51
|
Lucotti S, Kenific CM, Zhang H, Lyden D. Extracellular vesicles and particles impact the systemic landscape of cancer. EMBO J 2022; 41:e109288. [PMID: 36052513 PMCID: PMC9475536 DOI: 10.15252/embj.2021109288] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022] Open
Abstract
Intercellular cross talk between cancer cells and stromal and immune cells is essential for tumor progression and metastasis. Extracellular vesicles and particles (EVPs) are a heterogeneous class of secreted messengers that carry bioactive molecules and that have been shown to be crucial for this cell-cell communication. Here, we highlight the multifaceted roles of EVPs in cancer. Functionally, transfer of EVP cargo between cells influences tumor cell growth and invasion, alters immune cell composition and function, and contributes to stromal cell activation. These EVP-mediated changes impact local tumor progression, foster cultivation of pre-metastatic niches at distant organ-specific sites, and mediate systemic effects of cancer. Furthermore, we discuss how exploiting the highly selective enrichment of molecules within EVPs has profound implications for advancing diagnostic and prognostic biomarker development and for improving therapy delivery in cancer patients. Altogether, these investigations into the role of EVPs in cancer have led to discoveries that hold great promise for improving cancer patient care and outcome.
Collapse
Affiliation(s)
- Serena Lucotti
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Candia M Kenific
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Haiying Zhang
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - David Lyden
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
52
|
Bai R, Zhang T, Gao Y, Shu T, Zhou Y, Wang F, Chang X, Tang W, Zhu Y, Han X. Rab31, a receptor of advanced glycation end products (RAGE) interacting protein, inhibits AGE induced pancreatic β-cell apoptosis through the pAKT/BCL2 pathway. Endocr J 2022; 69:1015-1026. [PMID: 35314532 DOI: 10.1507/endocrj.ej21-0594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [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
Receptor of advanced glycation end products (RAGE) mediates diverse signal transduction following ligand stimulation and plays an important role in diabetes complications and aging associated disease. We have previously verified that advanced glycation end products (AGE) bind to RAGE to cause pancreatic β-cell apoptosis through the mitochondrial pathway. However, the direct interacting protein(s) of RAGE in β cells has never been appreciated. In the present study, we utilized GST pull-down assay combined with mass spectrometry to identify the interacting proteins of the RAGE intracellular domain (C-terminal 43 amino acid of RAGE). Overall four RAGE interacting proteins, including Rab31, were identified with scores over 160. Rab31 was detected in three β-cell lines and confirmed to have interacted with RAGE via co-immunoprecipitation and immunostaining assays. This interaction was further enhanced by glycation-serum (GS) stimulation due to membrane distribution of Rab31 following treatment with GS. We further confirmed that Rab31 promoted RAGE endocytosis and inhibited GS-induced β-cell apoptosis through the pAKT/BCL2 pathway. These findings reveal a new RAGE interaction protein Rab31 that prevents AGE/RAGE-induced pancreatic β-cell apoptosis. Rab31 is therefore a promising therapeutic target for preserving functional β cells under diabetes conditions.
Collapse
Affiliation(s)
- Rongjie Bai
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Tao Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Yan Gao
- Institute of Suzhou Biobank, Suzhou Center for Disease Prevention and Control, Suzhou 215004, China
- Suzhou Institute of Advanced Study in Public Health, Gusu School, Nanjing Medical University, Suzhou 215004, China
| | - Tingting Shu
- Department of Endocrinology, Geriatric Hospital of Nanjing Medical University, Nanjing 210024, China
| | - Yuncai Zhou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Fuqiang Wang
- Analysis Center, Nanjing Medical University, Nanjing 210029, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Wei Tang
- Department of Endocrinology, Geriatric Hospital of Nanjing Medical University, Nanjing 210024, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
53
|
Sharari S, Kabeer B, Mohammed I, Haris B, Pavlovski I, Hawari I, Bhat AA, Toufiq M, Tomei S, Mathew R, Syed N, Nisar S, Maacha S, Grivel JC, Chaussabel D, Ericsson J, Hussain K. Understanding the Role of GLUT2 in Dysglycemia Associated with Fanconi-Bickel Syndrome. Biomedicines 2022; 10:biomedicines10092114. [PMID: 36140215 PMCID: PMC9495670 DOI: 10.3390/biomedicines10092114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 12/04/2022] Open
Abstract
Fanconi−Bickel Syndrome (FBS) is a rare disorder of carbohydrate metabolism that is characterized by the accumulation of glycogen mainly in the liver. It is inherited in an autosomal recessive manner due to mutations in the SLC2A2 gene. SLC2A2 encodes for the glucose transporter GLUT2 and is expressed in tissues that are involved in glucose homeostasis. The molecular mechanisms of dysglycemia in FBS are still not clearly understood. In this study, we report two cases of FBS with classical phenotypes of FBS associated with dysglycemia. Genomic DNA was extracted and analyzed by whole-genome and Sanger sequencing, and patient PBMCs were used for molecular analysis. One patient had an exonic SLC2A2 mutation (c.1093C>T in exon 9, R365X), while the other patient had a novel intronic SLC2A2 mutation (c.613-7T>G). Surprisingly, the exonic mutation resulted in the overexpression of dysfunctional GLUT2, resulting in the dysregulated expression of other glucose transporters. The intronic mutation did not affect the coding sequence of GLUT2, its expression, or glucose transport activity. However, it was associated with the expression of miRNAs correlated with type 1 diabetes mellitus, with a particular significant overexpression of hsa-miR-29a-3p implicated in insulin production and secretion. Our findings suggest that SLC2A2 mutations cause dysglycemia in FBS either by a direct effect on GLUT2 expression and/or activity or, indirectly, by the dysregulated expression of miRNAs implicated in glucose homeostasis.
Collapse
Affiliation(s)
- Sanaa Sharari
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha 34110, Qatar
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha 26999, Qatar
| | | | - Idris Mohammed
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha 34110, Qatar
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha 26999, Qatar
| | - Basma Haris
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha 26999, Qatar
| | | | - Iman Hawari
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha 34110, Qatar
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha 26999, Qatar
| | | | | | - Sara Tomei
- Research Branch, Sidra Medicine, Doha 26999, Qatar
| | | | - Najeeb Syed
- Research Branch, Sidra Medicine, Doha 26999, Qatar
| | - Sabah Nisar
- Research Branch, Sidra Medicine, Doha 26999, Qatar
| | - Selma Maacha
- Research Branch, Sidra Medicine, Doha 26999, Qatar
| | | | | | - Johan Ericsson
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha 34110, Qatar
- School of Medicine and Medical Science, University College Dublin, Belfield, 4 D4 Dublin, Ireland
| | - Khalid Hussain
- Department of Pediatric Medicine, Division of Endocrinology, Sidra Medicine, Doha 26999, Qatar
- Correspondence:
| |
Collapse
|
54
|
Diagnostic and Therapeutic Roles of Extracellular Vesicles in Aging-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6742792. [PMID: 35979398 PMCID: PMC9377967 DOI: 10.1155/2022/6742792] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/19/2022] [Indexed: 01/10/2023]
Abstract
Aging shows a decline in overall physical function, and cellular senescence is the powerful catalyst leading to aging. Considering that aging will be accompanied with the emergence of various aging-related diseases, research on new antiaging drugs is still valuable. Extracellular vesicles (EVs), as tools for intercellular communication, are important components of the senescence-associated secretory phenotype (SASP), and they can play pathological roles in the process of cellular senescence. In addition, EVs are similar to their original cells in functions. Therefore, EVs derived from pathological tissues or body fluids may be closely related to the progression of diseases and become potential biomarkers, while those from healthy cells may have therapeutic effects. Moreover, EVs are satisfactory drug carriers. At present, numerous studies have supported the idea that engineered EVs could improve drug targeting ability and utilization efficiency. Here, we summarize the characteristics of EVs and cellular senescence and focus on the diagnostic and therapeutic potential of EVs in various aging-related diseases, including Alzheimer disease, osteoporosis, cardiovascular disease, diabetes mellitus and its complications, and skin aging.
