1
|
Ji Y, Chen H, Pang L, Chen C, Wang S, Chen J, Fang L, Liu B, Cheng Y, Liu S, Zhong Y. AGE induced macrophage-derived exosomes induce endothelial dysfunction in diabetes via miR-22-5p/FOXP1. Cardiovasc Diabetol 2025; 24:158. [PMID: 40205587 PMCID: PMC11983961 DOI: 10.1186/s12933-025-02715-7] [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: 11/29/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Endothelial dysfunction is a pivotal contributor to cardiovascular complications in individuals with diabetes. However, the precise role of macrophages and their exosomes in the diabetic milieu remains elusive. METHODS Exosomes (Exos) were isolated from the supernatants of macrophages treated with advanced glycation end products (AGE) or bovine serum albumin (BSA) using ultracentrifugation. Following coculture with AGE-Exos or BSA-Exos, human umbilical vein endothelial cells (HUVECs) were subjected to CCK-8, EdU, cell migration, monocyte adhesion, and tube formation assays. ELISA and Western blotting were employed to assess inflammatory cytokine release and protein expression levels in HUVECs. The miRNA expression profiles of AGE-Exos and BSA-Exos were analysed using miRNA arrays. Potential targets of miR-22-5p were predicted via miRNA databases and validated through RT‒qPCR, dual-luciferase reporter assays, and rescue experiments. Furthermore, a Rab27a knockout mouse model of type 2 diabetes mellitus (T2DM) was established by intraperitoneal injection of Streptozotocin. Aortic tissues were analysed via immunofluorescence for CD63 and CD31 expression, immunohistochemistry for VCAM-1 and ICAM-1 expression, and Western blotting for FOXP1 expression. RESULTS AGE stimulation increased the secretion of exosomes from macrophages. Compared with BSA-Exos, AGE-Exos significantly impaired endothelial cell proliferation, migration, and tube formation capabilities while increasing monocyte adhesion and proinflammatory cytokine release without affecting cell viability. miR-22-5p was enriched in AGE-Exos, which were subsequently transferred to HUVECs, specifically targeting FOXP1, resulting in endothelial dysfunction. Overexpression of miR-22-5p in HUVECs using lentiviral vectors recapitulated the inflammatory effects observed with AGE-Exos, whereas anti-miR-22-5p conferred protective effects. Rab27a knockout significantly reduced exosome accumulation in T2DM model mouse aortic tissues, alleviating endothelial discontinuity, downregulating VCAM-1 and ICAM-1 expression, and upregulating FOXP1 expression. CONCLUSIONS AGE-induced release of macrophage-derived exosomes may partially depend on Rab27a transport, which delivers miR-22-5p to ECs. This miR-22-5p targets FOXP1 in ECs, leading to inflammation and resulting in endothelial dysfunction that accelerates the development of diabetic vascular lesions.
Collapse
MESH Headings
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Animals
- Exosomes/metabolism
- Exosomes/pathology
- Exosomes/drug effects
- Exosomes/transplantation
- Humans
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/pathology
- Human Umbilical Vein Endothelial Cells/drug effects
- Glycation End Products, Advanced/toxicity
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Cell Movement
- Male
- Signal Transduction
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/physiopathology
- rab27 GTP-Binding Proteins/genetics
- rab27 GTP-Binding Proteins/metabolism
- rab27 GTP-Binding Proteins/deficiency
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/physiopathology
- Neovascularization, Physiologic
- Serum Albumin, Bovine/toxicity
- Inflammation Mediators/metabolism
- Mice
- Cell Adhesion
- Repressor Proteins
Collapse
Affiliation(s)
- Yang Ji
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
- Department of Emergency, The Second Affliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Huanzhen Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
- DongGuan SongShan Lake Tungwah Hospital, Dongguan, Guangdong, China
| | - Lihua Pang
- Department of Emergency, The Second Affliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Changnong Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Sha Wang
- Department of Emergency, The Second Affliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Jing Chen
- Department of Cardiology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Lei Fang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Benrong Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Yongruo Cheng
- Department of Emergency, The Second Affliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China.
| | - Yun Zhong
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Afliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, China.
