1
|
Cho H, Ha SE, Singh R, Kim D, Ro S. microRNAs in Type 1 Diabetes: Roles, Pathological Mechanisms, and Therapeutic Potential. Int J Mol Sci 2025; 26:3301. [PMID: 40244147 PMCID: PMC11990060 DOI: 10.3390/ijms26073301] [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: 02/21/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by the progressive destruction of pancreatic β-cells, leading to insulin deficiency. The primary drivers of β-cell destruction in T1D involve autoimmune-mediated processes that trigger chronic inflammation and ultimately β-cell loss. Regulatory microRNAs (miRNAs) play a crucial role in modulating these processes by regulating gene expression through post-transcriptional suppression of target mRNAs. Dysregulated miRNAs have been implicated in T1D pathogenesis, serving as both potential diagnostic biomarkers and therapeutic targets. This review explores the role of miRNAs in T1D, highlighting their involvement in disease mechanisms across both rodent models and human patients. While current antidiabetic therapies manage T1D symptoms, they do not prevent β-cell destruction, leaving patients reliant on lifelong insulin therapy. By summarizing key miRNA expression profiles in diabetic animal models and patients, this review explores the potential of miRNA-based therapies to restore β-cell function and halt or slow the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Seungil Ro
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (H.C.); (S.E.H.); (R.S.); (D.K.)
| |
Collapse
|
2
|
Mostafa EA, Ismail NA, El Din Abd El Baky AMN, ElShaer TF, Ashmawy I, Wahby AA, Wahed MMA, Hamdy Abd El Aziz S. MiR-375: it could be a general biomarker of metabolic changes and inflammation in type 1 diabetes patients and their siblings. J Endocrinol Invest 2025; 48:757-764. [PMID: 39453571 PMCID: PMC11876202 DOI: 10.1007/s40618-024-02474-4] [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: 05/14/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE Type 1 diabetes (T1D) is a chronic autoimmune illness that results in loss of pancreatic beta cells and insulin insufficiency. MicroRNAs (miRNAs) are linked to immune system functions contributing to the pathophysiology of T1D, miRNA-375 is significantly expressed in the human pancreas and its circulatory levels might correspond to beta cell alterations. Pancreatic islet cell antibodies (ICA) and Glutamic acid decarboxylase antibodies (GADA) have roles in autoimmune pathogenesis and are predictive markers of T1D. The aim of this work was to detect serum level changes of miRNA-375, ICA, and GADA in T1D patients, and their siblings compared to healthy controls and correlate them with T1D biochemical parameters. METHODS The study included 66 T1D patients (32 males and 34 females; age range 3-18 years), 22 patients' siblings (13 males and 9 females; age range 4-17 years), and 23 healthy controls (7 males and 16 females; age range 4-17 years). MiRNA-375 levels were measured using quantitative reverse transcription polymerase chain reaction (RT-qPCR), while ICA and GADA levels were measured using enzyme-linked immunosorbent assay (ELISA). Data analysis was done utilizing SPSS-17 software. RESULTS MiR-375 levels were downregulated in T1D patients and further decreased in their siblings when compared to healthy controls. Furthermore, miR-375 exhibited inverse correlations with HbA1c levels but no correlations with Total Insulin Dose, disease duration, or autoantibodies (GADA & ICA). CONCLUSION Our study indicates that miR-375 is significantly downregulated in children with T1D and their siblings, suggesting its potential role as a biomarker for beta-cell function and glycemic control.
Collapse
Affiliation(s)
- Eman A Mostafa
- National Research Center, Department of Pediatrics, El Buhouth St., P. O. 12622, Dokki, Cairo, Egypt.
| | - Nagwa Abdallah Ismail
- National Research Center, Department of Pediatrics, El Buhouth St., P. O. 12622, Dokki, Cairo, Egypt
| | | | - Tarek F ElShaer
- National Research Center, Department of Pediatrics, El Buhouth St., P. O. 12622, Dokki, Cairo, Egypt
| | - Ingy Ashmawy
- National Research Center, Department of Clinical and Chemical Pathology, Cairo, Egypt
| | - Aliaa Ahmed Wahby
- National Research Center, Department of Clinical and Chemical Pathology, Cairo, Egypt
| | - Mai Magdy Abdel Wahed
- National Research Center, Department of Clinical and Chemical Pathology, Cairo, Egypt
| | | |
Collapse
|
3
|
Lalani S, Knudsen J, Kenney J, Hober D, DiPersio CM, Gerber A. A novel microRNA promotes coxsackievirus B4 infection of pancreatic β cells. Front Immunol 2024; 15:1414894. [PMID: 39697323 PMCID: PMC11652211 DOI: 10.3389/fimmu.2024.1414894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/17/2024] [Indexed: 12/20/2024] Open
Abstract
The epidemiological association of coxsackievirus B infection with type 1 diabetes suggests that therapeutic strategies that reduce viral load could delay or prevent disease onset. Moreover, recent studies suggest that treatment with antiviral agents against coxsackievirus B may help preserve insulin levels in type 1 diabetic patients. In the current study, we performed small RNA-sequencing to show that infection of immortalized trophoblast cells with coxsackievirus caused differential regulation of several miRNAs. One of these, hsa-miR-AMC1, was similarly upregulated in human pancreatic β cells infected with coxsackievirus B4. Moreover, treatment of β cells with non-cytotoxic concentrations of an antagomir that targets hsa-miR-AMC1 led to decreased CVB4 infection, suggesting a positive feedback loop wherein this microRNA further promotes viral infection. Interestingly, some predicted target genes of hsa-miR-AMC1 are shared with hsa-miR-184, a microRNA that is known to suppress genes that regulate insulin production in pancreatic β cells. Consistently, treatment of coxsackievirus B4-infected β cells with the hsa-miR-AMC1 antagomir was associated with a trend toward increased insulin production. Taken together, our findings implicate novel hsa-miR-AMC1 as a potential early biomarker of coxsackievirus B4-induced type 1 diabetes and suggest that inhibiting hsa-miR-AMC1 may provide therapeutic benefit to type 1 diabetes patients. Our findings also support the use of trophoblast cells as a model for identifying microRNAs that might be useful diagnostic markers or therapeutic targets for coxsackievirus B-induced type 1 diabetes.
Collapse
Affiliation(s)
- Salima Lalani
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Joseph Knudsen
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - James Kenney
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, United States
| | - Didier Hober
- Laboratoire de Virologie ULR3610, Univ Lille, Centre Hospitalier Universitaire de Lille, Lille, France
| | - C. Michael DiPersio
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, United States
- Department of Surgery, Albany Medical College, Albany, NY, United States
| | - Allen Gerber
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, United States
- Department of Neurology, Castle Point Medical Center, Wappingers Falls, NY, United States
| |
Collapse
|
4
|
Cabiati M, Federico G, Del Ry S. Importance of Studying Non-Coding RNA in Children and Adolescents with Type 1 Diabetes. Biomedicines 2024; 12:1988. [PMID: 39335501 PMCID: PMC11429055 DOI: 10.3390/biomedicines12091988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Type 1 diabetes (T1D) mellitus is a chronic illness in children and teens, with rising global incidence rates. It stems from an autoimmune attack on pancreatic β cells, leading to insufficient insulin production. Genetic susceptibility and environmental triggers initiate this process. Early detection is possible by identifying multiple autoantibodies, which aids in predicting future T1D development. A new staging system highlights T1D's onset with islet autoimmunity rather than symptoms. Family members of T1D patients face a significantly increased risk of T1D. Italy recently passed a law mandating national T1D screening for pediatric populations. Measurements of β cell function continue to be essential in assessing efficacy, and different models have been proposed, but more appropriate biomarkers are mandatory for both progression studies before the onset of diabetes and during therapeutic monitoring. Biomarkers like microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) play key roles in T1D pathogenesis by regulating gene expression. Understanding their roles offers insights into T1D mechanisms and potential therapeutic targets. In this review, we summarized recent progress in the roles of some non-coding RNAs (ncRNAs) in the pathogenesis of T1D, with particular attention to miRNAs, lncRNAs, and circRNAs.
Collapse
Affiliation(s)
- Manuela Cabiati
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Giovanni Federico
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Del Ry
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| |
Collapse
|
5
|
Joglekar MV, Kaur S, Pociot F, Hardikar AA. Prediction of progression to type 1 diabetes with dynamic biomarkers and risk scores. Lancet Diabetes Endocrinol 2024; 12:483-492. [PMID: 38797187 DOI: 10.1016/s2213-8587(24)00103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 05/29/2024]
Abstract
Identifying biomarkers of functional β-cell loss is an important step in the risk stratification of type 1 diabetes. Genetic risk scores (GRS), generated by profiling an array of single nucleotide polymorphisms, are a widely used type 1 diabetes risk-prediction tool. Type 1 diabetes screening studies have relied on a combination of biochemical (autoantibody) and GRS screening methodologies for identifying individuals at high-risk of type 1 diabetes. A limitation of these screening tools is that the presence of autoantibodies marks the initiation of β-cell loss, and is therefore not the best biomarker of progression to early-stage type 1 diabetes. GRS, on the other hand, represents a static biomarker offering a single risk score over an individual's lifetime. In this Personal View, we explore the challenges and opportunities of static and dynamic biomarkers in the prediction of progression to type 1 diabetes. We discuss future directions wherein newer dynamic risk scores could be used to predict type 1 diabetes risk, assess the efficacy of new and emerging drugs to retard, or prevent type 1 diabetes, and possibly replace or further enhance the predictive ability offered by static biomarkers, such as GRS.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | | | - Flemming Pociot
- Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
6
|
Sebastiani G, Grieco GE, Bruttini M, Auddino S, Mori A, Toniolli M, Fignani D, Licata G, Aiello E, Nigi L, Formichi C, Fernandez-Tajes J, Pugliese A, Evans-Molina C, Overbergh L, Tree T, Peakman M, Mathieu C, Dotta F. A set of circulating microRNAs belonging to the 14q32 chromosome locus identifies two subgroups of individuals with recent-onset type 1 diabetes. Cell Rep Med 2024; 5:101591. [PMID: 38838677 PMCID: PMC11228666 DOI: 10.1016/j.xcrm.2024.101591] [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: 10/19/2023] [Revised: 02/02/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Circulating microRNAs (miRNAs) are linked to the onset and progression of type 1 diabetes mellitus (T1DM), thus representing potential disease biomarkers. In this study, we employed a multiplatform sequencing approach to analyze circulating miRNAs in an extended cohort of prospectively evaluated recent-onset T1DM individuals from the INNODIA consortium. Our findings reveal that a set of miRNAs located within T1DM susceptibility chromosomal locus 14q32 distinguishes two subgroups of individuals. To validate our results, we conducted additional analyses on a second cohort of T1DM individuals, confirming the identification of these subgroups, which we have named cluster A and cluster B. Remarkably, cluster B T1DM individuals, who exhibit increased expression of a set of 14q32 miRNAs, show better glycemic control and display a different blood immunomics profile. Our findings suggest that this set of circulating miRNAs can identify two different T1DM subgroups with distinct blood immunomics at baseline and clinical outcomes during follow-up.