Collapse
|
55
|
Lee YS. The Mechanism for Adipose Endotrophin Production. Diabetes 2022; 71:1617-1619. [PMID: 35881834 PMCID: PMC9490353 DOI: 10.2337/dbi22-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022]
|
56
|
Jo W, Kim M, Oh J, Kim CS, Park C, Yoon S, Lee C, Kim S, Nam D, Park J. MicroRNA-29 Ameliorates Fibro-Inflammation and Insulin Resistance in HIF1α-Deficient Obese Adipose Tissue by Inhibiting Endotrophin Generation. Diabetes 2022; 71:1746-1762. [PMID: 35167651 DOI: 10.2337/db21-0801] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022]
Abstract
Dysregulation of extracellular matrix proteins in obese adipose tissue (AT) induces systemic insulin resistance. The metabolic roles of type VI collagen and its cleavage peptide endotrophin in obese AT are well established. However, the mechanisms regulating endotrophin generation remain elusive. Herein, we identified that several endotrophin-containing peptides (pre-endotrophins) were generated from the COL6A3 chain in a stepwise manner for the efficient production of mature endotrophin, partly through the action of hypoxia-induced matrix metalloproteinases (MMPs), including MMP2, MMP9, and MMP16. Hypoxia is an upstream regulator of COL6A3 expression and the proteolytic processing that regulates endotrophin generation. Hypoxia-inducible factor 1α (HIF1α) and the hypoxia-associated suppression of microRNA-29 (miR-29) cooperatively control the levels of COL6A3 and MMPs, which are responsible for endotrophin generation in hypoxic ATs. Adipocyte-specific Hif1α knock-out (APN-HIF1αKO) mice fed a chronic high-fat diet exhibited the significant amelioration of both local fibro-inflammation in AT and systemic insulin resistance compared with their control littermates, partly through the inhibition of endotrophin generation. Strikingly, adenovirus-mediated miR-29 overexpression in the ATs of APN-HIF1αKO mice in obesity significantly decreased endotrophin levels, suggesting that miR-29, combined with HIF1α inhibition in AT, could be a promising therapeutic strategy for treating obesity and related metabolic diseases.
Collapse
|
57
|
Lee S, Ko JH, Kim SN. The Extracellular MicroRNAs on Inflammation: A Literature Review of Rodent Studies. Biomedicines 2022; 10:1601. [PMID: 35884901 PMCID: PMC9312877 DOI: 10.3390/biomedicines10071601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammation is an indispensable biological process stimulated by infection and injuries. Inflammatory mechanisms related to extracellular vesicles (EVs), which are small membrane structures carrying various molecules, were summarized in this review. Emerging evidence from animal studies has highlighted the role of EVs in modulating inflammatory responses, by transporting various molecules involved in host defense. In this review, we have discussed the role of EV miRNAs in inflammation. Rodent studies associated with extracellular miRNAs in inflammatory diseases, published from 2012 to 2022, were explored from PUBMED, EMBASE, and MEDLINE. A total of 95 studies were reviewed. In summary, EV-associated miRNAs play a key role in various diseases, including organ injury, immune dysfunction, neurological disease, metabolic syndrome, vesicular disease, arthritis, cancer, and other inflammatory diseases. Diverse EV-associated miRNAs regulate inflammasome activation and pro- and anti-inflammatory cytokine levels by targeting genes.
Collapse
Affiliation(s)
- Seri Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
- Graduate School, Dongguk University, Seoul 04620, Korea
| | - Jade Heejae Ko
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
| | - Seung-Nam Kim
- College of Korean Medicine, Dongguk University, Goyang 10326, Korea; (S.L.); (J.H.K.)
| |
Collapse
|
58
|
Bauer KM, Round JL, O'Connell RM. No small matter: emerging roles for exosomal miRNAs in the immune system. FEBS J 2022; 289:4021-4037. [PMID: 34087046 PMCID: PMC9545694 DOI: 10.1111/febs.16052] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/25/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Extracellular communication is critical to the function of an organism. Exosomes, small lipid extracellular vesicles, have been recently appreciated to participate in this vital function. Within these vesicles lie critical bioactive molecules including mRNAs, proteins, and a plethora of noncoding RNAs, including microRNAs (miRNAs). Exosomal miRNAs have been shown to be produced by, trafficked between, and function in many distinct donor and recipient cell types, including cells of the immune system. For instance, loss of these critical communicators can alter the cellular response to endotoxin, and when tumor cells lose the ability to secrete these vesicles, the immune system is able to effectively suppress tumor growth. This review will highlight key findings on the known communication to and from the immune system, highlighting exosomal miRNA research in macrophages, dendritic cells, B lymphocytes, and T cells. Additionally, we will focus on three major areas of exosomal studies that involve immune responses including mucosal barriers, adipose tissue, and the tumor microenvironment. These environments are heterogeneous and dynamic, and rapidly respond to the microbiota, metabolic shifts, and immunotherapies, respectively. It is clear that exosomal miRNAs play pivotal roles in regulating cross-talk between cells in these tissues, and this represents a novel layer of cellular communication proving critical in human health and disease.
Collapse
Affiliation(s)
- Kaylyn M. Bauer
- Divison of Microbiology and ImmunologyDepartment of PathologyUniversity of UtahSalt Lake CityUTUSA
| | - June L. Round
- Divison of Microbiology and ImmunologyDepartment of PathologyUniversity of UtahSalt Lake CityUTUSA
- Hunstman Cancer InstituteUniversity of UtahSalt Lake CityUTUSA
| | - Ryan M. O'Connell
- Divison of Microbiology and ImmunologyDepartment of PathologyUniversity of UtahSalt Lake CityUTUSA
- Hunstman Cancer InstituteUniversity of UtahSalt Lake CityUTUSA
| |
Collapse
|
59
|
Dalgaard LT, Sørensen AE, Hardikar AA, Joglekar MV. The microRNA-29 family - role in metabolism and metabolic disease. Am J Physiol Cell Physiol 2022; 323:C367-C377. [PMID: 35704699 DOI: 10.1152/ajpcell.00051.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The microRNA-29a family members miR-29a-3p, miR-29b-3p and miR-29c-3p are ubiquitously expressed and consistently increased in various tissues and cell types in conditions of metabolic disease; obesity, insulin resistance and type 2 diabetes. In pancreatic beta cells, miR-29a is required for normal exocytosis, but increased levels are associated with impaired beta cell function. Similarly, in liver miR-29 species are higher in models of insulin resistance and type 2 diabetes, and either knock-out or depletion using a microRNA inhibitor improves hepatic insulin resistance. In skeletal muscle, miR-29 upregulation is associated with insulin resistance and altered substrate oxidation, and similarly, in adipocytes over-expression of miR-29a leads to insulin resistance. Blocking miR-29a using nucleic acid antisense therapeutics show promising results in preclinical animal models of obesity and type 2 diabetes, although the widespread expression pattern of miR-29 family members complicates the exploration of single target tissues. However, in fibrotic diseases, such as in late complications of diabetes and metabolic disease (diabetic kidney disease, non-alcoholic steatohepatitis), miR-29 expression is suppressed by TGFβ allowing increased extracellular matrix collagen to form. In the clinical setting circulating levels of miR-29a and miR-29b are consistently increased in type 2 diabetes and in gestational diabetes, and are also possible prognostic markers for deterioration of glucose tolerance. In conclusion, miR-29 plays an essential role in various organs relevant to intermediary metabolism and its upregulation contribute to impaired glucose metabolism, while it suppresses fibrosis development. Thus, a correct balance of miR-29a levels seems important for cellular and organ homeostasis in metabolism.
