‐treated H4IIEC3 cells. Moreover, NCDase‐enriched exosomes from INS‐1 cells rescued PA‐induced H4IIEC3 insulin resistance and blocked PA‐induced reactive oxygen species production in which ceramide was involved.
Collapse
Affiliation(s)
- Qun Zhu
- Department of Endocrinology the Second Affiliated Hospital of Nanjing Medical University Jiangsu China; China-USA Lipids in Health and Disease Research Center Guilin Medical University Guangxi China
| | - Rongping Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery Affiliated Hospital of Guilin Medical University Guangxi China
| | - Junfei Jin
- China-USA Lipids in Health and Disease Research Center Guilin Medical University Guangxi China; Laboratory of Hepatobiliary and Pancreatic Surgery Affiliated Hospital of Guilin Medical University Guangxi China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair Guilin Medical University Guangxi China
| |
Collapse
|
31
|
Jin J, Liao W, Yao W, Zhu R, Li Y, He S. Aldo-keto Reductase Family 1 Member B 10 Mediates Liver Cancer Cell Proliferation through Sphingosine-1-Phosphate. Sci Rep 2016; 6:22746. [PMID: 26948042 PMCID: PMC4780005 DOI: 10.1038/srep22746] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/19/2016] [Indexed: 02/07/2023] Open
Abstract
AKR1B10 is involved in hepatocarcinogenesis via modulation of fatty acid and lipid synthesis. AKR1B10 inhibition results in apoptosis of tumor cells whose lipids, especially phospholipids, were decreased by over 50%, suggesting involvement of phospholipids like sphingosine-1-phosphate (S1P) in AKR1B10's oncogenic function. Using a co-culture system, we found that co-culture of QSG-7701 (human hepatocyte) with HepG2 (hepatoma cell line) increases QSG-7701's proliferation, in which AKR1B10-S1P signaling plays a pivotal role. Consistent with previous findings, AKR1B10 mRNA and protein levels were higher in primary hepatocellular carcinoma (PHC) tissues than in peri-tumor tissues. Interestingly, the level of S1P was also higher in PHC tissues than in peri-tumor tissues. After analyzing the correlation between AKR1B10 mRNA expression in PHC tissues and the clinical data, we found that AKR1B10 mRNA expression was associated with serum alpha-fetoprotein (AFP), tumor-node-metastasis (TNM) stage, and lymph node metastasis, but not with other clinicopathologic variables. A higher AKR1B10 mRNA expression level is related to a shorter DFS (disease free survival) and OS (overall survival), serving as an independent predictor of DFS and OS in PHC patients with surgical resection.
Collapse
Affiliation(s)
- Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.,China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Wenmin Yao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Rongping Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Yulan Li
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.,China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Songqing He
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| |
Collapse
|
32
|
Lakhter AJ, Sims EK. Minireview: Emerging Roles for Extracellular Vesicles in Diabetes and Related Metabolic Disorders. Mol Endocrinol 2015; 29:1535-48. [PMID: 26393296 PMCID: PMC4627606 DOI: 10.1210/me.2015-1206] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs), membrane-contained vesicles released by most cell types, have attracted a large amount of research interest over the past decade. Because of their ability to transfer cargo via regulated processes, causing functional impacts on recipient cells, these structures may play important roles in cell-cell communication and have implications in the physiology of numerous organ systems. In addition, EVs have been described in most human biofluids and have wide potential as relatively noninvasive biomarkers of various pathologic conditions. Specifically, EVs produced by the pancreatic β-cell have been demonstrated to regulate physiologic and pathologic responses to β-cell stress, including β-cell proliferation and apoptosis. β-Cell EVs are also capable of interacting with immune cells and may contribute to the activation of autoimmune processes that trigger or propagate β-cell inflammation and destruction during the development of diabetes. EVs from adipose tissue have been shown to contribute to the development of the chronic inflammation and insulin resistance associated with obesity and metabolic syndrome via interactions with other adipose, liver, and muscle cells. Circulating EVs may also serve as biomarkers for metabolic derangements and complications associated with diabetes. This minireview describes the properties of EVs in general, followed by a more focused review of the literature describing EVs affecting the β-cell, β-cell autoimmunity, and the development of insulin resistance, which all have the potential to affect development of type 1 or type 2 diabetes.
Collapse
Affiliation(s)
- Alexander J Lakhter
- Department of Pediatrics (A.J.L., E.K.S.), Center for Diabetes and Metabolic Diseases, and Section of Pediatric Endocrinology and Diabetology (E.K.S.), Indiana University, Indianapolis, Indiana 46202
| | - Emily K Sims
- Department of Pediatrics (A.J.L., E.K.S.), Center for Diabetes and Metabolic Diseases, and Section of Pediatric Endocrinology and Diabetology (E.K.S.), Indiana University, Indianapolis, Indiana 46202
| |
Collapse
|
33
|
Guay C, Menoud V, Rome S, Regazzi R. Horizontal transfer of exosomal microRNAs transduce apoptotic signals between pancreatic beta-cells. Cell Commun Signal 2015; 13:17. [PMID: 25880779 PMCID: PMC4371845 DOI: 10.1186/s12964-015-0097-7] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
Background Diabetes mellitus is a common metabolic disorder characterized by dysfunction of insulin-secreting pancreatic beta-cells. MicroRNAs are important regulators of beta-cell activities. These non-coding RNAs have recently been discovered to exert their effects not only inside the cell producing them but, upon exosome-mediated transfer, also in other recipient cells. This novel communication mode remains unexplored in pancreatic beta-cells. In the present study, the microRNA content of exosomes released by beta-cells in physiological and physiopathological conditions was analyzed and the biological impact of their transfer to recipient cells investigated. Results Exosomes were isolated from the culture media of MIN6B1 and INS-1 derived 832/13 beta-cell lines and from mice, rat or human islets. Global profiling revealed that the microRNAs released in MIN6B1 exosomes do not simply reflect the content of the cells of origin. Indeed, while a subset of microRNAs was preferentially released in exosomes others were selectively retained in the cells. Moreover, exposure of MIN6B1 cells to inflammatory cytokines changed the release of several microRNAs. The dynamics of microRNA secretion and their potential transfer to recipient cells were next investigated. As a proof-of-concept, we demonstrate that if cel-miR-238, a C. Elegans microRNA not present in mammalian cells, is expressed in MIN6B1 cells a fraction of it is released in exosomes and is transferred to recipient beta-cells. Furthermore, incubation of untreated MIN6B1 or mice islet cells in the presence of microRNA-containing exosomes isolated from the culture media of cytokine-treated MIN6B1 cells triggers apoptosis of recipient cells. In contrast, exosomes originating from cells not exposed to cytokines have no impact on cell survival. Apoptosis induced by exosomes produced by cytokine-treated cells was prevented by down-regulation of the microRNA-mediating silencing protein Ago2 in recipient cells, suggesting that the effect is mediated by the non-coding RNAs. Conclusions Taken together, our results suggest that beta-cells secrete microRNAs that can be transferred to neighboring beta-cells. Exposure of donor cells to pathophysiological conditions commonly associated with diabetes modifies the release of microRNAs and affects survival of recipient beta-cells. Our results support the concept that exosomal microRNAs transfer constitutes a novel cell-to-cell communication mechanism regulating the activity of pancreatic beta-cells. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0097-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| | - Véronique Menoud
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| | - Sophie Rome
- CarMeN Laboratory (INSERM U.1060/INRA 1397, INSA), University of Lyon, Faculty of Medicine Lyon-Sud, Ouillons, France.
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| |
Collapse
|