Collapse
Affiliation(s)
- Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Giuseppina Emanuela Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Marco Bruttini
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| | - Stefano Auddino
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Alessia Mori
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| | - Mattia Toniolli
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Daniela Fignani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Giada Licata
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Elena Aiello
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | - Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy
| | | | - Alberto Pugliese
- Diabetes Research Institute, Leonard Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases and the Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lut Overbergh
- Katholieke Universiteit Leuven/Universitaire Ziekenhuizen, Leuven, Belgium
| | - Timothy Tree
- Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Mark Peakman
- Immunology & Inflammation Research Therapeutic Area, Sanofi, Boston, MA, USA
| | - Chantal Mathieu
- Katholieke Universiteit Leuven/Universitaire Ziekenhuizen, Leuven, Belgium
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario ONLUS c/o Toscana Life Science, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy.
| |
Collapse
|
7
|
Beltramo E, Mazzeo A, Porta M. Release of Pro-Inflammatory/Angiogenic Factors by Retinal Microvascular Cells Is Mediated by Extracellular Vesicles Derived from M1-Activated Microglia. Int J Mol Sci 2023; 25:15. [PMID: 38203187 PMCID: PMC10778795 DOI: 10.3390/ijms25010015] [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: 10/25/2023] [Revised: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
The interactions between the neuronal and vascular sides of the retina during diabetic retinopathy (DR) have gained increasing attention. Microglia is responsible for the immune response to inflammation inside the retina, which could be mediated by paracrine signals carried by extracellular vesicles (EVs). We aimed to characterize EVs released from immortalized human microglial cells in inflammation and investigate their effects on the retinal microvasculature and the anti-inflammatory potential of thiamine in this context. M1 pro-inflammatory polarization in microglia was induced through a cytokine cocktail. EVs were isolated from the supernatants, characterized, and used to stimulate human retinal endothelial cells (HRECs) and pericytes (HRPs). Microvascular cell functions and their release of pro-inflammatory/angiogenic factors were assessed. M1-derived EVs showed increased content of miR-21, miR-155, CCL2, MMP2, and MMP9, and enhanced apoptosis, proliferation, migration, and ROS production in HRPs and HRECs. IL-1β, IL-6, MMP9, CCL2, and VEGF release increased in HRPs exposed to M1-derived EVs, while HRECs showed augmented IL-6, Ang2, VEGF, and PDFG-B. Addition of thiamine to M1-microglial cultures reverted most of these effects. In conclusion, M1-derived EVs stimulate functional changes and secretion of pro-inflammatory/angiogenic molecules in microvascular cells, exacerbating inflammatory damage and retinopathy features. Thiamine added to microglia exerts anti-inflammatory effects.
Collapse
Affiliation(s)
- Elena Beltramo
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (A.M.); (M.P.)
| | | | | |
Collapse
|
8
|
Shi M, Lu Q, Zhao Y, Ding Z, Yu S, Li J, Ji M, Fan H, Hou S. miR-223: a key regulator of pulmonary inflammation. Front Med (Lausanne) 2023; 10:1187557. [PMID: 37465640 PMCID: PMC10350674 DOI: 10.3389/fmed.2023.1187557] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Small noncoding RNAs, known as microRNAs (miRNAs), are vital for the regulation of diverse biological processes. miR-223, an evolutionarily conserved anti-inflammatory miRNA expressed in cells of the myeloid lineage, has been implicated in the regulation of monocyte-macrophage differentiation, proinflammatory responses, and the recruitment of neutrophils. The biological functions of this gene are regulated by its expression levels in cells or tissues. In this review, we first outline the regulatory role of miR-223 in granulocytes, macrophages, endothelial cells, epithelial cells and dendritic cells (DCs). Then, we summarize the possible role of miR-223 in chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), coronavirus disease 2019 (COVID-19) and other pulmonary inflammatory diseases to better understand the molecular regulatory networks in pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Mingyu Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Ziling Ding
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sifan Yu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Junfeng Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Mengjun Ji
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| |
Collapse
|
9
|
Singh S, Saini H, Sharma A, Gupta S, Huddar VG, Tripathi R. Breast cancer: miRNAs monitoring chemoresistance and systemic therapy. Front Oncol 2023; 13:1155254. [PMID: 37397377 PMCID: PMC10312137 DOI: 10.3389/fonc.2023.1155254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
With a high mortality rate that accounts for millions of cancer-related deaths each year, breast cancer is the second most common malignancy in women. Chemotherapy has significant potential in the prevention and spreading of breast cancer; however, drug resistance often hinders therapy in breast cancer patients. The identification and the use of novel molecular biomarkers, which can predict response to chemotherapy, might lead to tailoring breast cancer treatment. In this context, accumulating research has reported microRNAs (miRNAs) as potential biomarkers for early cancer detection, and are conducive to designing a more specific treatment plan by helping analyze drug resistance and sensitivity in breast cancer treatment. In this review, miRNAs are discussed in two alternative ways-as tumor suppressors to be used in miRNA replacement therapy to reduce oncogenesis and as oncomirs to lessen the translation of the target miRNA. Different miRNAs like miR-638, miR-17, miR-20b, miR-342, miR-484, miR-21, miR-24, miR-27, miR-23 and miR-200 are involved in the regulation of chemoresistance through diverse genetic targets. For instance, tumor-suppressing miRNAs like miR-342, miR-16, miR-214, and miR-128 and tumor-promoting miRNAs like miR101 and miR-106-25 cluster regulate the cell cycle, apoptosis, epithelial to mesenchymal transition and other pathways to impart breast cancer drug resistance. Hence, in this review, we have discussed the significance of miRNA biomarkers that could assist in providing novel therapeutic targets to overcome potential chemotherapy resistance to systemic therapy and further facilitate the design of tailored therapy for enhanced efficacy against breast cancer.
Collapse
Affiliation(s)
- Shivam Singh
- Department of Radiation Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Heena Saini
- Integrated translational Molecular Biology laboratory, Department of Rog Nidan and Vikriti vigyan (Pathology), All India Institute of Ayurveda (AIIA), New Delhi, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhash Gupta
- Department of Radiation Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - V. G. Huddar
- Department of Kaya Chikitsa (Internal Medicine), All India Institute of Ayurveda (AIIA), New Delhi, India
| | - Richa Tripathi
- Integrated translational Molecular Biology laboratory, Department of Rog Nidan and Vikriti vigyan (Pathology), All India Institute of Ayurveda (AIIA), New Delhi, India
| |
Collapse
|
10
|
Morales-Sánchez P, Lambert C, Ares-Blanco J, Suárez-Gutiérrez L, Villa-Fernández E, Garcia AV, García-Villarino M, Tejedor JR, Fraga MF, Torre EM, Pujante P, Delgado E. Circulating miRNA expression in long-standing type 1 diabetes mellitus. Sci Rep 2023; 13:8611. [PMID: 37244952 DOI: 10.1038/s41598-023-35836-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Type 1 diabetes is a chronic autoimmune disease which results in inefficient regulation of glucose homeostasis and can lead to different vascular comorbidities through life. In this study we aimed to analyse the circulating miRNA expression profile of patients with type 1 diabetes, and with no other associated pathology. For this, fasting plasma was obtained from 85 subjects. Next generation sequencing analysis was firstly performed to identify miRNAs that were differentially expressed between groups (20 patients vs. 10 controls). hsa-miR-1-3p, hsa-miR-200b-3p, hsa-miR-9-5p, and hsa-miR-1200 expression was also measured by Taqman RT-PCR to validate the observed changes (34 patients vs. 21 controls). Finally, through a bioinformatic approach, the main pathways affected by the target genes of these miRNAs were studied. Among the studied miRNAs, hsa-miR-1-3p expression was found significantly increased in patients with type 1 diabetes compared to controls, and positively correlated with glycated haemoglobin levels. Additionally, by using a bioinformatic approach, we could observe that changes in hsa-miR-1-3p directly affect genes involved in vascular development and cardiovascular pathologies. Our results suggest that, circulating hsa-miR-1-3p in plasma, together with glycaemic control, could be used as prognostic biomarkers in type 1 diabetes, helping to prevent the development of vascular complications in these patients.
Collapse
Affiliation(s)
- Paula Morales-Sánchez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Lambert
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- University of Barcelona, Barcelona, Spain.
| | - Jessica Ares-Blanco
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Lorena Suárez-Gutiérrez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Elsa Villa-Fernández
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Ana Victoria Garcia
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Miguel García-Villarino
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Juan Ramón Tejedor
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Mario F Fraga
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Edelmiro Menéndez Torre
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Pedro Pujante
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
| | - Elías Delgado
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain.
| |
Collapse
|
11
|
Brenu EW, Harris M, Hamilton-Williams EE. Circulating biomarkers during progression to type 1 diabetes: A systematic review. Front Endocrinol (Lausanne) 2023; 14:1117076. [PMID: 36817583 PMCID: PMC9935596 DOI: 10.3389/fendo.2023.1117076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
AIM Progression to type 1 diabetes (T1D) is defined in stages and clinical disease is preceded by a period of silent autoimmunity. Improved prediction of the risk and rate of progression to T1D is needed to reduce the prevalence of diabetic ketoacidosis at presentation as well as for staging participants for clinical trials. This systematic review evaluates novel circulating biomarkers associated with future progression to T1D. METHODS PubMed, Ovid, and EBSCO databases were used to identify a comprehensive list of articles. The eligibility criteria included observational studies that evaluated the usefulness of circulating markers in predicting T1D progression in at-risk subjects <20 years old. RESULTS Twenty-six studies were identified, seventeen were cohort studies and ten were case control studies. From the 26 studies, 5 found evidence for protein and lipid dysregulation, 11 identified molecular markers while 12 reported on changes in immune parameters during progression to T1D. An increased risk of T1D progression was associated with the presence of altered gene expression, immune markers including regulatory T cell dysfunction and higher short-lived effector CD8+ T cells in progressors. DISCUSSION Several circulating biomarkers are dysregulated before T1D diagnosis and may be useful in predicting either the risk or rate of progression to T1D. Further studies are required to validate these biomarkers and assess their predictive accuracy before translation into broader use. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero, identifier (CRD42020166830).
Collapse
Affiliation(s)
- Ekua W. Brenu
- School of Medicine, University of Notre Dame, Sydney, NSW, Australia
| | - Mark Harris
- Endocrinology Department, Queensland Children’s Hospital, South Brisbane, QLD, Australia
| | - Emma E. Hamilton-Williams
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- *Correspondence: Emma E. Hamilton-Williams,
| |
Collapse
|
12
|
Li S, Wu Q, Jiang Z, Wu Y, Li Y, Ni B, Xiao J, Zhai Z. miR-31-5p Regulates Type I Interferon by Targeting SLC15A4 in Plasmacytoid Dendritic Cells of Systemic Lupus Erythematosus. J Inflamm Res 2022; 15:6607-6616. [PMID: 36510495 PMCID: PMC9739073 DOI: 10.2147/jir.s383623] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/12/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Plasmacytoid dendritic cells (pDCs) are the main producers of type I interferon (IFN-I), and the excessive production of IFN-I is a hallmark of systemic lupus erythematosus (SLE). Both SLC15A4 and miR-31-5p are SLE susceptibility-related genes, and SLC15A4 has been implicated an important role in endolysosomal toll-like receptor (TLR) activation in pDCs. However, whether miR-31-5p exerts a regulating effect on SLC15A4 expression in pDCs is unclear. METHODS The expression of SLC15A4 and miR-31-5p in peripheral blood mononuclear cells (PBMCs) of SLE patients was measured by RT-qPCR analyses. The quantitative analysis of IFN-α secretion in the patients' serum was performed by ELISA assay. Luciferase-reporter assay was applied to confirm the interaction between miR-31-5p and SLC15A4. The expression of miR-31-5p, SLC15A4 and IFN-stimulated genes (ISGs, such as MX1, OAS1 and IFIT3) was detected by Western blot and RT-qPCR assays and further IRF5 phosphorylation was evaluated by immunofluorescence after transfected with miR-31-5p mimics or inhibitor in THP-1 and CAL-1 cells. RESULTS The expression of miR-31-5p was downregulated and negatively correlated with the overexpression of SLC15A4 in PBMCs of SLE patients. In addition to this, the secretion of IFN-α was overexpressed in sera of SLE and positively correlated with SLC15A4 level. We found that miR-31-5p directly targeted SLC15A4 and negatively regulated the expression of SLC15A4 in THP-1 and CAL-1 cells. In vitro inhibition of miR-31-5p increased the phosphorylation of IRF5 and the induction of ISGs stimulated by R848, overexpression of miR-31-5p get the reverse results. CONCLUSION miR-31-5p might involve in SLE pathogenesis through regulating IFN-I expression by negatively regulating SLC15A4 to increase the levels of IFN-α and ISGs in pDCs.