Collapse
Affiliation(s)
- Louise T Dalgaard
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Anja E Sørensen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Mugdha V Joglekar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| |
Collapse
|
60
|
Karimova MV, Gvazava IG, Vorotelyak EA. Overcoming the Limitations of Stem Cell-Derived Beta Cells. Biomolecules 2022; 12:biom12060810. [PMID: 35740935 PMCID: PMC9221417 DOI: 10.3390/biom12060810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
Great advances in type 1 diabetes (T1D) and type 2 diabetes (T2D) treatment have been made to this day. However, modern diabetes therapy based on insulin injections and cadaveric islets transplantation has many disadvantages. That is why researchers are developing new methods to regenerate the pancreatic hormone-producing cells in vitro. The most promising approach is the generation of stem cell-derived beta cells that could provide an unlimited source of insulin-secreting cells. Recent studies provide methods to produce beta-like cell clusters that display glucose-stimulated insulin secretion—one of the key characteristics of the beta cell. However, in comparison with native beta cells, stem cell-derived beta cells do not undergo full functional maturation. In this paper we review the development and current state of various protocols, consider advantages, and propose ways to improve them. We examine molecular pathways, epigenetic modifications, intracellular components, and the microenvironment as a possible leverage to promote beta cell functional maturation. A possibility to create islet organoids from stem cell-derived components, as well as their encapsulation and further transplantation, is also examined. We try to combine modern research on beta cells and their crosstalk to create a holistic overview of developing insulin-secreting systems.
Collapse
Affiliation(s)
- Mariana V. Karimova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.K.); (I.G.G.)
| | - Inessa G. Gvazava
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.K.); (I.G.G.)
| | - Ekaterina A. Vorotelyak
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia; (M.V.K.); (I.G.G.)
- Department of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence:
| |
Collapse
|
61
|
Wang Y, Liu X, Xia P, Li Z, FuChen X, Shen Y, Yu P, Zhang J. The Regulatory Role of MicroRNAs on Phagocytes: A Potential Therapeutic Target for Chronic Diseases. Front Immunol 2022; 13:901166. [PMID: 35634335 PMCID: PMC9130478 DOI: 10.3389/fimmu.2022.901166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/19/2022] [Indexed: 11/27/2022] Open
Abstract
An effective acute inflammatory response results in the elimination of infectious microorganisms, followed by a smooth transition to resolution and repair. During the inflammatory response, neutrophils play a crucial role in antimicrobial defense as the first cells to reach the site of infection damage. However, if the neutrophils that have performed the bactericidal effect are not removed in time, the inflammatory response will not be able to subside. Anti-inflammatory macrophages are the main scavengers of neutrophils and can promote inflammation towards resolution. MicroRNAs (miRNAs) have great potential as clinical targeted therapy and have attracted much attention in recent years. This paper summarizes the involvement of miRNAs in the process of chronic diseases such as atherosclerosis, rheumatoid arthritis and systemic lupus erythematosus by regulating lipid metabolism, cytokine secretion, inflammatory factor synthesis and tissue repair in two types of cells. This will provide a certain reference for miRNA-targeted treatment of chronic diseases.
Collapse
Affiliation(s)
- Yongbo Wang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Xingyu Liu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Xinxi FuChen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yunfeng Shen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
62
|
Xiao YC, Wang W, Gao Y, Li WY, Tan X, Wang YK, Wang WZ. The Peripheral Circulating Exosomal microRNAs Related to Central Inflammation in Chronic Heart Failure. J Cardiovasc Transl Res 2022; 15:500-513. [PMID: 35501543 DOI: 10.1007/s12265-022-10266-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 04/21/2022] [Indexed: 12/11/2022]
Abstract
Sympathetic hyperactivity plays an important role in the progression of chronic heart failure (CHF). It is reported that inflammation in the rostral ventrolateral medulla (RVLM), a key region for sympathetic control, excites the activity of neurons and leads to an increase in sympathetic outflow. Exosome, as the carrier of microRNAs (miRNAs), has the function of crossing the blood-brain barrier. The present study was designed to investigate the effect of exosomal miRNAs on central inflammation via peripheral-central interaction in CHF. The miRNA microarray detection was performed to compare the difference between circulating exosomes and the RVLM in CHF rats. It was shown that the expression of miR-214-3p was significantly up-regulated, whereas let-7g-5p and let-7i-5p were significantly down-regulated in circulating exosomes and the RVLM. Further studies in PC12 cells revealed that miR-214-3p enhanced the inflammatory response, while let-7g-5p and let-7i-5p reduced the neuroinflammation. The direct interaction between the miRNA and its inflammatory target gene (miR-214-3p, Traf3; let-7g-5p, Smad2; and let-7i-5p, Mapk6) was confirmed by the dual-luciferase reporter assay. These results suggest that the circulating exosomes participate in the enhancement of inflammatory response in the RVLM through their packaged miRNAs, which may further contribute to sympathetic hyperactivity in CHF.
Collapse
Affiliation(s)
- Yu-Chen Xiao
- Department of Marine Biomedicine and Polar Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Wen Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yuan Gao
- Department of Marine Biomedicine and Polar Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Wan-Yang Li
- School of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Xing Tan
- Department of Marine Biomedicine and Polar Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yang-Kai Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, China.
| | - Wei-Zhong Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, China.