| |
Collapse
|
2
|
Zhang B, Liu S, Sun Y, Xu D. Endosulfan induced kidney cell injury by modulating ACE2 through up-regulating miR-429 in HK-2 cells. Toxicology 2023; 484:153392. [PMID: 36513242 DOI: 10.1016/j.tox.2022.153392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Endosulfan, a typical organochlorine pesticide, is widely used in agricultural countries and was detected in blood samples from the general population. Studies have shown a positive correlation between chronic kidney disease of unknown aetiology (CKDu) and endosulfan. CKDu has become endemic in agricultural countries, with clinical manifestations of tubulointerstitial fibrosis.The goal of this study was to investigate the effects of endosulfan in kidney cell injury in human renal tubular epithelial cells (HK-2), focusing on apoptosis, inflammatory response, and epithelial-mesenchymal transition (EMT). We found that endosulfan induced apoptosis in HK-2 cells by up-regulating the expression of BAX, APAF-1, Caspase-3 and mitochondrial Cytochrome c was released into the cytosol. Endosulfan caused an inflammatory response, showing the increase in the secretion and mRNA expression levels of IL-6/IL-8. Endosulfan triggered EMT, characterized by downregulation of E-cadherin and upregulation of Vimentin. Western blot results showed that p-Smad3 and Smad3 protein expression were elevated while the expression of Smad7 were decreased in endosulfan-exposed groups. Dual luciferase reporter assay confirmed the potential binding capacity of miR-429 to 3'-UTR of ACE2. Endosulfan causes upregulation of miR-429 and downregulation of ACE2 in HK-2 cells. Overexpression of miR-429 or silencing of ACE2 in HK-2 cells caused apoptosis, inflammation and EMT through TGF signaling pathway. These findings suggest that endosulfan can lead to kidney cell injury by modulating ACE2 through up-regulating miR-429, providing new evidence for the pathogenesis of CKDu.
Collapse
Affiliation(s)
- Boxiang Zhang
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China.
| | - Shiwen Liu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China.
| | - Yeqing Sun
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China.
| | - Dan Xu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian 116026, PR China.
| |
Collapse
|
3
|
Wang Y, Guo Y, Lu Y, Sun Y, Xu D. The effects of endosulfan on cell migration and invasion in prostate cancer cells via the KCNQ1OT1/miR-137-3p/PTP4A3 axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 845:157252. [PMID: 35817112 DOI: 10.1016/j.scitotenv.2022.157252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
Endosulfan belongs to persistent organic pollutants (POPs), closely related to an increased risk of prostate cancer (PCa). The existing evidence shows that lncRNAs compete with miRNAs for binding sites and contribute to the onset and progression of human malignancies. In this study we investigate how endosulfan promotes cell migration and invasion in DU145 and PC3 prostate cancer cells through epigenetic mechanism of lncRNA-miRNA regulation. Based on our past research we focused on PTP4A3 and constructed wild-type (WT) and mutant PTP4A3 plasmids for further analysis. Our results revealed that transfection of PTP4A3-WT can lead to changes in the expression of epithelial-mesenchymal transition (EMT) biomarkers and critical proteins in the TGF-β signaling pathway, and promote cell migration and invasion in PCa cells. Bioinformatics analysis shows that there were complementary sequences in PTP4A3 3'-UTR and KCNQ1OT1 3'-UTR to the seed sequence of hsa-miR-137-3p, and dual luciferase reporter assay indicates the potential binding capacity of miR-137-3p to 3'-UTR of PTP4A3 and KCNQ1OT1. We found that miR-137-3p mimic inhibited cell migration and invasion, as well as repressed alterations of EMT biomarkers and critical proteins in the TGF-β signaling pathway. Rescue experiment results revealed that co-transfection of miR-137-3p mimic and PTP4A3-WT plasmid reversed these changes following transfection with miR-137-3p mimic alone. We found that KCNQ1OT1 was predominantly distributed in the cytoplasm from a subcellular fractionation assay. Functionally, silencing of KCNQ1OT1 repressed cell migration and invasion, and caused alterations of EMT biomarkers and critical proteins in the TGF-β signaling pathway, which were all restored by co-transfection with anti-miR-137-3p or PTP4A3-WT plasmid. Furthermore, overexpression of miR-137-3p or silencing of KCNQ1OT1 dramatically rescued the effects of endosulfan on promoting cell migration and invasion. These findings suggest that endosulfan can indeed promote cell migration and invasion via the KCNQ1OT1/miR-137-3p/PTP4A3 axis in PCa cells.
