101
|
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: 1.6] [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
|
102
|
Qi L, Wang R, Shi Q, Yuan M, Jin M, Li D. Umbilical cord mesenchymal stem cell conditioned medium restored the expression of collagen II and aggrecan in nucleus pulposus mesenchymal stem cells exposed to high glucose. J Bone Miner Metab 2019; 37:455-466. [PMID: 30187277 DOI: 10.1007/s00774-018-0953-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/21/2018] [Indexed: 12/16/2022]
Abstract
Diabetes can cause intervertebral disc degeneration by accelerating apoptosis and senescence of nucleus pulposus mesenchymal stem cells (NPMSCs). The aim of this study was to determine the effect of umbilical cord mesenchymal stem cells (UCMSCs) conditioned medium on high glucose (HG) induced degradation of NPMSCs produced extracellular matrix. NPMSCs were isolated from the inner intervertebral disc tissue using type XI collagenase digestion. According to Annexin V/propidium iodide (PI) flow cytometry analysis; HG leads to an increase in the rate of NPMSCs apoptosis. HG injury also resulted in a marked decrease in the percentage of cells in G0/G1 phase and an increase in cells in S and G2/M phases, indicating that HG induces cell cycle arrest of NPMSCs. Treatment with MSC-CM abolished the effect of HG on cell senescence. HG also significantly inhibited collagen II and aggrecan expression in NPMSCs. After MSC-CM treatment, the expression of these two extracellular matrix components was restored. Exposure to HG resulted in phosphorylation of p38 MAPK, while the levels of total p38 MAPK were not affected. When treated with MSC-CM, phosphorylated p38 MAPK levels of NPMSCs were lower than those without CM treatment. Our data also showed that p38 MAPK inhibitor SB203580 can attenuated phosphorylation of p38 MAPK and resumed the collagen II and aggrecan expression in NPMSCs. In summary, this study demonstrated that MSC-CM has the potential to alleviate HG induced extracellular matrix degradation via the p38 MAPK pathway.
Collapse
Affiliation(s)
- Lei Qi
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Ran Wang
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Qing Shi
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Ming Yuan
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Min Jin
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
- Stem Cell and Regenerative Medicine Research Center of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
103
|
Castaño C, Novials A, Párrizas M. Exosomes and diabetes. Diabetes Metab Res Rev 2019; 35:e3107. [PMID: 30513130 DOI: 10.1002/dmrr.3107] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Diabetes is a group of metabolic diseases characterized by elevated blood glucose levels that drive the development of life-threatening complications. Diabetes results from a situation of insufficient insulin action, either by deficient production of the hormone by the pancreas, or by the development of insulin resistance in peripheral tissues such as liver, muscle, or the adipose depots. Communication between organs is thus central to the maintenance of glucose homoeostasis. Recently, several studies are evidencing that small vesicles called exosomes released by, amongst other, the adipose tissue can regulate gene expression in other tissues, hence modulating interorgan crosstalk. Therefore, exosomes participate in the development of diabetes and its associated complications. Their study holds the potential of providing us with novel biomarkers for the early diagnosis and stratification of patients at risk of developing diabetes, hence allowing the timely implementation of more personalized therapies. On the other hand, the molecular dissection of the pathways initiated by exosomes under situations of metabolic stress could help to gain a deeper knowledge of the pathophysiology of diabetes and its associated metabolic diseases.
Collapse
Affiliation(s)
- Carlos Castaño
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| | - Marcelina Párrizas
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Barcelona, Spain
| |
Collapse
|
104
|
Zhao C, Lai S, Wu D, Liu D, Zou X, Ismail A, El-Seedi H, Arroo RR, Xiao J. miRNAs as Regulators of Antidiabetic Effects of Fucoidans. EFOOD 2019. [DOI: 10.2991/efood.k.190822.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
105
|
Suksangrat T, Phannasil P, Jitrapakdee S. miRNA Regulation of Glucose and Lipid Metabolism in Relation to Diabetes and Non-alcoholic Fatty Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:129-148. [DOI: 10.1007/978-3-030-12668-1_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
106
|
Comparison of Cardiac miRNA Transcriptomes Induced by Diabetes and Rapamycin Treatment and Identification of a Rapamycin-Associated Cardiac MicroRNA Signature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8364608. [PMID: 30647817 PMCID: PMC6311877 DOI: 10.1155/2018/8364608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/16/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
Abstract