Collapse
Affiliation(s)
- Shifei Li
- Department of Dermatology, Southwest Hospital of Third Military Medical University, Chongqing, People’s Republic of China
| | - Qijun Wu
- Department of Dermatology, Southwest Hospital of Third Military Medical University, Chongqing, People’s Republic of China
| | - Zhuyan Jiang
- Department of Dermatology, Southwest Hospital of Third Military Medical University, Chongqing, People’s Republic of China
| | - Yaguang Wu
- Department of Dermatology, Southwest Hospital of Third Military Medical University, Chongqing, People’s Republic of China
| | - Yuhong Li
- Department of Cell Biology, Third Military Medical University, Chongqing, People’s Republic of China
| | - Bing Ni
- Department of Pathophysiology, Third Military Medical University, Chongqing, People’s Republic of China
| | - Jun Xiao
- Department of Cardiovascular Medicine, Chongqing University Central Hospital, Chongqing, People’s Republic of China
| | - Zhifang Zhai
- Department of Dermatology, Southwest Hospital of Third Military Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
13
|
Shi H, Bi X, Zhang J, Duan S, Yan J, Jia H. Simple and sensitive detection of microRNA based on guanine-rich DNA-enhanced fluorescence of DNA-templated silver clusters. Talanta 2022. [DOI: 10.1016/j.talanta.2022.124065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
14
|
Xiao Z, He F, Feng M, Liu Z, Liu Z, Li S, Wang W, Yao H, Wu J. Engineered coxsackievirus B3 containing multiple organ-specific miRNA targets showed attenuated viral tropism and protective immunity. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 103:105316. [PMID: 35718333 DOI: 10.1016/j.meegid.2022.105316] [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: 11/30/2021] [Revised: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Coxsackievirus B3 (CVB3) can cause viral myocarditis, pancreatitis, and aseptic meningitis. This study aimed to construct an engineered CVB3 harboring three different tissue-specific miRNA targets (CVB3-miR3*T) to decrease the virulence of CVB3 in muscles, pancreas, and brain. CVB3-miR3*T and CVB3-miR-CON (containing three sequences not found in the human genome) were engineered and replicated in HELA cells. A viral plaque assay was used to determine the titers in HELA cells and TE671 cells (high miRNA-206 expression), MIN-6 cells (high miRNA-29a-3p expression), and mouse astrocytes (high miRNA-124-3p expression). We found that engineered CVB3 showed attenuated replication and reduced cytotoxicity, the variability of each type of cell was also increased in the CVB3-miR3*T group. Male BALB/c mice were infected to determine the LD50 and examine heart, pancreas, and brain titers and injury. Viral replication of the engineered viruses was restricted in infected mouse heart, pancreas, and brain, and viral plaques were about 100 fold lower compared with the control group. Mice immunized using CVB3-miR3*T, UV-inactivated CVB3-WT, and CVB3-miR-CON were infected with 100 × LD50 of CVB3-WT to determine neutralization. CVB3-miRT*3-preimmunized mice exhibited complete protection and remained alive after lethal virus infection, while only 5/15 were alive in the UV-inactivated mice, and all 15 mice were dead in the PBS-immunized group. The results demonstrate that miR-206-, miRNA-29a-3p-, and miRNA-124-3p-mediated CVB3 detargeting from the pancreas, heart, and brain might be a highly effective strategy for viral vaccine development.
Collapse
Affiliation(s)
- Zonghui Xiao
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Feng He
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Miao Feng
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Zhuo Liu
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Zhewei Liu
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Sen Li
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Wei Wang
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Hailan Yao
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China.
| | - Jianxin Wu
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China; Beijing Municipal Key Laboratory of Child Development and Nutriomics, Beijing, China; Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Wang R, Wei A, Zhang Y, Xu G, Nong X, Liu C, Zeng Y, Huang H, Pang X, Wei W, Wang C, Huang H. Association between genetic variants of microRNA-21 and microRNA-155 and systemic lupus erythematosus: A case-control study from a Chinese population. J Clin Lab Anal 2022; 36:e24518. [PMID: 35707883 PMCID: PMC9279951 DOI: 10.1002/jcla.24518] [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: 02/04/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a common autoimmune disease, and its pathogenesis remains unclear. The alteration of genetic materials is believed to play a role in SLE development. This study evaluated the association between the genetic variants of microRNA‐21 (miR‐21) and microRNA‐155 (miR‐155) and SLE. Methods The SNaPshot genotyping method was used to detect the genotypes of selected SNPs in patients and controls. The expression of miR‐21 and miR‐155 was analyzed using reverse transcription‐quantitative polymerase chain reaction (RT‐qPCR). The functional annotation and the biological effects of SNPs were assessed by HaploReg V4.1 and Regulome DB V2.0 software. The Hardy–Weinberg equilibrium test was used to gather statistics, and odds ratios (ORs) and 95% confidence intervals (CIs) were evaluated by logistic regression. Results The distribution difference of TA genotype in rs767649 was observed (TA vs. T/T: OR = 0.68, 95%CI, 0.48–0.95, p = 0.026). There was a significant difference in the T/A + A/A (T/A + A/A vs. T/T: OR = 0.68, 95%CI, 0.49–0.94, p = 0.020). A significant difference in T allele distribution was found in the depressed complement of SLE (T vs. A: OR = 0.67, 95%CI, 0.47–0.95, p = 0.026). There were significant differences in genetic variants of rs13137 between the positive and the negative SSB antibodies (Anti‐SSB) (T vs. A: OR = 0.67, 95%CI, 0.47–0.95, p = 0.026; T/A + T/T vs. AA: OR = 2.23, 1.18–4.49, p = 0.013). The expression levels of miR‐21 and miR‐155 were significantly higher in patients than in controls (p < 0.001). Conclusions This study provides novel insight that genetic variants of rs767649 and rs13137 are associated with susceptibility to SLE.
Collapse
Affiliation(s)
- Rong Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Anji Wei
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yingjie Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Guidan Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xuejuan Nong
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Chunhong Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yonglong Zeng
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Huatuo Huang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xiaoxia Pang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Wujun Wei
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Chunfang Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Huayi Huang
- Department of Laboratory Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.,Mindray North America, 800 MacArthur Boulevard, Mahwah, New Jersey, USA.,Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carton Streets, Buffalo, New York, USA
| |
Collapse
|
16
|
Liu J, Zhang Y, Tian Y, Huang W, Tong N, Fu X. Integrative biology of extracellular vesicles in diabetes mellitus and diabetic complications. Theranostics 2022; 12:1342-1372. [PMID: 35154494 PMCID: PMC8771544 DOI: 10.7150/thno.65778] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/11/2021] [Indexed: 11/14/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic systemic disease with increasing prevalence globally. An important aspect of diabetic pathogenesis is cellular crosstalk and information exchange between multiple metabolic organs and tissues. In the past decade, increasing evidence suggested that extracellular vesicles (EVs), a class of cell-derived membrane vesicles that transmit information and perform inter-cellular and inter-organ communication, are involved in the pathological changes of insulin resistance (IR), inflammation, and endothelial injury, and implicated in the development of DM and its complications. The biogenesis and cargo sorting machinery dysregulation of EVs may mediate their pathogenic roles under diabetic conditions. Moreover, the biogenesis of EVs, their ubiquitous production by different cells, their function as mediators of inter-organ communication, and their biological features in body fluids have generated great promise as biomarkers and clinical treatments. In this review, we summarize the components of EV generation and sorting machinery and highlight their role in the pathogenesis of DM and associated complications. Furthermore, we discuss the emerging clinical implications of EVs as potential biomarkers and therapeutic strategies for DM and diabetic complications. A better understanding of EVs will deepen our knowledge of the pathophysiology of DM and its complications and offer attractive approaches to improve the prevention, diagnosis, treatment, and prognosis of these disorders.
Collapse
Affiliation(s)
- Jing Liu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yanyan Zhang
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Department of Geriatric Medicine, Lanzhou University Secondary Hospital, Lanzhou, Gansu, China
| | - Yan Tian
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Wei Huang
- Department and Laboratory of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Nanwei Tong
- Division of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, Laboratory of Diabetes and Islet Transplantation Research, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
17
|
Micro-RNA Implications in Type-1 Diabetes Mellitus: A Review of Literature. Int J Mol Sci 2021; 22:ijms222212165. [PMID: 34830046 PMCID: PMC8621893 DOI: 10.3390/ijms222212165] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/24/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Type-1 diabetes mellitus (T1DM) is one of the most well-defined and complex metabolic disorders, characterized by hyperglycemia, with a constantly increasing incidence in children and adolescents. While current knowledge regarding the molecules related to the pathogenesis and diagnosis of T1DM is vast, the discovery of new molecules, such as micro ribonucleic acids (micro-RNAs, miRNAs), as well as their interactions with T1DM, has spurred novel prospects in the diagnosis of the disease. This review aims at summarizing current knowledge regarding miRNAs' biosynthesis and action pathways and their role as gene expression regulators in T1DM. MiRNAs follow a complex biosynthesis pathway, including cleaving and transport from nucleus to cytoplasm. After assembly of their final form, they inhibit translation or cause messenger RNA (mRNA) degradation, resulting in the obstruction of protein synthesis. Many studies have reported miRNA involvement in T1DM pathogenesis, mainly through interference with pancreatic b-cell function, insulin production and secretion. They are also found to contribute to β-cell destruction, as they aid in the production of autoreactive agents. Due to their elevated accumulation in various biological specimens, as well as their involvement in T1DM pathogenesis, their role as biomarkers in early preclinical T1DM diagnosis is widely hypothesized, with future studies concerning their diagnostic value deemed a necessity.
Collapse
|
18
|
Flowers E, Allen IE, Kanaya AM, Aouizerat BE. Circulating microRNAs are associated with variability in fasting blood glucose over 12-months and target pathways related to type 2 diabetes: A pilot study. Diab Vasc Dis Res 2021; 18:14791641211055837. [PMID: 34846185 PMCID: PMC8761879 DOI: 10.1177/14791641211055837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION MicroRNAs (miRs) may be important regulators of risk for type 2 diabetes (T2D). Circulating miRs may provide information about which individuals are at risk for T2D. The purpose of this study was to assess longitudinal associations between circulating miR expression and variability in fasting blood glucose (FBG) and to identify miR-targeted genes and biological pathways. METHODS Variability in FBG was estimated using standard deviation from participants (n = 20) in a previously completed yoga trial. Expression of 402 miRs was measured using hydrogel particle lithography. MirTarBase was used to identify mRNAs, and miRPathDB was used to identify pathways targeted by differentially expressed miRs. RESULTS Six circulating miRs (miR-192, miR-197, miR-206, miR-424, miR-486, and miR-93) were associated with variability in FBG and targeted 143 genes and 23 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Six mRNAs (AKT1, CCND1, ESR1, FASN, SMAD7, and VEGFA) were targeted by at least two miRs and four of those were located in miR-targeted KEGG pathways. CONCLUSIONS Circulating miRs are associated with variability in FBG in individuals at risk for T2D. Further studies are needed to determine whether miRs may be prodromal biomarkers that can identify which individuals are at greatest risk to progress to T2D and which biological pathways underlie this risk.
Collapse
Affiliation(s)
- Elena Flowers
- Department of Physiological Nursing, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Elena Flowers, San Francisco Department of Physiological Nursing, University of California, 2 Koret Way, #605L, San Francisco, CA 94143-0610, USA.