| |
Collapse
|
63
|
Fu K, Zhou H, Wang C, Gong L, Ma C, Zhang Y, Li Y. A review: Pharmacology and pharmacokinetics of Schisandrin A. Phytother Res 2022; 36:2375-2393. [DOI: 10.1002/ptr.7456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/20/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
64
|
Wang X, Zhang Z, Wang M. MiR-29a regulates cardiomyocyte apoptosis by targeting Bak1 in diabetic cardiomyopathy. J Biochem 2022; 171:663-671. [PMID: 35274138 DOI: 10.1093/jb/mvac025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/06/2022] [Indexed: 11/14/2022] Open
Abstract
This study sought to investigate the association between microRNA-29a (miR-29a) and cardiomyocyte apoptosis in diabetic cardiomyopathy (DCM). DCM rat model was established by treating rats with streptozotocin (STZ), followed by injection of NC or miR-29a-3p mimics into the myocardium of rats. High glucose (HG)-treated H9c2 cells were transfected with NC and miR-29a-3p mimics. DCM rats presented elevated levels of blood glucose, HbA1c, and blood pressure and urine output and decreased body weight and cardiac contractile function after modeling. MiR-29a was lowly expressed in STZ-treated rats and HG-treated H9c2 cells. Upregulation of miR-29a improved cardiac structure and function and attenuated, alleviated myocardial histological abnormalities and fibrosis, and lowered cardiomyocyte apoptosis in DCM rats. Meanwhile, HG promoted H9c2 cell apoptosis, while miR-29a overexpression attenuated the function of HG. Compared with control group, the protein expression of Bax, cleaved-caspase3 and Bak1 in DCM and HG group were significantly upregulated, and the expression of Bcl-2 and Mcl-1 were downregulated, while miR-29a overexpression exerted opposite effect. Bioinformatics prediction method and western blot revealed that miR-29a directly targeted Bak1 and downregulated Bak1 expression. Overall, miR-29a regulated STZ- and HG-induced cardiomyocyte apoptosis by targeting Bak1, providing a novel understanding of the pathogenesis of DCM.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050004, China.,Department of Cardiology, The Third Hospital of Shijiazhuang Affiliated to Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Zhitao Zhang
- Department of Experimental Center, Hebei Medical University Clinical College, Shijiazhuang, Hebei 050031, China
| | - Mei Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050004, China
| |
Collapse
|
65
|
Makhijani P, McGaha TL. Myeloid Responses to Extracellular Vesicles in Health and Disease. Front Immunol 2022; 13:818538. [PMID: 35320943 PMCID: PMC8934876 DOI: 10.3389/fimmu.2022.818538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/15/2022] [Indexed: 01/04/2023] Open
Abstract
Extracellular vesicles are mediators of cell-cell communication playing a key role in both steady-state and disease conditions. Extracellular vesicles carry diverse donor-derived cargos, including DNA, RNA, proteins, and lipids that induce a complex network of signals in recipient cells. Due to their ability to capture particulate matter and/or capacity to polarize and orchestrate tissue responses, myeloid immune cells (e.g., dendritic cells, macrophages, etc.) rapidly respond to extracellular vesicles, driving local and systemic effects. In cancer, myeloid-extracellular vesicle communication contributes to chronic inflammation, self-tolerance, and therapeutic resistance while in autoimmune disease, extracellular vesicles support inflammation and tissue destruction. Here, we review cellular mechanisms by which extracellular vesicles modulate myeloid immunity in cancer and autoimmune disease, highlighting some contradictory results and outstanding questions. We will also summarize how understanding of extracellular vesicle biology is being utilized for novel therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Priya Makhijani
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Tumor Immunotherapy Program, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Tracy L. McGaha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Tumor Immunotherapy Program, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- *Correspondence: Tracy L. McGaha,
| |
Collapse
|
66
|
Entezari M, Hashemi D, Taheriazam A, Zabolian A, Mohammadi S, Fakhri F, Hashemi M, Hushmandi K, Ashrafizadeh M, Zarrabi A, Ertas YN, Mirzaei S, Samarghandian S. AMPK signaling in diabetes mellitus, insulin resistance and diabetic complications: A pre-clinical and clinical investigation. Biomed Pharmacother 2022; 146:112563. [PMID: 35062059 DOI: 10.1016/j.biopha.2021.112563] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM) is considered as a main challenge in both developing and developed countries, as lifestyle has changed and its management seems to be vital. Type I and type II diabetes are the main kinds and they result in hyperglycemia in patients and related complications. The gene expression alteration can lead to development of DM and related complications. The AMP-activated protein kinase (AMPK) is an energy sensor with aberrant expression in various diseases including cancer, cardiovascular diseases and DM. The present review focuses on understanding AMPK role in DM. Inducing AMPK signaling promotes glucose in DM that is of importance for ameliorating hyperglycemia. Further investigation reveals the role of AMPK signaling in enhancing insulin sensitivity for treatment of diabetic patients. Furthermore, AMPK upregulation inhibits stress and cell death in β cells that is of importance for preventing type I diabetes development. The clinical studies on diabetic patients have shown the role of AMPK signaling in improving diabetic complications such as brain disorders. Furthermore, AMPK can improve neuropathy, nephropathy, liver diseases and reproductive alterations occurring during DM. For exerting such protective impacts, AMPK signaling interacts with other molecular pathways such as PGC-1α, PI3K/Akt, NOX4 and NF-κB among others. Therefore, providing therapeutics based on AMPK targeting can be beneficial for amelioration of DM.
Collapse
Affiliation(s)
- Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Danial Hashemi
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirhossein Zabolian
- Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Golestan, Iran
| | - Shima Mohammadi
- Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Farima Fakhri
- Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonosis, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer 34396, Istanbul, Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Turkey
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
67
|
Leitão AL, Enguita FJ. A Structural View of miRNA Biogenesis and Function. Noncoding RNA 2022; 8:ncrna8010010. [PMID: 35202084 PMCID: PMC8874510 DOI: 10.3390/ncrna8010010] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/16/2022] Open
Abstract
Micro-RNAs (miRNAs) are a class of non-coding RNAs (ncRNAs) that act as post-transcriptional regulators of gene expression. Since their discovery in 1993, they have been the subject of deep study due to their involvement in many important biological processes. Compared with other ncRNAs, miRNAs are generated from devoted transcriptional units which are processed by a specific set of endonucleases. The contribution of structural biology methods for understanding miRNA biogenesis and function has been essential for the dissection of their roles in cell biology and human disease. In this review, we summarize the application of structural biology for the characterization of the molecular players involved in miRNA biogenesis (processors and effectors), starting from the X-ray crystallography methods to the more recent cryo-electron microscopy protocols.
Collapse
Affiliation(s)
- Ana Lúcia Leitão
- MEtRICs, Department of Sciences and Technology of Biomass, NOVA School of Science and Technology, FCT NOVA, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
68
|
Tang W, Guo ZD, Chai WN, Du DL, Yang XM, Cao L, Chen H, Zhou C, Cheng CJ, Sun XC, Huang ZJ, Zhong JJ. Downregulation of miR-491-5p promotes neovascularization after traumatic brain injury. Neural Regen Res 2022; 17:577-586. [PMID: 34380897 PMCID: PMC8504397 DOI: 10.4103/1673-5374.314326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
MicroRNA-491-5p (miR-491-5p) plays an important role in regulating cell proliferation and migration; however, the effect of miR-491-5p on neovascularization after traumatic brain injury remains poorly understood. In this study, a controlled cortical injury model in C57BL/6 mice and an oxygen-glucose deprivation model in microvascular endothelial cells derived from mouse brain were established to simulate traumatic brain injury in vivo and in vitro, respectively. In the in vivo model, quantitative real-time-polymerase chain reaction results showed that the expression of miR-491-5p increased or decreased following the intracerebroventricular injection of an miR-491-5p agomir or antagomir, respectively, and the expression of miR-491-5p decreased slightly after traumatic brain injury. To detect the neuroprotective effects of miR-491-p, neurological severity scores, Morris water maze test, laser speckle techniques, and immunofluorescence staining were assessed, and the results revealed that miR-491-5p downregulation alleviated neurological dysfunction, promoted the recovery of regional cerebral blood flow, increased the number of lectin-stained microvessels, and increased the survival of neurons after traumatic brain injury. During the in vitro experiments, the potential mechanism of miR-491-5p on neovascularization was explored through quantitative real-time-polymerase chain reaction, which showed that miR-491-5p expression increased or decreased in brain microvascular endothelial cells after transfection with an miR-491-5p mimic or inhibitor, respectively. Dual-luciferase reporter and western blot assays verified that metallothionein-2 was a target gene for miR-491-5p. Cell counting kit 8 (CCK-8) assay, flow cytometry, and 2?,7?-dichlorofluorescein diacetate (DCFH-DA) assay results confirmed that the downregulation of miR-491-5p increased brain microvascular endothelial cell viability, reduced cell apoptosis, and alleviated oxidative stress under oxygen-glucose deprivation conditions. Cell scratch assay, Transwell assay, tube formation assay, and western blot assay results demonstrated that miR-491-5p downregulation promoted the migration, proliferation, and tube formation of brain microvascular endothelial cells through a metallothionein-2-dependent hypoxia-inducible factor-1α/vascular endothelial growth factor pathway. These findings confirmed that miR-491-5p downregulation promotes neovascularization, restores cerebral blood flow, and improves the recovery of neurological function after traumatic brain injury. The mechanism may be mediated through a metallothionein-2-dependent hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway and the alleviation of oxidative stress. All procedures were approved by Ethics Committee of the First Affiliated Hospital of Chongqing Medical University, China (approval No. 2020-304) on June 22, 2020.