Collapse
Affiliation(s)
- Yue Wang
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China
| | - Yubing Guo
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China
| | - Yanyuan Lu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China
| | - Yeqing Sun
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China
| | - Dan Xu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China.
| |
Collapse
|
4
|
Gjorgjieva M, Ay AS, Correia de Sousa M, Delangre E, Dolicka D, Sobolewski C, Maeder C, Fournier M, Sempoux C, Foti M. MiR-22 Deficiency Fosters Hepatocellular Carcinoma Development in Fatty Liver. Cells 2022; 11:cells11182860. [PMID: 36139435 PMCID: PMC9496902 DOI: 10.3390/cells11182860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/24/2022] Open
Abstract
MiR-22 is mostly considered as a hepatic tumor-suppressor microRNA based on in vitro analyses. Yet, whether miR-22 exerts a tumor-suppressive function in the liver has not been investigated in vivo. Herein, in silico analyses of miR-22 expression were performed in hepatocellular carcinomas from human patient cohorts and different mouse models. Diethylnitrosamine-induced hepatocellular carcinomas were then investigated in lean and diet-induced obese miR-22-deficient mice. The proteome of liver tissues from miR-22-deficient mice prior to hepatocellular carcinoma development was further analyzed to uncover miR-22 regulated factors that impact hepatocarcinogenesis with miR-22 deficiency. MiR-22 downregulation was consistently observed in hepatocellular carcinomas from all human cohorts and mouse models investigated. The time of appearance of the first tumors was decreased and the number of tumoral foci induced by diethylnitrosamine was significantly increased by miR-22-deficiency in vivo, two features which were further drastically exacerbated with diet-induced obesity. At the molecular level, we provide evidence that the loss of miR-22 significantly affects the energetic metabolism and mitochondrial functions of hepatocytes, and the expression of tumor-promoting factors such as thrombospondin-1. Our study demonstrates that miR-22 acts as a hepatic tumor suppressor in vivo by restraining pro-carcinogenic metabolic deregulations through pleiotropic mechanisms and the overexpression of relevant oncogenes.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Anne-Sophie Ay
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Marta Correia de Sousa
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Etienne Delangre
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Dobrochna Dolicka
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christine Maeder
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Margot Fournier
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christine Sempoux
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Correspondence:
| |
Collapse
|
5
|
Tang G, Yu C, Xiang K, Gao M, Liu Z, Yang B, Yang M, Zhao S. Inhibition of ANXA2 regulated by SRF attenuates the development of severe acute pancreatitis by inhibiting the NF-κB signaling pathway. Inflamm Res 2022; 71:1067-1078. [PMID: 35900381 DOI: 10.1007/s00011-022-01609-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is an inflammatory process of the pancreas resulting from biliary obstruction or alcohol consumption. Approximately, 10-20% of AP can evolve into severe AP (SAP). In this study, we sought to explore the physiological roles of the transcription factor serum response factor (SRF), annexin A2 (ANXA2), and nuclear factor-kappaB (NF-κB) in SAP. METHODS C57BL/6 mice and rat pancreatic acinar cells (AR42J) were used to establish an AP model in vivo and in vitro by cerulein with or without lipopolysaccharide (LPS). Production of pro-inflammatory cytokines (IL-1β and TNF-α) were examined by ELISA and immunoblotting analysis. Hematoxylin and eosin (HE) staining and TUNEL staining were performed to evaluate pathological changes in the course of AP. Apoptosis was examined by flow cytometric and immunoblotting analysis. Molecular interactions were tested by dual luciferase reporter, ChIP, and Co-IP assays. RESULTS ANXA2 was overexpressed in AP and correlated to the severity of AP. ANXA2 knockdown rescued pancreatic acinar cells against inflammation and apoptosis induced by cerulein with or without LPS. Mechanistic investigations revealed that SRF bound with the ANXA2 promoter region and repressed its expression. ANXA2 could activate the NF-κB signaling pathway by inducing the nuclear translocation of p50. SRF-mediated transcriptional repression of ANXA2-protected pancreatic acinar cells against AP-like injury through repressing the NF-κB signaling pathway. CONCLUSION Our study highlighted a regulatory network consisting of SRF, ANXA2, and NF-κB that was involved in AP progression, possibly providing some novel targets for treating SAP.