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Alka M Kanaya
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Bradley E Aouizerat
- Department of Oral and Maxillofacial Surgery, New York University, New York, NY, USA
- Bluestone Center for Clinical Research, New York University, New York, NY, USA
| |
Collapse
|
19
|
Ma GM, Huo LW, Tong YX, Wang YC, Li CP, Jia HX. Label-free and sensitive MiRNA detection based on turn-on fluorescence of DNA-templated silver nanoclusters coupled with duplex-specific nuclease-assisted signal amplification. Mikrochim Acta 2021; 188:355. [PMID: 34585278 DOI: 10.1007/s00604-021-05001-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022]
Abstract
A novel strategy for microRNAs (miRNAs) detection has been developed utilizing duplex-specific nuclease-assisted signal amplification (DSNSA) and guanine-rich DNA-enhanced fluorescence of DNA-templated silver nanoclusters (AgNCs). The combination between target miRNA, DSNSA, and AgNCs is achieved by the unique design of DNA sequences. Target miRNA opens the hairpin structure of the Hairpin DNA probe (HP) by hybridizing with the HP and initiates the duplex-specific nuclease-assisted signal amplification (DSNSA) reaction. The DSNSA reaction generates the release of the guanine-rich DNA sequence, which can turn on the fluorescence of the dark AgNCs by hybridizing with the DNA template of the dark AgNCs. The fluorescence intensity of AgNCs corresponds to the dosage of the target miRNA. This is measured at 630 nm by exciting at 560 nm. The constructed method exhibits a low detection limit (~8.3 fmol), a great dynamic range of more than three orders of magnitude, and excellent selectivity. Moreover, it has a good performance for miR-21 detection in complex biological samples. A novel strategy for microRNAs (miRNAs) detection has been developed utilizing duplex-specific nuclease-assisted signal amplification (DSNSA) and guanine-rich DNA-enhanced fluorescence of DNA-templated silver nanoclusters (AgNCs).
Collapse
Affiliation(s)
- Gui-Min Ma
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education; Key Laboratory of Analytical Science and Technology of Hebei Province; Institute of Life Science and Green Development; College of Chemistry and Environmental Science, Hebei University, Baoding, 071002, Hebei Province, People's Republic of China
| | - Li-Wei Huo
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education; Key Laboratory of Analytical Science and Technology of Hebei Province; Institute of Life Science and Green Development; College of Chemistry and Environmental Science, Hebei University, Baoding, 071002, Hebei Province, People's Republic of China
| | - Yin-Xia Tong
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education; Key Laboratory of Analytical Science and Technology of Hebei Province; Institute of Life Science and Green Development; College of Chemistry and Environmental Science, Hebei University, Baoding, 071002, Hebei Province, People's Republic of China
| | - Yu-Cong Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education; Key Laboratory of Analytical Science and Technology of Hebei Province; Institute of Life Science and Green Development; College of Chemistry and Environmental Science, Hebei University, Baoding, 071002, Hebei Province, People's Republic of China
| | - Cui-Ping Li
- Key Laboratory of Public Health Safety of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education; College of Public Health, Hebei University, Baoding, 071002, People's Republic of China
| | - Hong-Xia Jia
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education; Key Laboratory of Analytical Science and Technology of Hebei Province; Institute of Life Science and Green Development; College of Chemistry and Environmental Science, Hebei University, Baoding, 071002, Hebei Province, People's Republic of China.
| |
Collapse
|
20
|
Jiang Y, Gai Y, Long Y, Liu Q, Liu C, Zhang Y, Lan X. Application and Evaluation of [ 99mTc]-Labeled Peptide Nucleic Acid Targeting MicroRNA-155 in Breast Cancer Imaging. Mol Imaging 2021; 19:1536012120916124. [PMID: 32559121 PMCID: PMC7307583 DOI: 10.1177/1536012120916124] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been reported that dysregulation of microRNA-155 expression and function is associated with tumorigenesis, growth, tumor subtypes, invasion, and poor survival rates. Peptide nucleic acid (PNA), an artificially synthesized nucleic acid mimic, has been applied for molecular diagnosis. In this study, a PNA sequence that undergoes complementary binding to miR-155 was labeled with 99mTc to evaluate whether the tracer could visualize the expression of miR-155 in breast cancer. Both antisense PNA (anti-PNA, fully complementary bound to human mature miR-155, referred to as “anti-PNA-155”) and mismatched PNA (referred to as “mis-PNA”) single strands containing 23-mer were synthesized. The relative expression of miR-155 in MCF-7 cells and tumors was higher than that in MDA-MB-231 cells and tumors. Single-photon emission computed tomography (SPECT) scan showed that radioactivity mainly accumulated in kidney. MCF-7 tumors, but not MDA-MB-231 tumors, were clearly visualized after [99mTc]anti-PNA-155 injection. MCF-7 tumors were less visible when coinjected with 100-fold excess of anti-PNA-155 or injected with [99mTc]mis-PNA, which suggested specific binding. Biodistribution study results were consistent with SPECT imaging. We successfully demonstrated that [99mTc]anti-PNA-155 could visualize miR-155 expression in vivo, suggesting it may be a promising probe applied in breast cancer.
Collapse
Affiliation(s)
- Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chunbao Liu
- Department of Nuclear Medicine, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
21
|
Margaritis K, Margioula-Siarkou G, Margioula-Siarkou C, Petousis S, Kotanidou EP, Christoforidis A, Pavlou E, Galli-Tsinopoulou A. Circulating serum and plasma levels of micro-RNA in type-1 diabetes in children and adolescents: A systematic review and meta-analysis. Eur J Clin Invest 2021; 51:e13510. [PMID: 33565089 DOI: 10.1111/eci.13510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a complex metabolic disorder characterized by hyperglycaemia, with constantly increasing incidence in paediatric population. The discovery of new molecules, such as microRNAs, and their possible interactions with T1DM create novel aspects in the diagnosis of the disease. METHODS This systematic review and meta-analysis adhered to PRISMA guidelines. MEDLINE, SCOPUS, Cochrane CENTRAL and Clinicaltrials.gov. were searched up to 20 April 2020. Inclusion criteria for individual studies were quantification of microRNAs in serum/plasma samples and study groups consisting of children and adolescents with T1DM and healthy controls. Primary outcome of the study was the qualitative expression of microRNAs between the two groups. Statistical analysis was performed with Comprehensive Meta-Analysis Software v3.0. Methodological quality of included studies was assessed using Newcastle-Ottawa scale. RESULTS A total of 484 studies were retrieved from the initial search of the databases. These were subsequently limited to seven included studies. Seven microRNAs demonstrated contrasting expression between the two groups, with two of them showing significant overexpression in T1DM group (miR-181:95% CI: 0.429 to 1.341 P < .001, miR-210:95% CI: 0.381 to 0.852, P < .001) and one micro-RNA being significantly overexpressed in control group (miR-375:95% CI: 0.293 to 1.459, P = .003). CONCLUSION A total of three micro-RNA molecules appeared to have a significantly different expression in T1DM patients, serving as a possible diagnostic panel of biomarkers. These findings may contribute as reference for future research to further support the use of microRNAs as a novel diagnostic tool in T1DM.
Collapse
Affiliation(s)
- Kosmas Margaritis
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| | - Georgia Margioula-Siarkou
- 2nd Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Chrysoula Margioula-Siarkou
- 2nd Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Stamatios Petousis
- 2nd Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Eleni P Kotanidou
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| | - Athanasios Christoforidis
- 1st Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| | - Evangelos Pavlou
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| | - Assimina Galli-Tsinopoulou
- 2nd Department of Paediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA University General Hospital, Thessaloniki, Greece
| |
Collapse
|
22
|
Tamara A, Coulson DJ, Latief JS, Bakhashab S, Weaver JU. Upregulated anti-angiogenic miR-424-5p in type 1 diabetes (model of subclinical cardiovascular disease) correlates with endothelial progenitor cells, CXCR1/2 and other parameters of vascular health. Stem Cell Res Ther 2021; 12:249. [PMID: 33985567 PMCID: PMC8120744 DOI: 10.1186/s13287-021-02332-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 01/14/2023] Open
Abstract
Background In spite of clinical progress, cardiovascular disease (CVD) remains the predominant cause of mortality worldwide. Overexpression studies in animals have proven miR-424-5p to have anti-angiogenic properties. As type 1 diabetes mellitus (T1DM) without CVD displays endothelial dysfunction and reduced circulating endothelial progenitor cells (cEPCs), it offers a model of subclinical CVD. Therefore, we explored miR-424-5p, cytokines and vascular health in T1DM. Methods Twenty-nine well-controlled T1DM patients with no CVD and 20-matched controls were studied. Cytokines IL8, TNF-α, IL7, VEGF-C, cEPCs/CD45dimCD34+CD133+ cells and ex-vivo proangiogenic cells (PACs)/fibronectin adhesion assay (FAA) were measured. MiR-424-5p in plasma and peripheral blood mononuclear cells (PBMC) along with mRNAs in PBMC was evaluated. Results We found an elevation of IL7 (p = 0.008), IL8 (p = 0.003), TNF-α (p = 0.041), VEGF-C (p = 0.013), upregulation of mRNA CXCR1 (p = 0.009), CXCR2 (p < 0.001) and reduction of cEPCs (p < 0.001), PACs (p < 0.001) and FAA (p = 0.017) in T1DM. MiR-424-5p was upregulated in T1DM in PBMC (p < 0.001). MiR-424-5p was negatively correlated with cEPCs (p = 0.006), PACs (p = 0.005) and FAA (p < 0.001) and positively with HbA1c (p < 0.001), IL7 (p = 0.008), IL8 (p = 0.017), VEGF-C (p = 0.007), CXCR1 (p = 0.02) and CXCR2 (p = 0.001). ROC curve analyses showed (1) miR-424-5p to be a biomarker for T1DM (p < 0.001) and (2) significant upregulation of miR-424-5p, defining subclinical CVD, occurred at HbA1c of 46.5 mmol/mol (p = 0.002). Conclusion We validated animal research on anti-angiogenic properties of miR-424-5p in T1DM. MiR-424-5p may be a biomarker for onset of subclinical CVD at HbA1c of 46.5 mmol/mol (pre-diabetes). Thus, miR-424-5p has potential use for CVD monitoring whilst anti-miR-424-5p-based therapies may be used to reduce CVD morbidity/mortality in T1DM.
Collapse
Affiliation(s)
- Alice Tamara
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - David J Coulson
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Jevi Septyani Latief
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - Sherin Bakhashab
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 80218, Saudi Arabia
| | - Jolanta U Weaver
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK. .,Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne, NE9 6SH, UK. .,Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
23
|
Zarnegarnia Y, Messinger S. A conditional approach for the receiver operating characteristic curve construction to evaluate diagnostic test performance in a family-matched case-control design. Stat Methods Med Res 2021; 30:1249-1272. [PMID: 33826452 DOI: 10.1177/0962280221995956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Receiver operating characteristic curves are widely used in medical research to illustrate biomarker performance in binary classification, particularly with respect to disease or health status. Study designs that include related subjects, such as siblings, usually have common environmental or genetic factors giving rise to correlated biomarker data. The design could be used to improve detection of biomarkers informative of increased risk, allowing initiation of treatment to stop or slow disease progression. Available methods for receiver operating characteristic construction do not take advantage of correlation inherent in this design to improve biomarker performance. This paper will briefly review some developed methods for receiver operating characteristic curve estimation in settings with correlated data from case-control designs and will discuss the limitations of current methods for analyzing correlated familial paired data. An alternative approach using conditional receiver operating characteristic curves will be demonstrated. The proposed approach will use information about correlation among biomarker values, producing conditional receiver operating characteristic curves that evaluate the ability of a biomarker to discriminate between affected and unaffected subjects in a familial paired design.