Collapse
Affiliation(s)
- Wei Tang
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zong-Duo Guo
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei-Na Chai
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong-Lin Du
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Min Yang
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lang Cao
- Department of Ophthalmology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Zhou
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chong-Jie Cheng
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Chuan Sun
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Jian Huang
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Jun Zhong
- Department of Neurosurgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
69
|
Xiong L, Chen L, Wu L, He W, Chen D, Peng Z, Li J, Zhu X, Su L, Li Y, Gong Y, Xiao H. Lipotoxicity-induced circGlis3 impairs beta cell function and is transmitted by exosomes to promote islet endothelial cell dysfunction. Diabetologia 2022; 65:188-205. [PMID: 34751796 DOI: 10.1007/s00125-021-05591-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/30/2021] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Lipotoxicity constitutes the major driving force for type 2 diabetes. Circular RNAs (circRNAs) play important roles in regulating beta cell function and exosomes are essential mediators of intercellular communication. The role of exosomal circRNAs in type 2 diabetes remains largely unknown. We aimed to examine whether lipotoxicity induces dysregulation of circRNAs in beta cell-derived exosomes and to determine the contribution of exosomal circRNAs to the development of type 2 diabetes. METHODS Exosomes were extracted from MIN6 cells treated with palmitate or BSA, and RNA sequencing was performed. CircGlis3 (Gli-similar 3) expression level was validated by qPCR. The impact of circGlis3 on beta cell function and the deleterious effects of exosomal circGlis3 on islet endothelial cells (islet ECs) were investigated in vitro and in vivo in human and mouse models by gain or loss of function assays. The molecular mechanism of circGlis3 was explored by RNA pull-down and immunoprecipitation assays. RESULTS Beta cell-derived exosomal circGlis3 was significantly upregulated under lipotoxic conditions, and exosomal circGlis3 levels were also elevated in the serum of mouse models of diabetes and participants with type 2 diabetes. CircGlis3 participated in lipotoxicity-induced beta cell dysfunction in vitro and in vivo. Moreover, beta cell-derived exosomal circGlis3 could be transferred to islet ECs and reduce the cell viability, cell migration and angiogenesis of islet ECs. Mechanistically, circGlis3 promoted the degradation of glucocorticoid modulatory element-binding protein 1 (GMEB1) by facilitating the interaction between GMEB1 and mindbomb E3 ubiquitin protein ligase 2 (MIB2), thus suppressing the phosphorylation of heat shock protein 27 (HSP27). CONCLUSIONS/INTERPRETATION Our study points to the involvement of circGlis3 in diabetes development, and exosomal circGlis3 transfer as a communication mode between beta cells and islet ECs, suggesting that circGlis3 might be a potential biomarker and therapeutic target for type 2 diabetes. DATA AVAILABILITY The RNA-sequencing data have been deposited in the NCBI Sequence Read Archive (SRA) database, with accession number PRJNA689673. Mass spectrometry data are available via ProteomeXchange with identifier PXD024693.
Collapse
Affiliation(s)
- Li Xiong
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Chen
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liting Wu
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiman He
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dubo Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zishan Peng
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jin Li
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaonan Zhu
- Department of Pharmacology, Zhong‑Shan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong, China
| | - Lei Su
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingying Gong
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Haipeng Xiao
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
70
|
Li C, Wang D, Jiang Z, Gao Y, Sun L, Li R, Chen M, Lin C, Liu D. Non-coding RNAs in diabetes mellitus and diabetic cardiovascular disease. Front Endocrinol (Lausanne) 2022; 13:961802. [PMID: 36147580 PMCID: PMC9487522 DOI: 10.3389/fendo.2022.961802] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
More than 10% of the world's population already suffers from varying degrees of diabetes mellitus (DM), but there is still no cure for the disease. Cardiovascular disease (CVD) is one of the most common and dangerous of the many health complications that can be brought on by DM, and has become the leading cause of death in people with diabetes. While research on DM and associated CVD is advancing, the specific mechanisms of their development are still unclear. Given the threat of DM and CVD to humans, the search for new predictive markers and therapeutic ideas is imminent. Non-coding RNAs (ncRNAs) have been a popular subject of research in recent years. Although they do not encode proteins, they play an important role in living organisms, and they can cause disease when their expression is abnormal. Numerous studies have observed aberrant ncRNAs in patients with DM complications, suggesting that they may play an important role in the development of DM and CVD and could potentially act as biomarkers for diagnosis. There is additional evidence that treatment with existing drugs for DM, such as metformin, alters ncRNA expression levels, suggesting that regulation of ncRNA expression may be a key mechanism in future DM treatment. In this review, we assess the role of ncRNAs in the development of DM and CVD, as well as the evidence for ncRNAs as potential therapeutic targets, and make use of bioinformatics to analyze differential ncRNAs with potential functions in DM.
Collapse
Affiliation(s)
- Chengshun Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Ziping Jiang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yongjian Gao
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Liqun Sun
- Department of Pediatrics, First Hospital of Jilin University, Changchun, China
| | - Rong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Minqi Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dianfeng Liu
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
- *Correspondence: Dianfeng Liu,
| |
Collapse
|
71
|
Gauthier BR, Cobo-Vuilleumier N, López-Noriega L. Roles of extracellular vesicles associated non-coding RNAs in Diabetes Mellitus. Front Endocrinol (Lausanne) 2022; 13:1057407. [PMID: 36619588 PMCID: PMC9814720 DOI: 10.3389/fendo.2022.1057407] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs), especially exosomes (50 to 150 nm), have been shown to play important roles in a wide range of physiological and pathological processes, including metabolic diseases such as Diabetes Mellitus (DM). In the last decade, several studies have demonstrated how EVs are involved in cell-to-cell communication. EVs are enriched in proteins, mRNAs and non-coding RNAs (miRNAs, long non-coding RNAs and circRNAS, among others) which are transferred to recipient cells and may have a profound impact in either their survival or functionality. Several studies have pointed out the contribution of exosomal miRNAs, such as miR-l42-3p and miR-26, in the development of Type 1 and Type 2 DM (T1DM and T2DM), respectively. In addition, some miRNA families such as miR-let7 and miR-29 found in exosomes have been associated with both types of diabetes, suggesting that they share common etiological features. The knowledge about the role of exosomal long non-coding RNAs in this group of diseases is more immature, but the exosomal lncRNA MALAT1 has been found to be elevated in the plasma of individuals with T2DM, while more than 169 lncRNAs were reported to be differentially expressed between healthy donors and people with T1DM. Here, we review the current knowledge about exosomal non-coding RNAs in DM and discuss their potential as novel biomarkers and possible therapeutic targets.