Collapse
Affiliation(s)
- Guanxiu Tang
- The Department of Gerontology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Can Yu
- The Department of Intensive Care Unit (ICU), The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Kaimin Xiang
- The Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Min Gao
- The Department of Intensive Care Unit (ICU), The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Zuoliang Liu
- The Department of Intensive Care Unit (ICU), The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Bingchang Yang
- The Department of Intensive Care Unit (ICU), The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Mingshi Yang
- The Department of Intensive Care Unit (ICU), The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Shangping Zhao
- The Department of Intensive Care Unit (ICU), The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China.
| |
Collapse
|
6
|
Valencia-Quintana R, Bahena-Ocampo IU, González-Castañeda G, Bonilla E, Milić M, Bonassi S, Sánchez-Alarcón J. miRNAs: A potentially valuable tool in pesticide toxicology assessment-current experimental and epidemiological data review. CHEMOSPHERE 2022; 295:133792. [PMID: 35104543 DOI: 10.1016/j.chemosphere.2022.133792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
miRNAs are responsible for the regulation of many cellular processes such as development, cell differentiation, proliferation, apoptosis, and tumor growth. Several studies showed that they can also serve as specific, stable, and sensitive markers of chemical exposure. In this review, current experimental and epidemiological data evidencing deregulation in miRNA expression in response to fungicides, insecticides or herbicides were analyzed. As shown by Venn's diagrams, miR-363 and miR-9 deregulation is associated with fungicide exposure in vitro and in vivo, while let-7, miR-155, miR-181 and miR-21 were found to be commonly deregulated by at least three different insecticides. Furthermore, let-7, miR-30, miR-126, miR-181 and miR-320 were commonly deregulated by 3 different herbicides. Notably, these 5 miRNAs were also found to be deregulated by one or more insecticides, suggesting their participation in the cellular response to pesticides, regardless of their chemical structure. All these miRNAs have been proposed as potential biomarkers for fungicide, insecticide, or herbicide exposure. These results allow us to improve our understanding of the molecular mechanisms of toxicity upon pesticide exposure, although further studies are needed to confirm these miRNAs as definitive (not potential) biomarkers of pesticide exposure.
Collapse
Affiliation(s)
- Rafael Valencia-Quintana
- Laboratorio "Rafael Villalobos-Pietrini" de Toxicología Genómica y Química Ambiental, Facultad de Agrobiología, Universidad Autónoma de Tlaxcala, CA Ambiente y Genética UATLX-CA-223 Red Temática de Toxicología de Plaguicidas, Tlaxcala, 90000, Mexico.
| | | | | | - Edmundo Bonilla
- Departamento de Ciencias de La Salud, UAM-Iztapalapa, Mexico.
| | - Mirta Milić
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, 10000, Croatia.
| | - Stefano Bonassi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, 00166, Italy; Unit of Clinical and Molecular Epidemiology IRCCS San Raffaele Pisana, Rome, 00166, Italy.
| | - Juana Sánchez-Alarcón
- Laboratorio "Rafael Villalobos-Pietrini" de Toxicología Genómica y Química Ambiental, Facultad de Agrobiología, Universidad Autónoma de Tlaxcala, CA Ambiente y Genética UATLX-CA-223 Red Temática de Toxicología de Plaguicidas, Tlaxcala, 90000, Mexico.
| |
Collapse
|
7
|
Lozano-Velasco E, Garcia-Padilla C, del Mar Muñoz-Gallardo M, Martinez-Amaro FJ, Caño-Carrillo S, Castillo-Casas JM, Sanchez-Fernandez C, Aranega AE, Franco D. Post-Transcriptional Regulation of Molecular Determinants during Cardiogenesis. Int J Mol Sci 2022; 23:ijms23052839. [PMID: 35269981 PMCID: PMC8911333 DOI: 10.3390/ijms23052839] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/26/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular development is initiated soon after gastrulation as bilateral precardiac mesoderm is progressively symmetrically determined at both sides of the developing embryo. The precardiac mesoderm subsequently fused at the embryonic midline constituting an embryonic linear heart tube. As development progress, the embryonic heart displays the first sign of left-right asymmetric morphology by the invariably rightward looping of the initial heart tube and prospective embryonic ventricular and atrial chambers emerged. As cardiac development progresses, the atrial and ventricular chambers enlarged and distinct left and right compartments emerge as consequence of the formation of the interatrial and interventricular septa, respectively. The last steps of cardiac morphogenesis are represented by the completion of atrial and ventricular septation, resulting in the configuration of a double circuitry with distinct systemic and pulmonary chambers, each of them with distinct inlets and outlets connections. Over the last decade, our understanding of the contribution of multiple growth factor signaling cascades such as Tgf-beta, Bmp and Wnt signaling as well as of transcriptional regulators to cardiac morphogenesis have greatly enlarged. Recently, a novel layer of complexity has emerged with the discovery of non-coding RNAs, particularly microRNAs and lncRNAs. Herein, we provide a state-of-the-art review of the contribution of non-coding RNAs during cardiac development. microRNAs and lncRNAs have been reported to functional modulate all stages of cardiac morphogenesis, spanning from lateral plate mesoderm formation to outflow tract septation, by modulating major growth factor signaling pathways as well as those transcriptional regulators involved in cardiac development.