Collapse
Affiliation(s)
- Yalda Zarnegarnia
- Biostatistics & Design Program, Oregon Health and Science University, OR, USA
| | - Shari Messinger
- Department of Public Health Sciences, University of Miami, Miami, FL, USA
| |
Collapse
|
24
|
Snowhite I, Pastori R, Sosenko J, Messinger Cayetano S, Pugliese A. Baseline Assessment of Circulating MicroRNAs Near Diagnosis of Type 1 Diabetes Predicts Future Stimulated Insulin Secretion. Diabetes 2021; 70:638-651. [PMID: 33277338 PMCID: PMC7881864 DOI: 10.2337/db20-0817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes is an autoimmune disease resulting in severely impaired insulin secretion. We investigated whether circulating microRNAs (miRNAs) are associated with residual insulin secretion at diagnosis and predict the severity of its future decline. We studied 53 newly diagnosed subjects enrolled in placebo groups of TrialNet clinical trials. We measured serum levels of 2,083 miRNAs, using RNA sequencing technology, in fasting samples from the baseline visit (<100 days from diagnosis), during which residual insulin secretion was measured with a mixed meal tolerance test (MMTT). Area under the curve (AUC) C-peptide and peak C-peptide were stratified by quartiles of expression of 31 miRNAs. After adjustment for baseline C-peptide, age, BMI, and sex, baseline levels of miR-3187-3p, miR-4302, and the miRNA combination of miR-3187-3p/miR-103a-3p predicted differences in MMTT C-peptide AUC/peak levels at the 12-month visit; the combination miR-3187-3p/miR-4723-5p predicted proportions of subjects above/below the 200 pmol/L clinical trial eligibility threshold at the 12-month visit. Thus, miRNA assessment at baseline identifies associations with C-peptide and stratifies subjects for future severity of C-peptide loss after 1 year. We suggest that miRNAs may be useful in predicting future C-peptide decline for improved subject stratification in clinical trials.
Collapse
Affiliation(s)
- Isaac Snowhite
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Ricardo Pastori
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Jay Sosenko
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Shari Messinger Cayetano
- Department of Public Health Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Alberto Pugliese
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
- Department of Microbiology and Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
25
|
Gubitosi-Klug RA, Braffett BH, Hitt S, Arends V, Uschner D, Jones K, Diminick L, Karger AB, Paterson AD, Roshandel D, Marcovina S, Lachin JM, Steffes M, Palmer JP. Residual β cell function in long-term type 1 diabetes associates with reduced incidence of hypoglycemia. J Clin Invest 2021; 131:143011. [PMID: 33529168 PMCID: PMC7843223 DOI: 10.1172/jci143011] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDWe investigated residual β cell function in Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) study participants with an average 35-year duration of type 1 diabetes mellitus (T1DM).METHODSSerum C-peptide was measured during a 4-hour mixed-meal tolerance test. Associations with metabolic outcomes and complications were explored among nonresponders (all C-peptide values after meal <0.003 nmol/L) and 3 categories of responders, classified by peak C-peptide concentration (nmol/L) as high (>0.2), intermediate (>0.03 to ≤0.2), and low (≥ 0.003 to ≤0.03).RESULTSOf the 944 participants, 117 (12.4%) were classified as responders. Residual C-peptide concentrations were associated with higher DCCT baseline concentrations of stimulated C-peptide (P value for trend = 0.0001). Residual C-peptide secretion was not associated with current or mean HbA1c, HLA high-risk haplotypes for T1DM, or the current presence of T1DM autoantibodies. The proportion of subjects with a history of severe hypoglycemia was lower with high (27%) and intermediate (48%) residual C-peptide concentrations than with low (74%) and no (70%) residual C-peptide concentrations (P value for trend = 0.0001). Responders and nonresponders demonstrated similar rates of advanced microvascular complications.CONCLUSIONβ Cell function can persist in long-duration T1DM. With a peak C-peptide concentration of >0.03 nmol/L, we observed clinically meaningful reductions in the prevalence of severe hypoglycemia.TRIAL REGISTRATIONClinicalTrials.gov NCT00360815 and NCT00360893.FUNDINGDivision of Diabetes Endocrinology and Metabolic Diseases of the National Institute of Diabetes and Digestive and Kidney Diseases (DP3-DK104438, U01 DK094176, and U01 DK094157).
Collapse
Affiliation(s)
- Rose A. Gubitosi-Klug
- Rainbow Babies and Children’s Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | - Barbara H. Braffett
- The Biostatistics Center, George Washington University, Rockville, Maryland, USA
| | - Susan Hitt
- University of Missouri, Columbia, Missouri, USA
| | | | - Diane Uschner
- The Biostatistics Center, George Washington University, Rockville, Maryland, USA
| | | | - Lisa Diminick
- The Biostatistics Center, George Washington University, Rockville, Maryland, USA
| | - Amy B. Karger
- University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew D. Paterson
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Divisions of Epidemiology and Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Delnaz Roshandel
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - John M. Lachin
- The Biostatistics Center, George Washington University, Rockville, Maryland, USA
| | | | | | | |
Collapse
|
26
|
Garavelli S, Bruzzaniti S, Tagliabue E, Di Silvestre D, Prattichizzo F, Mozzillo E, Fattorusso V, La Sala L, Ceriello A, Puca AA, Mauri P, Strollo R, Marigliano M, Maffeis C, Petrelli A, Bosi E, Franzese A, Galgani M, Matarese G, de Candia P. Plasma circulating miR-23~27~24 clusters correlate with the immunometabolic derangement and predict C-peptide loss in children with type 1 diabetes. Diabetologia 2020; 63:2699-2712. [PMID: 32728892 DOI: 10.1007/s00125-020-05237-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS We aimed to analyse the association between plasma circulating microRNAs (miRNAs) and the immunometabolic profile in children with type 1 diabetes and to identify a composite signature of miRNAs/immunometabolic factors able to predict type 1 diabetes progression. METHODS Plasma samples were obtained from children at diagnosis of type 1 diabetes (n = 88) and at 12 (n = 32) and 24 (n = 30) months after disease onset and from healthy control children with similar sex and age distribution (n = 47). We quantified 60 robustly expressed plasma circulating miRNAs by quantitative RT-PCR and nine plasma immunometabolic factors with a recognised role at the interface of metabolic and immune alterations in type 1 diabetes. Based on fasting C-peptide loss over time, children with type 1 diabetes were stratified into the following groups: those who had lost >90% of C-peptide compared with diagnosis level; those who had lost <10% of C-peptide; those showing an intermediate C-peptide loss. To evaluate the modulation of plasma circulating miRNAs during the course of type 1 diabetes, logistic regression models were implemented and the correlation between miRNAs and immunometabolic factors was also assessed. Results were then validated in an independent cohort of children with recent-onset type 1 diabetes (n = 18). The prognostic value of the identified plasma signature was tested by a neural network-based model. RESULTS Plasma circulating miR-23~27~24 clusters (miR-23a-3p, miR-23b-3p, miR-24-3p, miR-27a-3p and miR-27b-3p) were upmodulated upon type 1 diabetes progression, showed positive correlation with osteoprotegerin (OPG) and were negatively correlated with soluble CD40 ligand, resistin, myeloperoxidase and soluble TNF receptor in children with type 1 diabetes but not in healthy children. The combination of plasma circulating miR-23a-3p, miR-23b-3p, miR-24-3p, miR-27b-3p and OPG, quantified at disease onset, showed a significant capability to predict the decline in insulin secretion 12 months after disease diagnosis in two independent cohorts of children with type 1 diabetes. CONCLUSIONS/INTERPRETATIONS We have pinpointed a novel miR-23a-3p/miR-23b-3p/miR-24-3p/miR-27b-3p/OPG plasma signature that may be developed into a novel blood-based method to better stratify patients with type 1 diabetes and predict C-peptide loss.
Collapse
Affiliation(s)
- Silvia Garavelli
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy
| | - Sara Bruzzaniti
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | - Elena Tagliabue
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
| | | | | | - Enza Mozzillo
- Centre of Paediatric Diabetology, Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Valentina Fattorusso
- Centre of Paediatric Diabetology, Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Lucia La Sala
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
| | | | - Annibale A Puca
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Pierluigi Mauri
- Institute of Biomedical Technologies, C. N. R, Segrate, Milan, Italy
| | - Rocky Strollo
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico, Rome, Italy
| | - Marco Marigliano
- Paediatric Diabetes and Metabolic Disorders Unit, University of Verona, Verona, Italy
| | - Claudio Maffeis
- Paediatric Diabetes and Metabolic Disorders Unit, University of Verona, Verona, Italy
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Adriana Franzese
- Centre of Paediatric Diabetology, Department of Translational Medical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Mario Galgani
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II', via Pansini 5, 80131, Naples, Italy.
| | - Giuseppe Matarese
- Institute for Endocrinology and Experimental Oncology 'G. Salvatore', C.N.R, via Pansini 5, 80131, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II', via Pansini 5, 80131, Naples, Italy.
| | - Paola de Candia
- IRCCS MultiMedica, via G. Fantoli 16/15, 20138, Milan, Italy.
| |
Collapse
|
27
|
Wang H. MicroRNA, Diabetes Mellitus and Colorectal Cancer. Biomedicines 2020; 8:biomedicines8120530. [PMID: 33255227 PMCID: PMC7760221 DOI: 10.3390/biomedicines8120530] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus (DM) is an endocrinological disorder that is due to either the pancreas not producing enough insulin, or the body does not respond appropriately to insulin. There are many complications of DM such as retinopathy, nephropathy, and peripheral neuropathy. In addition to these complications, DM was reported to be associated with different cancers. In this review, we discuss the association between DM and colorectal cancer (CRC). CRC is the third most commonly diagnosed cancer worldwide that mostly affects older people, however, its incidence and mortality are rising among young people. We discuss the relationship between DM and CRC based on their common microRNA (miRNA) biomarkers. miRNAs are non-coding RNAs playing important functions in cell differentiation, development, regulation of cell cycle, and apoptosis. miRNAs can inhibit cell proliferation and induce apoptosis in CRC cells. miRNAs also can improve glucose tolerance and insulin sensitivity. Therefore, investigating the common miRNA biomarkers of both DM and CRC can shed a light on how these two diseases are correlated and more understanding of the link between these two diseases can help the prevention of both DM and CRC.