Collapse
Affiliation(s)
- Benoit R. Gauthier
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Madrid, Spain
- *Correspondence: Benoit R. Gauthier, ; Livia López-Noriega,
| | - Nadia Cobo-Vuilleumier
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - Livia López-Noriega
- Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- *Correspondence: Benoit R. Gauthier, ; Livia López-Noriega,
| |
Collapse
|
72
|
Liu Q, Wang M, Xu T, Liang W, Yang F. Significance of serum miR-29a in the occurrence and progression of diabetic nephropathy: A cross-sectional study. J Clin Lab Anal 2021; 36:e24210. [PMID: 34964177 PMCID: PMC8842137 DOI: 10.1002/jcla.24210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN), a common microvascular complication of type 2 diabetes mellitus (T2DM), is an important factor causing chronic kidney disease. However, the relationship between miR-29a and DN remains unknown. Therefore, a cross-sectional study was conducted to identify a potential molecular biomarker for DN prevention and management by detecting the serum miR-29a levels. METHODS The serum miR-29a levels were measured in 360 subjects (180 T2DM patients and 180 healthy controls) using quantitative reverse transcription PCR (qRT-PCR), and other conventional indicators were measured and analysed. A binary logistic regression was used to evaluate the DN risk factors; a receiver operating characteristic (ROC) curve was applied to analyse the diagnostic efficacy of miR-29a for DN, and a Spearman's rank correlation analysis was used to evaluate the correlation between serum miR-29a and cystatin C. RESULTS The serum miR-29 levels in the T2DM patients were higher than those in the healthy subjects and significantly increased with the progression of DN (p < 0.05). Serum miR-29a and cystatin C are independent predictors of the occurrence of DN. Compared with a single indicator, the combination of serum miR-29a and cystatin C has better DN diagnostic performance. In addition, the serum miR-29a levels were positively correlated with cystatin C in the patients with DN (r = 0.521, p < 0.001). CONCLUSION The expression of serum miR-29a was significantly associated with the occurrence and progression of DN and is expected to become a potential biomarker for the diagnosis of DN.
Collapse
Affiliation(s)
- Qian Liu
- Department of Laboratory Medicine, The Second People's Hospital of Lianyungang, Lianyungang, China.,Department of Laboratory Medicine, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, China
| | - Menglin Wang
- Department of Laboratory Medicine, Suqian First Hospital, Suqian, China
| | - Tongdao Xu
- Department of Endocrinology, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Wei Liang
- Department of Laboratory Medicine, The Second People's Hospital of Lianyungang, Lianyungang, China.,Department of Laboratory Medicine, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, China
| | - Fumeng Yang
- Department of Laboratory Medicine, The Second People's Hospital of Lianyungang, Lianyungang, China.,Department of Laboratory Medicine, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, China
| |
Collapse
|
73
|
Zhang S, Xu M, Shen Z, Shang C, Zhang W, Chen S, Liu C. Green light exposure aggravates high-fat diet feeding-induced hepatic steatosis and pancreatic dysfunction in male mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112802. [PMID: 34555719 DOI: 10.1016/j.ecoenv.2021.112802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
The increased incidence of metabolic syndrome (MetS) has been demonstrated to be closely associated with external environments, such as unhealthy ambient light exposure. Of note, spectral distribution of the light functions as a critical determinant of light's pathophysiological effects. However, the effects of the lighting spectrum on metabolic homeostasis and the specific target organs remain elusive. To address this concern, we in this study high-fat diet (HFD)-fed obese mice with different spectra of the light, and divided them into white light (WL)-treated group, green light (GL)-treated group and blue light (BL)-treated group. We found that compared with BL- or WL-treated obese mice, animals exposed to GL showed worsened metabolic status, including increased body weight gain, impaired glucose tolerance/insulin sensitivity, increased levels of serum lipids, and decreased levels of serum insulin. At the organ level, GL exposure particularly exacerbated hepatic lipid accumulation and enlarged the islet volume. Taking advantages of metabolomics and transcriptomics analyses, we screened out taurocholic acid (TCA) and adenosine (AD) as two promising metabolites mediating the deleterious effects of GL on the liver and islets, respectively. In detail, GL aggravates HFD-induced lipid synthesis and gluconeogenesis in the liver via the reduction of TCA, while triggering inflammation and cellular dysfunction in islets via the induction of AD. Collectively, our findings confirmed that GL and the HFD have a synergistic effect in the induction of metabolic disorders. DATA AVAILABILITY: All data supported the paper are present in the paper and/or the Supplementary Materials. The original datasets are also available from the corresponding author upon request.
Collapse
Affiliation(s)
- Shiyao Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengyi Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ziyue Shen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Changrui Shang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Chang Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology and Experimental Platform for Drug Chronological Intervention and Therapy, China Pharmaceutical University, Nanjing, Jiangsu, China; Key Laboratory of Active Components of Natural Medicine and Drug Release Technology, School of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
74
|
Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct Target Ther 2021; 6:383. [PMID: 34753929 PMCID: PMC8578673 DOI: 10.1038/s41392-021-00779-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes play a role as mediators of cell-to-cell communication, thus exhibiting pleiotropic activities to homeostasis regulation. Exosomal non-coding RNAs (ncRNAs), mainly microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are closely related to a variety of biological and functional aspects of human health. When the exosomal ncRNAs undergo tissue-specific changes due to diverse internal or external disorders, they can cause tissue dysfunction, aging, and diseases. In this review, we comprehensively discuss the underlying regulatory mechanisms of exosomes in human diseases. In addition, we explore the current knowledge on the roles of exosomal miRNAs, lncRNAs, and circRNAs in human health and diseases, including cancers, metabolic diseases, neurodegenerative diseases, cardiovascular diseases, autoimmune diseases, and infectious diseases, to determine their potential implication in biomarker identification and therapeutic exploration.
Collapse
|
75
|
Al-Selwi Y, Shaw JA, Kattner N. Understanding the Pancreatic Islet Microenvironment in Cystic Fibrosis and the Extrinsic Pathways Leading to Cystic Fibrosis Related Diabetes. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2021; 14:11795514211048813. [PMID: 34675737 PMCID: PMC8524685 DOI: 10.1177/11795514211048813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022]
Abstract
Cystic fibrosis (CF) is an autosomal recessive chronic condition
effecting approximately 70 000 to 100 000 people globally and is
caused by a loss-of-function mutation in the CF transmembrane
conductance regulator. Through improvements in clinical care, life
expectancy in CF has increased considerably associated with rising
incidence of secondary complications including CF-related diabetes
(CFRD). CFRD is believed to result from β-cell loss as well as
insufficient insulin secretion due to β-cell dysfunction, but the
underlying pathophysiology is not yet fully understood. Here we review
the morphological and cellular changes in addition to the
architectural remodelling of the pancreatic exocrine and endocrine
compartments in CF and CFRD pancreas. We consider also potential
underlying proinflammatory signalling pathways impacting on endocrine
and specifically β-cell function, concluding that further research
focused on these mechanisms may uncover novel therapeutic targets
enabling restoration of normal insulin secretion.