Rapamycin (Rap), an inhibitor of mTORC1, reduces obesity and improves lifespan in mice. However, hyperglycemia and lipid disorders are adverse side effects in patients receiving Rap treatment. We previously reported that diabetes induces pansuppression of cardiac cytokines in Zucker obese rats (ZO-C). Rap treatment (750 μg/kg/day for 12 weeks) reduced their obesity and cardiac fibrosis significantly; however, it increased their hyperglycemia and did not improve their cardiac diastolic parameters. Moreover, Rap treatment of healthy Zucker lean rats (ZL-C) induced cardiac fibrosis. Rap-induced changes in ZL-C's cardiac cytokine profile shared similarities with that of diabetes-induced ZO-C. Therefore, we hypothesized that the cardiac microRNA transcriptome induced by diabetes and Rap treatment could share similarities. Here, we compared the cardiac miRNA transcriptome of ZL-C to ZO-C, Rap-treated ZL (ZL-Rap), and ZO (ZO-Rap). We report that 80% of diabetes-induced miRNA transcriptome (40 differentially expressed miRNAs by minimum 1.5-fold in ZO-C versus ZL-C; p ≤ 0.05) is similar to 47% of Rap-induced miRNA transcriptome in ZL (68 differentially expressed miRNAs by minimum 1.5-fold in ZL-Rap versus ZL-C; p ≤ 0.05). This remarkable similarity between diabetes-induced and Rap-induced cardiac microRNA transcriptome underscores the role of miRNAs in Rap-induced insulin resistance. We also show that Rap treatment altered the expression of the same 17 miRNAs in ZL and ZO hearts indicating that these 17 miRNAs comprise a unique Rap-induced cardiac miRNA signature. Interestingly, only four miRNAs were significantly differentially expressed between ZO-C and ZO-Rap, indicating that, unlike the nondiabetic heart, Rap did not substantially change the miRNA transcriptome in the diabetic heart. In silico analyses showed that (a) mRNA-miRNA interactions exist between differentially expressed cardiac cytokines and miRNAs, (b) human orthologs of rat miRNAs that are strongly correlated with cardiac fibrosis may modulate profibrotic TGF-β signaling, and (c) changes in miRNA transcriptome caused by diabetes or Rap treatment include cardioprotective miRNAs indicating a concurrent activation of an adaptive mechanism to protect the heart in conditions that exacerbate diabetes.
Collapse
|
107
|
Sims EK, Evans-Molina C, Tersey SA, Eizirik DL, Mirmira RG. Biomarkers of islet beta cell stress and death in type 1 diabetes. Diabetologia 2018; 61:2259-2265. [PMID: 30112687 PMCID: PMC6160346 DOI: 10.1007/s00125-018-4712-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022]
Abstract
Recent work on the pathogenesis of type 1 diabetes has led to an evolving recognition of the heterogeneity of this disease, both with regards to clinical phenotype and responses to therapies to prevent or revert diabetes. This heterogeneity not only limits efforts to accurately predict clinical disease but also is reflected in differing responses to immunomodulatory therapeutics. Thus, there is a need for robust biomarkers of beta cell health, which could provide insight into pathophysiological differences in disease course, improve disease prediction, increase the understanding of therapeutic responses to immunomodulatory interventions and identify individuals most likely to benefit from these therapies. In this review, we outline current literature, limitations and future directions for promising circulating markers of beta cell stress and death in type 1 diabetes, including markers indicating abnormal prohormone processing, circulating RNAs and circulating DNAs.
Collapse
Affiliation(s)
- Emily K Sims
- Center for Diabetes and Metabolic Diseases, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 635 Barnhill Drive, MS2031, Indianapolis, IN, 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 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, 635 Barnhill Drive, MS2031, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, US Department of Veterans Affairs, Indianapolis, IN, USA
| | - Sarah A Tersey
- Center for Diabetes and Metabolic Diseases, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 635 Barnhill Drive, MS2031, Indianapolis, IN, 46202, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Raghavendra G Mirmira
- Center for Diabetes and Metabolic Diseases, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 635 Barnhill Drive, MS2031, Indianapolis, IN, 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
108
|
Yi L, Swensen AC, Qian WJ. Serum biomarkers for diagnosis and prediction of type 1 diabetes. Transl Res 2018; 201:13-25. [PMID: 30144424 PMCID: PMC6177288 DOI: 10.1016/j.trsl.2018.07.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/02/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes (T1D) culminates in the autoimmune destruction of the pancreatic βcells, leading to insufficient production of insulin and development of hyperglycemia. Serum biomarkers including a combination of glucose, glycated molecules, C-peptide, and autoantibodies have been well established for the diagnosis of T1D. However, these molecules often mark a late stage of the disease when ∼90% of the pancreatic insulin-producing β-cells have already been lost. With the prevalence of T1D increasing worldwide and because of the physical and psychological burden induced by this disease, there is a great need for prognostic biomarkers to predict T1D development or progression. This would allow us to identify individuals at high risk for early prevention and intervention. Therefore, considerable efforts have been dedicated to the understanding of disease etiology and the discovery of novel biomarkers in the last few decades. The advent of high-throughput and sensitive "-omics" technologies for the study of proteins, nucleic acids, and metabolites have allowed large scale profiling of protein expression and gene changes in T1D patients relative to disease-free controls. In this review, we briefly discuss the classical diagnostic biomarkers of T1D but mainly focus on the novel biomarkers that are identified as markers of β-cell destruction and screened with the use of state-of-the-art "-omics" technologies.