Collapse
Affiliation(s)
- Hsiuying Wang
- Institute of Statistics, National Chiao Tung University, Hsinchu 30010, Taiwan
| |
Collapse
|
28
|
Gebert M, Jaśkiewicz M, Moszyńska A, Collawn JF, Bartoszewski R. The Effects of Single Nucleotide Polymorphisms in Cancer RNAi Therapies. Cancers (Basel) 2020; 12:E3119. [PMID: 33113880 PMCID: PMC7694039 DOI: 10.3390/cancers12113119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Tremendous progress in RNAi delivery methods and design has allowed for the effective development of siRNA-based therapeutics that are currently under clinical investigation for various cancer treatments. This approach has the potential to revolutionize cancer therapy by providing the ability to specifically downregulate or upregulate the mRNA of any protein of interest. This exquisite specificity, unfortunately, also has a downside. Genetic variations in the human population are common because of the presence of single nucleotide polymorphisms (SNPs). SNPs lead to synonymous and non-synonymous changes and they occur once in every 300 base pairs in both coding and non-coding regions in the human genome. Much less common are the somatic mosaicism variations associated with genetically distinct populations of cells within an individual that is derived from postzygotic mutations. These heterogeneities in the population can affect the RNAi's efficacy or more problematically, which can lead to unpredictable and sometimes adverse side effects. From a more positive viewpoint, both SNPs and somatic mosaicisms have also been implicated in human diseases, including cancer, and these specific changes could offer the ability to effectively and, more importantly, selectively target the cancer cells. In this review, we discuss how SNPs in the human population can influence the development and success of novel anticancer RNAi therapies and the importance of why SNPs should be carefully considered.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - Maciej Jaśkiewicz
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - Adrianna Moszyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - James F. Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Rafał Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| |
Collapse
|
29
|
Martins B, Amorim M, Reis F, Ambrósio AF, Fernandes R. Extracellular Vesicles and MicroRNA: Putative Role in Diagnosis and Treatment of Diabetic Retinopathy. Antioxidants (Basel) 2020; 9:E705. [PMID: 32759750 PMCID: PMC7463887 DOI: 10.3390/antiox9080705] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [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: 08/02/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is a complex, progressive, and heterogenous retinal degenerative disease associated with diabetes duration. It is characterized by glial, neural, and microvascular dysfunction, being the blood-retinal barrier (BRB) breakdown a hallmark of the early stages. In advanced stages, there is formation of new blood vessels, which are fragile and prone to leaking. This disease, if left untreated, may result in severe vision loss and eventually legal blindness. Although there are some available treatment options for DR, most of them are targeted to the advanced stages of the disease, have some adverse effects, and many patients do not adequately respond to the treatment, which demands further research. Oxidative stress and low-grade inflammation are closely associated processes that play a critical role in the development of DR. Retinal cells communicate with each other or with another one, using cell junctions, adhesion contacts, and secreted soluble factors that can act in neighboring or long-distance cells. Another mechanism of cell communication is via secreted extracellular vesicles (EVs), through exchange of material. Here, we review the current knowledge on deregulation of cell-to-cell communication through EVs, discussing the changes in miRNA expression profiling in body fluids and their role in the development of DR. Thereafter, current and promising therapeutic agents for preventing the progression of DR will be discussed.
Collapse
Affiliation(s)
- Beatriz Martins
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (B.M.); (M.A.); (F.R.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Madania Amorim
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (B.M.); (M.A.); (F.R.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Flávio Reis
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (B.M.); (M.A.); (F.R.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (B.M.); (M.A.); (F.R.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| | - Rosa Fernandes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (B.M.); (M.A.); (F.R.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Emerging data have suggested that β-cell dysfunction may exacerbate the development and progression of type 1 diabetes (T1D). In this review, we highlight clinical and preclinical studies suggesting a role for β-cell dysfunction during the evolution of T1D and suggest agents that may promote β-cell health in T1D. RECENT FINDINGS Metabolic abnormalities exist years before development of hyperglycemia and exhibit a reproducible pattern reflecting progressive deterioration of β-cell function and increases in β-cell stress and death. Preclinical studies indicate that T1D may be prevented by modification of pathways impacting intrinsic β-cell stress and antigen presentation. Recent findings suggest that differences in metabolic phenotypes and β-cell stress may reflect differing endotypes of T1D. Multiple pathways representing potential drug targets have been identified, but most remain to be tested in human populations with preclinical disease. SUMMARY This cumulative body of work shows clear evidence that β-cell stress, dysfunction, and death are harbingers of impending T1D and likely contribute to progression of disease and insulin deficiency. Treatment with agents targeting β-cell health could augment interventions with immunomodulatory therapies but will need to be tested in intervention studies with endpoints carefully designed to capture changes in β-cell function and health.
Collapse
Affiliation(s)
- Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Roudebush VA Medical Center, Indianapolis, IN
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Regulatory T cells (Tregs) are critical contributors to immune homeostasis and their dysregulation can lead to the loss of immune tolerance and autoimmune diseases like type 1 diabetes (T1D). Recent studies have highlighted microRNAs (miRNAs) as important regulators of the immune system, by fine-tuning relevant genes in various immune cell types. In this review article, we discuss recent insights into miRNA regulation of immune tolerance and activation. Specifically, we discuss how the dysregulation of miRNAs in T cells contributes to their aberrant function and the onset of islet autoimmunity, as well as their potential as targets of novel intervention strategies to interfere with autoimmune activation. RECENT FINDINGS Several studies have shown that the dysregulation of individual miRNAs in T cells can contribute to impaired immune tolerance, contributing to onset and progression of islet autoimmunity. Importantly, the targeting of these miRNAs, including miR-92a, miR-142-3p and miR-181a, resulted in relevant effects on downstream pathways, improved Treg function and reduced islet autoimmunity in murine models. miRNAs are critical regulators of immune homeostasis and the dysregulation of individual miRNAs in T cells contributes to aberrant T cell function and autoimmunity. The specific targeting of individual miRNAs could improve Treg homeostasis and therefore limit overshooting T cell activation and islet autoimmunity.
Collapse
Affiliation(s)
- Martin G. Scherm
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, 80939 Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Ingolstaedter Landstrasse 1, 85764 Munich-, Neuherberg, Germany
| | - Carolin Daniel
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, 80939 Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Ingolstaedter Landstrasse 1, 85764 Munich-, Neuherberg, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| |
Collapse
|
32
|
Jiang ZF, Zhang L, Shen J. MicroRNA: Potential biomarker and target of therapy in acute lung injury. Hum Exp Toxicol 2020; 39:1429-1442. [PMID: 32495695 DOI: 10.1177/0960327120926254] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs stretching over 18-22 nucleotides and considered to be modifiers of many respiratory diseases. They are highly evolutionary conserved and have been implicated in several biological processes, including cell proliferation, apoptosis, differentiation, among others. Acute lung injury (ALI) is a fatal disease commonly caused by direct or indirect injury factors and has a high mortality rate in intensive care unit. Changes in expression of several types of miRNAs have been reported in patients with ALI. Some miRNAs suppress cellular injury and accelerate the recovery of ALI by targeting specific molecules and decreasing excessive immune response. For this reason, miRNAs are proposed as potential biomarkers for ALI and as therapeutic targets for this disease. This review summarizes current evidence supporting the role of miRNAs in ALI.
Collapse
Affiliation(s)
- Z-F Jiang
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - L Zhang
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - J Shen
- Center of Emergency & Intensive Care Unit, Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
33
|
Ventriglia G, Mancarella F, Sebastiani G, Cook DP, Mallone R, Mathieu C, Gysemans C, Dotta F. miR-409-3p is reduced in plasma and islet immune infiltrates of NOD diabetic mice and is differentially expressed in people with type 1 diabetes. Diabetologia 2020; 63:124-136. [PMID: 31659408 DOI: 10.1007/s00125-019-05026-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS MicroRNAs (miRNAs) are a novel class of potential biomarkers emerging in many diseases, including type 1 diabetes. Here, we aim to analyse a panel of circulating miRNAs in non-obese diabetic (NOD) mice and individuals with type 1 diabetes. METHODS We adopted standardised methodologies for extracting miRNAs from small sample volumes to evaluate a profiling panel of mature miRNAs in paired plasma and laser-captured microdissected immune-infiltrated islets of recently diabetic and normoglycaemic NOD mice. Moreover, we validated the findings during disease progression and remission after anti-CD3 therapy in NOD mice, as well as in individuals with type 1 diabetes. RESULTS Plasma levels of five miRNAs were downregulated in diabetic vs normoglycaemic mice. Of those, miR-409-3p was also downregulated in situ in the immune islet infiltrates of diabetic mice, suggesting an association with disease pathogenesis. Target-prediction tools linked miR-409-3p to immune- and metabolism-related signalling molecules. In situ miR-409-3p expression correlated with insulitis severity, and CD8+ central memory T cells were found to be enriched in miR-409-3p. Plasma miR-409-3p levels gradually decreased during diabetes development and improved with disease remission after anti-CD3 antibody therapy. Finally, plasma miR-409-3p levels were lower in people recently diagnosed with type 1 diabetes compared with a non-diabetic control group, and levels were inversely correlated with HbA1c levels. CONCLUSIONS/INTERPRETATION We propose that miR-409-3p may represent a new circulating biomarker of islet inflammation and type 1 diabetes severity.
Collapse
Affiliation(s)
- Giuliana Ventriglia
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16 - 53100, Siena, Italy
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, Siena, Italy
- Clinical and Experimental Endocrinology (CEE), Katholieke Universiteit Leuven (KU LEUVEN), Leuven, Belgium
| | - Francesca Mancarella
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16 - 53100, Siena, Italy
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16 - 53100, Siena, Italy
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, Siena, Italy
| | - Dana P Cook
- Clinical and Experimental Endocrinology (CEE), Katholieke Universiteit Leuven (KU LEUVEN), Leuven, Belgium
| | - Roberto Mallone
- Inserm, U1016, CNRS, UMR8104, Paris Descartes University, Sorbonne Paris Cité, Cochin Institute, Paris, France
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology (CEE), Katholieke Universiteit Leuven (KU LEUVEN), Leuven, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology (CEE), Katholieke Universiteit Leuven (KU LEUVEN), Leuven, Belgium
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, V.le Bracci, 16 - 53100, Siena, Italy.
- Fondazione Umberto Di Mario ONLUS c/o Toscana Life Sciences, Siena, Italy.
| |
Collapse
|
34
|
Scherm MG, Daniel C. miRNA-Mediated Immune Regulation in Islet Autoimmunity and Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:606322. [PMID: 33329406 PMCID: PMC7731293 DOI: 10.3389/fendo.2020.606322] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
The important role of microRNAs as major modulators of various physiological processes, including immune regulation and homeostasis, has been increasingly recognized. Consequently, aberrant miRNA expression contributes to the defective regulation of T cell development, differentiation, and function. This can result in immune activation and impaired tolerance mechanisms, which exert a cardinal function for the onset of islet autoimmunity and the progression to T1D. The specific impact of miRNAs for immune regulation and how miRNAs and their downstream targets are involved in the pathogenesis of islet autoimmunity and T1D has been investigated recently. These studies revealed that increased expression of individual miRNAs is involved in several layers of tolerance impairments, such as inefficient Treg induction and Treg instability. The targeted modulation of miRNAs using specific inhibitors, resulting in improved immune homeostasis, as well as improved methods for the targeting of miRNAs, suggest that miRNAs, especially in T cells, are a promising target for the reestablishment of immune tolerance.
Collapse
Affiliation(s)
- Martin G. Scherm
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Munich-Neuherberg, Germany
| | - Carolin Daniel
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Munich-Neuherberg, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, Munich, Germany
- *Correspondence: Carolin Daniel,
| |
Collapse
|
35
|
Xie Z, Chang C, Huang G, Zhou Z. The Role of Epigenetics in Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1253:223-257. [PMID: 32445098 DOI: 10.1007/978-981-15-3449-2_9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the interaction between genetic alterations and environmental factors. More than 60 susceptible genes or loci of T1D have been identified. Among them, HLA regions are reported to contribute about 50% of genetic susceptibility in Caucasians. There are many environmental factors involved in the pathogenesis of T1D. Environmental factors may change the expression of genes through epigenetic mechanisms, thus inducing individuals with susceptible genes to develop T1D; however, the underlying mechanisms remain poorly understood. The major epigenetic modifications include DNA methylation, histone modification, and non-coding RNA. There has been extensive research on the role of epigenetic mechanisms including aberrant DNA methylation, histone modification, and microRNA in the pathogenesis of T1D. DNA methylation and microRNA have been proposed as biomarkers to predict islet β cell death, which needs further confirmation before any clinical application can be developed. Small molecule inhibitors of histone deacetylases, histone methylation, and DNA methylation are potentially important for preventing T1D or in the reprogramming of insulin-producing cells. This chapter mainly focuses on T1D-related DNA methylation, histone modification, and non-coding RNA, as well as their possible translational potential in the early diagnosis and treatment of T1D.