Collapse
Affiliation(s)
- Yara Al-Selwi
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James Am Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Nicole Kattner
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
76
|
An W, Huang Y, Chen S, Teng T, Shi Y, Sun Z, Xu Y. Mechanisms of Rhizoma Coptidis against type 2 diabetes mellitus explored by network pharmacology combined with molecular docking and experimental validation. Sci Rep 2021; 11:20849. [PMID: 34675276 PMCID: PMC8531350 DOI: 10.1038/s41598-021-00293-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/05/2021] [Indexed: 02/08/2023] Open
Abstract
This study systematically explored the underlying mechanism of Rhizoma Coptidis against type 2 diabetes mellitus (T2DM) by using network pharmacology and molecular docking and experimental validation. We retrieved and screened active compounds of Rhizoma Coptidis and corresponding T2DM-related targets across multiple databases. PPI networks of the genes were constructed using STRING, and the core targets were screened via topological analysis. GO and KEGG enrichment analyses were performed by using DAVID. Finally, molecular docking and experimental studies were performed after bioinformatic analysis for verification. There were 14 active compounds and 19 core targets of Rhizoma Coptidis-T2DM, of which quercetin was identified as the main compound and IL6, VEGFA and TNF were the most significant core targets. GO and KEGG enrichment analyses showed that Rhizoma Coptidis ameliorated T2DM by regulating multiple biological processes and pathways. Docking studies indicated that IL6, VEGFA and TNF could stably bind with all active compounds of Rhizoma Coptidis. The results of our experiments revealed that Rhizoma Coptidis could inhibit the expression of IL6 and TNFα and enhance islet cell viability. This study suggests anti-inflammatory therapeutic effects of Rhizoma Coptidis on T2DM, thereby providing a scientific basis and new insight for further research on the antidiabetic effect of Rhizoma Coptidis.
Collapse
Affiliation(s)
- Wenrong An
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yanqin Huang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Shouqiang Chen
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 1 Jingba Road, Jinan, 250001, China
| | - Tao Teng
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 1 Jingba Road, Jinan, 250001, China
| | - Yingning Shi
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhenhai Sun
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 1 Jingba Road, Jinan, 250001, China.
| |
Collapse
|
77
|
Zhu YX, Zhou YC, Zhang Y, Sun P, Chang XA, Han X. Protocol for in vivo and ex vivo assessments of glucose-stimulated insulin secretion in mouse islet β cells. STAR Protoc 2021; 2:100728. [PMID: 34409308 PMCID: PMC8361272 DOI: 10.1016/j.xpro.2021.100728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pancreatic islet β cells secrete insulin in a biphasic manner when sensing high blood glucose level. This protocol describes the evaluation of different phases of insulin secretion, as well as basal, glucose-stimulated and total insulin secretion abilities, thereby enabling precise assessment of β cell function both in vivo and ex vivo. The in vivo assay consists of intravenous tube imbedding surgery and hyperglycemic clamp. The ex vivo assay consists of islet isolation, dynamic perfusion and static immersion. For complete details on the use and execution of this protocol, please refer to Sun et al. (2021). Glucose-stimulated biphasic insulin secretion (GSIS) in mouse islet β cells Optimized and detailed tube embedding surgery for in vivo GSIS assay Careful islet isolation and verification is essential for ex vivo GSIS assay Adaptable procedure to monitor both human and murine islet β-cell function ex vivo
Collapse
Affiliation(s)
- Yun-Xia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yun-Cai Zhou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao-Ai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
78
|
Qian B, Yang Y, Tang N, Wang J, Sun P, Yang N, Chen F, Wu T, Sun T, Li Y, Chang X, Zhu Y, Zhang Y, Han X. M1 macrophage-derived exosomes impair beta cell insulin secretion via miR-212-5p by targeting SIRT2 and inhibiting Akt/GSK-3β/β-catenin pathway in mice. Diabetologia 2021; 64:2037-2051. [PMID: 34117507 DOI: 10.1007/s00125-021-05489-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/25/2021] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS Macrophage levels are elevated in pancreatic islets, and the resulting inflammatory response is a major contributor to beta cell failure during obesity and type 2 diabetes mellitus. Previous studies by us and others have reported that exosomes released by macrophages play important roles in mediating cell-to-cell communication, and represent a class of inflammatory factors involved in the inflammatory process associated with type 2 diabetes mellitus. However, to date, no reports have demonstrated the effect of macrophage-derived exosomes on beta cells, and little is known regarding their underlying mechanisms in beta cell injury. Thus, we aimed to study the impact of macrophage-derived exosomes on islet beta cell injury in vitro and in vivo. METHODS The phenotypic profiles of islet-resident macrophages were analysed in C57BL/6J mice fed a high-fat diet (HFD). Exosomes were collected from the medium of cultured bone marrow-derived macrophages (BMDMs) and from isolated islet-resident macrophages of HFD-fed mice (HFD-Exos). The role of exosomes secreted by inflammatory M1 phenotype BMDMs (M1-Exos) and HFD-Exos on beta cell function was assessed. An miRNA microarray and quantitative real-time PCR (qPCR) were conducted to test the level of M1-Exos-derived miR-212-5p in beta cells. Then, miR-212-5p was overexpressed or inhibited in M1-Exos or beta cells to determine its molecular and functional impact. RESULTS M1-polarised macrophages were enriched in the islets of obese mice. M1 macrophages and islet-resident macrophages of HFD-fed mice impaired beta cell insulin secretion in an exosome-dependent manner. miR-212-5p was notably upregulated in M1-Exos and HFD-Exos. Enhancing the expression of miR-212-5p impaired beta cell insulin secretion. Blocking miR-212-5p elicited a significant improvement in M1-Exos-mediated beta cell insulin secretion during injury. Mechanistically, M1-Exos mediated an intercellular transfer of the miR-212-5p, targeting the sirtuin 2 gene and regulating the Akt/GSK-3β/β-catenin pathway in recipient beta cells to restrict insulin secretion. CONCLUSIONS/INTERPRETATION A novel exosome-modulated mechanism was delineated for macrophage-beta cell crosstalk that drove beta cell dysfunction and should be explored for its therapeutic utility.
Collapse
Affiliation(s)
- Bin Qian
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Ningyuan Tang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Jiahui Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Nan Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Tong Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China.