Collapse
Affiliation(s)
- Lian Yi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington.
| |
Collapse
|
109
|
Abstract
PURPOSE OF REVIEW The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine's extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|
110
|
Dotta F, Ventriglia G, Snowhite IV, Pugliese A. MicroRNAs: markers of β-cell stress and autoimmunity. Curr Opin Endocrinol Diabetes Obes 2018; 25:237-245. [PMID: 29846238 DOI: 10.1097/med.0000000000000420] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We discuss current knowledge about microRNAs (miRNAs) in type 1 diabetes (T1D), an autoimmune disease leading to severe loss of pancreatic β-cells. We describe: the role of cellular miRNAs in regulating immune functions and pathways impacting insulin secretion and β-cell survival; circulating miRNAs as disease biomarkers. RECENT FINDINGS Studies examined miRNAs in experimental models and patients, including analysis of tissues from organ donors, peripheral blood cells, and circulating miRNAs in serum, plasma, and exosomes. Studies employed diverse designs and methodologies to detect miRNAs and measure their levels. Selected miRNAs have been linked to the regulation of key biological pathways and disease pathogenesis; several circulating miRNAs are associated with having T1D, islet autoimmunity, disease progression, and immune and metabolic functions, for example, C-peptide secretion, in multiple studies. SUMMARY A growing literature reveals multiple roles of miRNAs in T1D, provide new clues into the regulation of disease mechanisms, and identify reproducible associations. Yet challenges remain, and the field will benefit from joint efforts to analyze results, compare methodologies, formally test the robustness of miRNA associations, and ultimately move towards validating robust miRNA biomarkers.
Collapse
Affiliation(s)
- Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena
- Fondazione Umberto di Mario, Toscana Life Sciences, Siena, Italy
| | - Giuliana Ventriglia
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena
- Fondazione Umberto di Mario, Toscana Life Sciences, Siena, Italy
| | | | - Alberto Pugliese
- Diabetes Research Institute
- Department of Medicine, Division of Endocrinology and Metabolism
- Department of Microbiology and Immunology, Leonard Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
111
|
Xu Y, Wu D, Jiang Z, Zhang Y, Wang S, Ma Z, Hui B, Wang J, Qian W, Ge Z, Sun L. MiR-616-3p modulates cell proliferation and migration through targeting tissue factor pathway inhibitor 2 in preeclampsia. Cell Prolif 2018; 51:e12490. [PMID: 30028057 DOI: 10.1111/cpr.12490] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/03/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Despite improvements in diagnosis and treatment, preeclampsia (PE) continues to pose a significant risk of maternal and foetal morbidity and mortality if not addressed promptly. An increasing number of studies have suggested that tissue factor pathway inhibitor 2 (TFPI2) acts as a suppressor gene, possibly inhibiting multiple serine proteases affecting cell proliferation and migration. It plays an essential role in the occurrence and development of PE, but the pathogenesis remains unclear. MATERIALS AND METHODS In our research, we performed western blotting, immunohistochemistry and qPCR assays to investigate TFPI2 and miR-616-3p expression in preeclamptic placental tissues. Cell assays were performed in HTR-8/SVneo and JEG3 cell lines. Cell proliferation and migration events were investigated by MTT, EdU and transwell assays. In conjunction with bioinformatics analysis, luciferase reporter assays were performed to elucidate the mechanism by which miR-616-3p binds to TFPI2 mRNA. RESULTS We established that TFPI2 protein levels were significantly upregulated in PE placental tissues. In addition, we found that miR-616-3p binds specifically to the 3'-UTR region of TFPI2 mRNA. Furthermore, miR-616-3p knockdown or TFPI2 overexpression substantially impaired cell growth and migration, whereas miR-616-3p upregulation or TFPI2 knockdown stimulated cell proliferation and migration. This miR-616-3p/TFPI2 axis was also found to affect the epithelial-mesenchymal transition process in PE. CONCLUSIONS Our results demonstrated that TFPI2 plays a vital role in the progression of PE and might provide a prospective therapeutic strategy to mitigate the severity of the disorder.
Collapse
Affiliation(s)
- Yetao Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of Obstetrics, Gynecology & Reproductive Sciences, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Dan Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ziyan Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuanyuan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sailan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhonghua Ma
- Department of Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Bingqing Hui
- Department of Oncology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jing Wang
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weiping Qian
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Shenzhen Hospital, FuTian District, Shenzhen, Guangdong, China
| | - Zhiping Ge
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lizhou Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|