Collapse
Affiliation(s)
- Zhiguo Xie
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, 410011, Hunan, China
| | - Christopher Chang
- Division of Pediatric Immunology and Allergy, Joe DiMaggio Children's Hospital, Hollywood, FL, 33021, USA.,Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, Davis, CA, 95616, USA
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, 410011, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, 410011, Hunan, China.
| |
Collapse
|
36
|
Yue HQ, Zhou YH, Guo Y, Tang CY, Wang F, Zhou HD. Serum miR-503 is a Candidate Biomarker for Differentiating Metabolic Healthy Obesity from Metabolic Unhealthy Obesity. Diabetes Metab Syndr Obes 2020; 13:2667-2676. [PMID: 32821139 PMCID: PMC7419640 DOI: 10.2147/dmso.s262888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/08/2020] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Overweight and obesity are associated with metabolic diseases. However, a subgroup of the overweight/obese population does not present metabolic abnormalities. Hence, there is an urgent need to identify biomarkers that can distinguish different obesity phenotypes and metabolic status. PATIENTS AND METHODS A total of 98 individuals were divided into three groups: metabolically healthy normal weight (MHNW), metabolically healthy obese (MHO), and metabolically unhealthy obese (MUO). Participants were evaluated for anthropometric and biochemical parameters and serum BMPR1A concentration and miR-503 level. Receiver operating characteristic (ROC) curve analysis and logistic regression analysis were performed. RESULTS The level of miR-503 was significantly higher in the MHO group compared with that in the MUO group, but no difference was observed between the MHNW and MHO groups. Meanwhile, no significant differences in serum BMPR1A concentration were observed between the three groups. ROC curve analysis showed that miR-503 could be used as a marker to distinguish the MUO from the MHO. Logistic regression analysis suggested that miR-503 was an important related factor associated with an unhealthy metabolic state in overweight/obese subjects. CONCLUSION miR-503 can be considered as a suitable biomarker to distinguish between the MUO and MHO, which may be a related factor for the incidence of metabolic disorders in overweight/obese subjects.
Collapse
Affiliation(s)
- Hai-Qing Yue
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of China
| | - Ying-Hui Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of China
| | - Yue Guo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of China
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Chen-Yi Tang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of China
| | - Fang Wang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of China
- Correspondence: Hou-De Zhou National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha410011, Hunan, People’s Republic of ChinaTel +86-731-85292223Fax +86-731-85533525 Email
| |
Collapse
|
37
|
Scherm MG, Serr I, Zahm AM, Schug J, Bellusci S, Manfredini R, Salb VK, Gerlach K, Weigmann B, Ziegler AG, Kaestner KH, Daniel C. miRNA142-3p targets Tet2 and impairs Treg differentiation and stability in models of type 1 diabetes. Nat Commun 2019; 10:5697. [PMID: 31836704 PMCID: PMC6910913 DOI: 10.1038/s41467-019-13587-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/14/2019] [Indexed: 12/31/2022] Open
Abstract
In type 1 diabetes, the appearance of islet autoantibodies indicates the onset of islet autoimmunity, often many years before clinical symptoms arise. While T cells play a major role in the destruction of pancreatic beta cells, molecular underpinnings promoting aberrant T cell activation remain poorly understood. Here, we show that during islet autoimmunity an miR142-3p/Tet2/Foxp3 axis interferes with the efficient induction of regulatory T (Treg) cells, resulting in impaired Treg stability in mouse and human. Specifically, we demonstrate that miR142-3p is induced in islet autoimmunity and that its inhibition enhances Treg induction and stability, leading to reduced islet autoimmunity in non-obese diabetic mice. Using various cellular and molecular approaches we identify Tet2 as a direct target of miR142-3p, thereby linking high miR142-3p levels to epigenetic remodeling in Tregs. These findings offer a mechanistic model where during islet autoimmunity miR142-3p/Tet2-mediated Treg instability contributes to autoimmune activation and progression.
Collapse
Affiliation(s)
- Martin G Scherm
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939, Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), 85764, Munich-Neuherberg, Germany
| | - Isabelle Serr
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939, Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), 85764, Munich-Neuherberg, Germany
| | - Adam M Zahm
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Saverio Bellusci
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, 35390, Giessen, Germany
| | - Rossella Manfredini
- Center for Regenerative Medicine, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Victoria K Salb
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939, Munich, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), 85764, Munich-Neuherberg, Germany
| | - Katharina Gerlach
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Benno Weigmann
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, at Klinikum rechts der Isar, 80333, Munich, Germany
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Carolin Daniel
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939, Munich, Germany.
- Deutsches Zentrum für Diabetesforschung (DZD), 85764, Munich-Neuherberg, Germany.
- Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, 80337, Munich, Germany.
| |
Collapse
|
38
|
Speake C, Skinner SO, Berel D, Whalen E, Dufort MJ, Young WC, Odegard JM, Pesenacker AM, Gorus FK, James EA, Levings MK, Linsley PS, Akirav EM, Pugliese A, Hessner MJ, Nepom GT, Gottardo R, Long SA. A composite immune signature parallels disease progression across T1D subjects. JCI Insight 2019; 4:126917. [PMID: 31671072 PMCID: PMC6962023 DOI: 10.1172/jci.insight.126917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
At diagnosis, most people with type 1 diabetes (T1D) produce measurable levels of endogenous insulin, but the rate at which insulin secretion declines is heterogeneous. To explain this heterogeneity, we sought to identify a composite signature predictive of insulin secretion, using a collaborative assay evaluation and analysis pipeline that incorporated multiple cellular and serum measures reflecting β cell health and immune system activity. The ability to predict decline in insulin secretion would be useful for patient stratification for clinical trial enrollment or therapeutic selection. Analytes from 12 qualified assays were measured in shared samples from subjects newly diagnosed with T1D. We developed a computational tool (DIFAcTO, Data Integration Flexible to Account for different Types of data and Outcomes) to identify a composite panel associated with decline in insulin secretion over 2 years following diagnosis. DIFAcTO uses multiple filtering steps to reduce data dimensionality, incorporates error estimation techniques including cross-validation and sensitivity analysis, and is flexible to assay type, clinical outcome, and disease setting. Using this novel analytical tool, we identified a panel of immune markers that, in combination, are highly associated with loss of insulin secretion. The methods used here represent a potentially novel process for identifying combined immune signatures that predict outcomes relevant for complex and heterogeneous diseases like T1D.
Collapse
Affiliation(s)
- Cate Speake
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Samuel O. Skinner
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Dror Berel
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Elizabeth Whalen
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Matthew J. Dufort
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - William Chad Young
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jared M. Odegard
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Anne M. Pesenacker
- University of British Columbia BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Frans K. Gorus
- Diabetes Research Center, Medical School and University Hospital (UZ Brussel), Brussels Free University Vrije Universiteit Brussel, Brussels, Belgium
| | - Eddie A. James
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Megan K. Levings
- University of British Columbia BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Peter S. Linsley
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Eitan M. Akirav
- Research Institute, Islet Biology, New York University Winthrop Hospital, Mineola, New York, USA
- Stony Brook University School of Medicine, Stony Brook, New York, USA
| | - Alberto Pugliese
- Diabetes Research Institute, Department of Medicine, Division of Diabetes Endocrinology and Metabolism, Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | | | - Gerald T. Nepom
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
- Immune Tolerance Network, Bethesda, Maryland, USA
| | - Raphael Gottardo
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - S. Alice Long
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| |
Collapse
|
39
|
Dwan BF, Moore A, Wang P. Nucleic acid-based theranostics in type 1 diabetes. Transl Res 2019; 214:50-61. [PMID: 31491371 DOI: 10.1016/j.trsl.2019.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/01/2019] [Accepted: 08/17/2019] [Indexed: 12/12/2022]
Abstract
Application of RNAi interference for type 1 diabetes (T1D) therapy bears tremendous potential. This review will discuss vehicles for oligonucleotide delivery, imaging modalities used for delivery monitoring, therapeutic targets, and different theranostic strategies that can be applied for T1D treatment.
Collapse
Affiliation(s)
- Bennett Francis Dwan
- Precision Health Program, Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, Michigan; College of Natural Science, Michigan State University, East Lansing, Michigan
| | - Anna Moore
- Precision Health Program, Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, Michigan
| | - Ping Wang
- Precision Health Program, Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
40
|
MicroRNA Signatures as Future Biomarkers for Diagnosis of Diabetes States. Cells 2019; 8:cells8121533. [PMID: 31795194 PMCID: PMC6953078 DOI: 10.3390/cells8121533] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/24/2022] Open
Abstract
Diabetes results from the inability of pancreatic islets to maintain blood glucose concentrations within a normal physiological range. Clinical features are usually not observed until islets begin to fail and irreversible damage has occurred. Diabetes is generally diagnosed based on elevated glucose, which does not distinguish between type 1 and 2 diabetes. Thus, new diagnostic approaches are needed to detect different modes of diabetes before manifestation of disease. During prediabetes (pre-DM), islets undergo stress and release micro (mi) RNAs. Here, we review studies that have measured and tracked miRNAs in the blood for those with recent-onset or longstanding type 1 diabetes, obesity, pre-diabetes, type 2 diabetes, and gestational diabetes. We summarize the findings on miRNA signatures with the potential to stage progression of different modes of diabetes. Advances in identifying selective biomarker signatures may aid in early detection and classification of diabetic conditions and treatments to prevent and reverse diabetes.
Collapse
|
41
|
Krishnan P, Syed F, Jiyun Kang N, G. Mirmira R, Evans-Molina C. Profiling of RNAs from Human Islet-Derived Exosomes in a Model of Type 1 Diabetes. Int J Mol Sci 2019; 20:ijms20235903. [PMID: 31775218 PMCID: PMC6928620 DOI: 10.3390/ijms20235903] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 12/30/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by the immune-mediated destruction of insulin-producing islet β cells. Biomarkers capable of identifying T1D risk and dissecting disease-related heterogeneity represent an unmet clinical need. Toward the goal of informing T1D biomarker strategies, we profiled coding and noncoding RNAs in human islet-derived exosomes and identified RNAs that were differentially expressed under proinflammatory cytokine stress conditions. Human pancreatic islets were obtained from cadaveric donors and treated with/without IL-1β and IFN-γ. Total RNA and small RNA sequencing were performed from islet-derived exosomes to identify mRNAs, long noncoding RNAs, and small noncoding RNAs. RNAs with a fold change ≥1.3 and a p-value <0.05 were considered as differentially expressed. mRNAs and miRNAs represented the most abundant long and small RNA species, respectively. Each of the RNA species showed altered expression patterns with cytokine treatment, and differentially expressed RNAs were predicted to be involved in insulin secretion, calcium signaling, necrosis, and apoptosis. Taken together, our data identify RNAs that are dysregulated under cytokine stress in human islet-derived exosomes, providing a comprehensive catalog of protein coding and noncoding RNAs that may serve as potential circulating biomarkers in T1D.
Collapse
Affiliation(s)
- Preethi Krishnan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (F.S.); (N.J.K.); (R.G.M.)
| | - Farooq Syed
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (F.S.); (N.J.K.); (R.G.M.)
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nicole Jiyun Kang
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (F.S.); (N.J.K.); (R.G.M.)
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G. Mirmira
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (F.S.); (N.J.K.); (R.G.M.)
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carmella Evans-Molina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (F.S.); (N.J.K.); (R.G.M.)