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
79
|
MicroRNA-29a in Osteoblasts Represses High-Fat Diet-Mediated Osteoporosis and Body Adiposis through Targeting Leptin. Int J Mol Sci 2021; 22:ijms22179135. [PMID: 34502056 PMCID: PMC8430888 DOI: 10.3390/ijms22179135] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/14/2022] Open
Abstract
Skeletal tissue involves systemic adipose tissue metabolism and energy expenditure. MicroRNA signaling controls high-fat diet (HFD)-induced bone and fat homeostasis dysregulation remains uncertain. This study revealed that transgenic overexpression of miR-29a under control of osteocalcin promoter in osteoblasts (miR-29aTg) attenuated HFD-mediated body overweight, hyperglycemia, and hypercholesterolemia. HFD-fed miR-29aTg mice showed less bone mass loss, fatty marrow, and visceral fat mass together with increased subscapular brown fat mass than HFD-fed wild-type mice. HFD-induced O2 underconsumption, respiratory quotient repression, and heat underproduction were attenuated in miR-29aTg mice. In vitro, miR-29a overexpression repressed transcriptomic landscapes of the adipocytokine signaling pathway, fatty acid metabolism, and lipid transport, etc., of bone marrow mesenchymal progenitor cells. Forced miR-29a expression promoted osteogenic differentiation but inhibited adipocyte formation. miR-29a signaling promoted brown/beige adipocyte markers Ucp-1, Pgc-1α, P2rx5, and Pat2 expression and inhibited white adipocyte markers Tcf21 and Hoxc9 expression. The microRNA also reduced peroxisome formation and leptin expression during adipocyte formation and downregulated HFD-induced leptin expression in bone tissue. Taken together, miR-29a controlled leptin signaling and brown/beige adipocyte formation of osteogenic progenitor cells to preserve bone anabolism, which reversed HFD-induced energy underutilization and visceral fat overproduction. This study sheds light on a new molecular mechanism by which bone integrity counteracts HFD-induced whole-body fat overproduction.
Collapse
|
80
|
Zhang B, Yu L, Sheng Y. Clinical value and role of microRNA-29c-3p in sepsis-induced inflammation and cardiac dysfunction. Eur J Med Res 2021; 26:90. [PMID: 34376255 PMCID: PMC8353850 DOI: 10.1186/s40001-021-00566-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/29/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The goal of this study was to investigate the diagnostic value of miR-29c-3p in sepsis and its role in sepsis-induced inflammatory response and cardiac dysfunction. METHODS Serum level of miR-29c-3p was detected by qRT-PCR. The ROC curve was used to evaluate the diagnostic value of miR-29c-3p for Sepsis. The cecal ligation and puncture method (CLP) was used to establish a rat sepsis model. To assess cardiac function, left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP) and maximum rate of rise/fall of left ventricle pressure (± dp/dtmax) in different experimental groups were detected, and the serum cardiac troponin I (cTnI), creative kinase isoenzyme MB (CK-MB) were measured by ELISA. Meanwhile, TNF-α, IL-1β, and IL-6 were detected by ELISA to assess the level of inflammatory response in animals. RESULTS miR-29c-3p level was upregulated in sepsis patients. ROC curve revealed that miR-29c-3p had the ability to distinguish sepsis patients from healthy controls. Cardiac dysfunction and inflammation were observed in sepsis rat, which were characterized by the decrease of LVSP and + dp/dtmax, the increase of LVEDP, - dp/dtmax, cTnI, CK-MB, TNF-α, IL-1β, IL-6. All effects were reversed by the injection of miR-29c-3p antagomir. Logistics regression analysis manifested miR-29c-3p is an independent factor in the occurrence of cardiac dysfunction in sepsis patients. CONCLUSIONS miR-29c-3p has potential as a biomarker for the diagnosis of sepsis, and inhibition of miR-29c-3p expression in animal models reduced sepsis-induced cardiac dysfunction and inflammatory response.
Collapse
Affiliation(s)
- Bingyu Zhang
- Department of Critical Care Medicine, Gongli Hospital of Pudong New Area of Shanghai, Shanghai, 200135, China
| | - Lin Yu
- Department of Critical Care Medicine, Gongli Hospital of Pudong New Area of Shanghai, Shanghai, 200135, China
| | - Ying Sheng
- Department of Emergency and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800, Gongwei Road, Pudong district, Shanghai, 201399, China.
| |
Collapse
|
81
|
Veluthakal R, Thurmond DC. Emerging Roles of Small GTPases in Islet β-Cell Function. Cells 2021; 10:1503. [PMID: 34203728 PMCID: PMC8232272 DOI: 10.3390/cells10061503] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
Several small guanosine triphosphatases (GTPases) from the Ras protein superfamily regulate glucose-stimulated insulin secretion in the pancreatic islet β-cell. The Rho family GTPases Cdc42 and Rac1 are primarily involved in relaying key signals in several cellular functions, including vesicle trafficking, plasma membrane homeostasis, and cytoskeletal dynamics. They orchestrate specific changes at each spatiotemporal region within the β-cell by coordinating with signal transducers, guanine nucleotide exchange factors (GEFs), GTPase-activating factors (GAPs), and their effectors. The Arf family of small GTPases is involved in vesicular trafficking (exocytosis and endocytosis) and actin cytoskeletal dynamics. Rab-GTPases regulate pre-exocytotic and late endocytic membrane trafficking events in β-cells. Several additional functions for small GTPases include regulating transcription factor activity and mitochondrial dynamics. Importantly, defects in several of these GTPases have been found associated with type 2 diabetes (T2D) etiology. The purpose of this review is to systematically denote the identities and molecular mechanistic steps in the glucose-stimulated insulin secretion pathway that leads to the normal release of insulin. We will also note newly identified defects in these GTPases and their corresponding regulatory factors (e.g., GDP dissociation inhibitors (GDIs), GEFs, and GAPs) in the pancreatic β-cells, which contribute to the dysregulation of metabolism and the development of T2D.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| |
Collapse
|
82
|
Mattke J, Vasu S, Darden CM, Kumano K, Lawrence MC, Naziruddin B. Role of Exosomes in Islet Transplantation. Front Endocrinol (Lausanne) 2021; 12:681600. [PMID: 34447351 PMCID: PMC8382876 DOI: 10.3389/fendo.2021.681600] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/13/2021] [Indexed: 12/22/2022] Open
Abstract
Exosomes are known for their ability to transport nucleic acid, lipid, and protein molecules, which allows for communication between cells and tissues. The cargo of the exosomes can have a variety of effects on a wide range of targets to mediate biological function. Pancreatic islet transplantation is a minimally invasive cell replacement therapy to prevent or reverse diabetes mellitus and is currently performed in patients with uncontrolled type 1 diabetes or chronic pancreatitis. Exosomes have become a focus in the field of islet transplantation for the study of diagnostic markers of islet cell viability and function. A growing list of miRNAs identified from exosomes collected during the process of isolating islets can be used as diagnostic biomarkers of islet stress and damage, leading to a better understanding of critical steps of the isolation procedure that can be improved to increase islet yield and quality. Exosomes have also been implicated as a possible contributor to islet graft rejection following transplantation, as they carry donor major histocompatibility complex molecules, which are then processed by recipient antigen-presenting cells and sensed by the recipient immune cells. Exosomes may find their way into the therapeutic realm of islet transplantation, as exosomes isolated from mesenchymal stem cells have shown promising results in early studies that have seen increased viability and functionality of isolated and grafted islets in vitro as well as in vivo. With the study of exosomes still in its infancy, continued research on the role of exosomes in islet transplantation will be paramount to understanding beta cell regeneration and improving long-term graft function.
Collapse
Affiliation(s)
- Jordan Mattke
- Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | - Srividya Vasu
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Carly M. Darden
- Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | - Kenjiro Kumano
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Michael C. Lawrence
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Bashoo Naziruddin
- Baylor Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, United States
- *Correspondence: Bashoo Naziruddin,
| |
Collapse
|