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
- Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-274-4145; Fax: +1-317-274-4107
| |
Collapse
|
42
|
Xu G, Thielen LA, Chen J, Grayson TB, Grimes T, Bridges SL, Tse HM, Smith B, Patel R, Li P, Evans-Molina C, Ovalle F, Shalev A. Serum miR-204 is an early biomarker of type 1 diabetes-associated pancreatic beta-cell loss. Am J Physiol Endocrinol Metab 2019; 317:E723-E730. [PMID: 31408375 PMCID: PMC6842918 DOI: 10.1152/ajpendo.00122.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pancreatic beta-cell death is a major factor in the pathogenesis of type 1 diabetes (T1D), but straightforward methods to measure beta-cell loss in humans are lacking, underlining the need for novel biomarkers. Using studies in INS-1 cells, human islets, diabetic mice, and serum samples of subjects with T1D at different stages, we have identified serum miR-204 as an early biomarker of T1D-associated beta-cell loss in humans. MiR-204 is a highly enriched microRNA in human beta-cells, and we found that it is released from dying beta-cells and detectable in human serum. We further discovered that serum miR-204 was elevated in children and adults with T1D and in autoantibody-positive at-risk subjects but not in type 2 diabetes or other autoimmune diseases and was inversely correlated with remaining beta-cell function in recent-onset T1D. Thus, serum miR-204 may provide a much needed novel approach to assess early T1D-associated human beta-cell loss even before onset of overt disease.
Collapse
Affiliation(s)
- Guanlan Xu
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lance A Thielen
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Junqin Chen
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Truman B Grayson
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tiffany Grimes
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - S Louis Bridges
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hubert M Tse
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Blair Smith
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rakesh Patel
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Peng Li
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carmella Evans-Molina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Fernando Ovalle
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anath Shalev
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
43
|
miR-342-5p inhibits expression of Bmp7 to regulate proliferation, differentiation and migration of osteoblasts. Mol Immunol 2019; 114:251-259. [DOI: 10.1016/j.molimm.2019.07.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 11/19/2022]
|
44
|
Abstract
BACKGROUND microRNAs (miRNAs) have emerged as critical contributors to immune regulation and homeostasis, and their dysregulation is involved in the aberrant differentiation and function of T cell subsets. In type 1 diabetes (T1D), the clinically overt disease is preceded by a presymptomatic phase which is marked by the presence of islet autoantibodies while the individual is still normoglycemic. Recent analyses revealed impaired regulatory T (Treg) cell induction from naive CD4+ T cells during this early phase of autoimmunity. SCOPE OF THE REVIEW In this review article, we aim to discuss important recent insights into miRNA regulation of immune homeostasis and activation. Specifically, we highlight the role of miRNAs as biomarkers in autoimmunity and T1D as well as the contribution of specific miRNAs and their downstream pathways to the onset and progression of islet immunity. Furthermore, we focus on critical next steps required to establish miRNAs as biomarkers to predict disease onset and progression and as novel targets of future prevention and treatment strategies to control autoimmunity. MAJOR CONCLUSIONS Several recent studies have provided considerable insight into the miRNA regulation of immune homeostasis and how dysregulated miRNAs contribute to onset and progression of islet autoimmunity. Specifically, high levels of individual miRNAs such as miR92a and miR181a are involved in impaired Treg induction during the onset of islet autoimmunity, thereby contributing to disease pathogenesis. The recent advancements in the field suggest miRNAs as potential biomarkers for islet autoimmunity and their direct targeting, especially in a T cell-specific manner, could contribute to the reestablishment of immune homeostasis and ultimately interfere with the onset of islet autoimmunity.
Collapse
Affiliation(s)
- Martin G Scherm
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, 80939, Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), Ingolstaedter Landstrasse 1, 85764, Munich-Neuherberg, Germany
| | - Isabelle Serr
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, 80939, Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), Ingolstaedter Landstrasse 1, 85764, Munich-Neuherberg, Germany
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Carolin Daniel
- Institute of Diabetes Research, Group Immune Tolerance in Type 1 Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Heidemannstrasse 1, 80939, Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), Ingolstaedter Landstrasse 1, 85764, Munich-Neuherberg, Germany; Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, 80337, Munich, Germany.
| |
Collapse
|
45
|
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
|
46
|
Mazzeo A, Lopatina T, Gai C, Trento M, Porta M, Beltramo E. Functional analysis of miR-21-3p, miR-30b-5p and miR-150-5p shuttled by extracellular vesicles from diabetic subjects reveals their association with diabetic retinopathy. Exp Eye Res 2019; 184:56-63. [DOI: 10.1016/j.exer.2019.04.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 10/27/2022]
|
47
|
Nakayasu ES, Qian WJ, Evans-Molina C, Mirmira RG, Eizirik DL, Metz TO. The role of proteomics in assessing beta-cell dysfunction and death in type 1 diabetes. Expert Rev Proteomics 2019; 16:569-582. [PMID: 31232620 PMCID: PMC6628911 DOI: 10.1080/14789450.2019.1634548] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/18/2019] [Indexed: 12/17/2022]
Abstract
Introduction: Type 1 diabetes (T1D) is characterized by autoimmune-induced dysfunction and destruction of the pancreatic beta cells. Unfortunately, this process is poorly understood, and the current best treatment for type 1 diabetes is the administration of exogenous insulin. To better understand these mechanisms and to develop new therapies, there is an urgent need for biomarkers that can reliably predict disease stage. Areas covered: Mass spectrometry (MS)-based proteomics and complementary techniques play an important role in understanding the autoimmune response, inflammation and beta-cell death. MS is also a leading technology for the identification of biomarkers. This, and the technical difficulties and new technologies that provide opportunities to characterize small amounts of sample in great depth and to analyze large sample cohorts will be discussed in this review. Expert opinion: Understanding disease mechanisms and the discovery of disease-associated biomarkers are highly interconnected goals. Ideal biomarkers would be molecules specific to the different stages of the disease process that are released from beta cells to the bloodstream. However, such molecules are likely to be present in trace amounts in the blood due to the small number of pancreatic beta cells in the human body and the heterogeneity of the target organ and disease process.
Collapse
Affiliation(s)
- Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Raghavendra G. Mirmira
- Center for Diabetes and Metabolic Diseases, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Thomas O. Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| |
Collapse
|
48
|
Wang F, Liang R, Tandon N, Matthews ER, Shrestha S, Yang J, Soibam B, Yang J, Liu Y. H19X-encoded miR-424(322)/-503 cluster: emerging roles in cell differentiation, proliferation, plasticity and metabolism. Cell Mol Life Sci 2019; 76:903-920. [PMID: 30474694 PMCID: PMC6394552 DOI: 10.1007/s00018-018-2971-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
miR-424(322)/-503 are mammal-specific members of the extended miR-15/107 microRNA family. They form a co-expression network with the imprinted lncRNA H19 in tetrapods. miR-424(322)/-503 regulate fundamental cellular processes including cell cycle, epithelial-to-mesenchymal transition, hypoxia and other stress response. They control tissue differentiation (cardiomyocyte, skeletal muscle, monocyte) and remodeling (mammary gland involution), and paradoxically participate in tumor initiation and progression. Expression of miR-424(322)/-503 is governed by unique mechanisms involving sex hormones. Here, we summarize current literature and provide a primer for future endeavors.
Collapse
Affiliation(s)
- Fan Wang
- Department of Oncology, The First Affiliated Hospital of Xian Jiaotong University, Xi'an, 710061, Shaanxi, China
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Rui Liang
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Neha Tandon
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Elizabeth R Matthews
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Shreesti Shrestha
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Jiao Yang
- Department of Oncology, The First Affiliated Hospital of Xian Jiaotong University, Xi'an, 710061, Shaanxi, China
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Benjamin Soibam
- Computer Science and Engineering Technology, University of Houston-Downtown, Houston, TX, 77002, USA
| | - Jin Yang
- Department of Oncology, The First Affiliated Hospital of Xian Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
49
|
Mollaei H, Safaralizadeh R, Rostami Z. MicroRNA replacement therapy in cancer. J Cell Physiol 2019; 234:12369-12384. [PMID: 30605237 DOI: 10.1002/jcp.28058] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Despite the recent progress in cancer management approaches, the mortality rate of cancer is still growing and there are lots of challenges in the clinics in terms of novel therapeutics. MicroRNAs (miRNA) are regulatory small noncoding RNAs and are already confirmed to have a great role in regulating gene expression level by targeting multiple molecules that affect cell physiology and disease development. Recently, miRNAs have been introduced as promising therapeutic targets for cancer treatment. Regulatory potential of tumor suppressor miRNAs, which enables regulation of entire signaling networks within the cells, makes them an interesting option for developing cancer therapeutics. In this regard, over recent decades, scientists have aimed at developing powerful and safe targeting approaches to restore these suppressive miRNAs in cancerous cells. The present review summarizes the function of miRNAs in tumor development and presents recent findings on how miRNAs have served as therapeutic agents against cancer, with a special focus on tumor suppressor miRNAs (mimics). Moreover, the latest investigations on the therapeutic strategies of miRNA delivery have been presented.
Collapse
Affiliation(s)
- Homa Mollaei
- Department of Biology, Faculty of Sciences, University of Birjand, Birjand, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zeinab Rostami
- Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
50
|
Liu Y, Yang Y, Wang Q, Kahaer A, Zhang J, Liao J, Abudureyimu M, Yahefu R, Qi J, Zhao L, Zhu J. Regulatory Effect of 1,25(OH)2D3 on TGF- β1 and miR-130b Expression in Streptozotocin-Induced Diabetic Nephropathy in Rats. Int J Endocrinol 2019; 2019:1231346. [PMID: 31781203 PMCID: PMC6875179 DOI: 10.1155/2019/1231346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To investigate the role of microRNA-130b in 1,25(OH)2D3 mediated improvement of renal fibrosis via transforming growth factor-beta 1 in a rat model of diabetic nephropathy (DN). METHODS DN was induced in 30 rats by intraperitoneal injection of streptozotocin. These rats were randomly allocated to the DN group, TGF-β1 overexpression group (in situ injection of TGF-β1 lentivirus to kidney tissues), and TGF-β1 siRNA group (in situ injection of TGF-β1 siRNA lentivirus to kidney tissues). Rats with different expression levels of TGF-β1 were administered 1,25(OH)2D3 (0.03 μg/kg/d) or peanut oil as control. DN rats were treated only with peanut oil. All rats were randomly divided into five groups (n = 6 per group): TGF-β1 overexpression + oil, TGF-β1 overexpression + 1,25(OH)2D3, TGF-β1 siRNA + oil, TGF-β1 siRNA + 1,25(OH)2D3, and DN + oil groups. After 37 days, kidney samples were collected and the expression of TGF-β1 and miR-130b was determined by real-time PCR, western blotting, and immunohistochemistry. Hematoxylin and eosin staining and Masson staining were used to evaluate kidney morphological and fibrogenic changes. Differences were determined using ANOVA and Student's t-test. RESULTS RT-PCR, western blotting, and immunohistochemistry revealed that interference of TGF-β1 significantly decreased mRNA and protein levels of TGF-β1 in renal tissues of DN rats compared to those in renal tissues of rats overexpressing TGF-β1 (p < 0.05). Histological analysis showed that upregulated TGF-β1 led to disorganized kidney structure and severe kidney fibrosis. The expression of miR-130b was significantly lowered upon lentivirus-mediated overexpression of TGF-β1 than upon downregulation of TGF-β1 (p < 0.05). Treatment with 1,25(OH)2D3 led to a significant reduction of TGF-β1 at the mRNA and protein levels (both p < 0.05), improvement of renal structure and fibrosis, and an increase in miR-130b expression (p < 0.05). CONCLUSION TGF-β1 can decrease the expression of miR-130b in kidney tissues of DN rats. Moreover, miR-130b may be involved in the protective effect of 1,25(OH)2D3 on renal fibrosis via TGF-β1.
Collapse
Affiliation(s)
- Yuetong Liu
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
| | - Ye Yang
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Qin Wang
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Apaer Kahaer
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jiyun Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jing Liao
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Mairemugu Abudureyimu
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Reyila Yahefu
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jing Qi
- Department of Chu Medical, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Lei Zhao
- Department of Chu Medical, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jun Zhu
- Department of Endocrinology, People's Hospital of Shenzhen Baoan District, The Second School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518101, China
| |
Collapse
|