Collapse
Affiliation(s)
- Estefania Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Carlos Garcia-Padilla
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Department of Anatomy, Embryology and Zoology, School of Medicine, University of Extremadura, 06006 Badajoz, Spain
| | - Maria del Mar Muñoz-Gallardo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Francisco Jose Martinez-Amaro
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
| | - Cristina Sanchez-Fernandez
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Amelia E. Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (C.G.-P.); (M.d.M.M.-G.); (F.J.M.-A.); (S.C.-C.); (J.M.C.-C.); (C.S.-F.); (A.E.A.)
- Fundación Medina, 18007 Granada, Spain
- Correspondence:
| |
Collapse
|
8
|
Kalinina T, Kononchuk V, Klyushova L, Gulyaeva L. Effects of Endocrine Disruptors o, p'-Dichlorodiphenyltrichloroethane, p, p'-Dichlorodiphenyltrichloroethane, and Endosulfan on the Expression of Estradiol-, Progesterone-, and Testosterone-Responsive MicroRNAs and Their Target Genes in MCF-7 Cells. TOXICS 2022; 10:25. [PMID: 35051067 PMCID: PMC8780485 DOI: 10.3390/toxics10010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 12/10/2022]
Abstract
Many studies have shown that dichlorodiphenyltrichloroethane (DDT) exposure raises breast cancer risk. Another insecticide with similar properties is endosulfan, which has been actively used in agriculture after DDT prohibition. Previously, we have identified some estradiol-, progesterone-, and testosterone-sensitive microRNAs (miRNAs, miRs). Because DDT and endosulfan have estrogenic, antiandrogenic, and antiprogesterone properties, we hypothesized that these miRNAs are affected by the insecticides. We quantified relative levels of miRNAs and expression levels of their target genes in breast cancer MCF-7 cells treated with p,p'-DDT, o,p'-DDT, or endosulfan. We also quantified miR-19b expression, which, as previously shown, is regulated by estrogen. Here, we observed that miR-19b expression increased in response not only to estradiol but also to testosterone and progesterone. Treatment of MCF-7 cells with p,p'-DDT or endosulfan decreased the protein levels of apoptosis regulators TP53INP1 and APAF1. In cells treated with o,p'-DDT, the TP53INP1 amount decreased after 24 h of incubation, but increased after 48 h of incubation with insecticide. OXTR expression, which is known to be associated with breast carcinogenesis, significantly diminished under the exposure of all insecticides. In cells treated with p,p'-DDT or o,p'-DDT, the observed changes were accompanied by alterations of the levels of hormone-responsive miRNAs: miR-324, miR-190a, miR-190b, miR-27a, miR-193b, and miR-19b.
Collapse
Affiliation(s)
- Tatiana Kalinina
- Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2/12, 630117 Novosibirsk, Russia; (V.K.); (L.K.); (L.G.)
| | - Vladislav Kononchuk
- Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2/12, 630117 Novosibirsk, Russia; (V.K.); (L.K.); (L.G.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Rechkunovskaya Str. 15, 630055 Novosibirsk, Russia
| | - Lyubov Klyushova
- Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2/12, 630117 Novosibirsk, Russia; (V.K.); (L.K.); (L.G.)
| | - Lyudmila Gulyaeva
- Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2/12, 630117 Novosibirsk, Russia; (V.K.); (L.K.); (L.G.)
- Institute for Medicine and Psychology, Novosibirsk State University, Pirogova Str. 2, 630090 Novosibirsk, Russia
| |
Collapse
|
9
|
Zhou X, Li S, Chao D, Chen Z, Zhang J, Lin J, Ji Y, Ji Q. Serum small extracellular vesicles promote M1 activation of microglia after cerebral ischemia/reperfusion injury. Neurosci Lett 2021; 766:136307. [PMID: 34737022 DOI: 10.1016/j.neulet.2021.136307] [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: 04/21/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
Microglial M1 activation is detrimental to stroke outcomes. Recent studies have shown that circulating small extracellular vesicles (sEVs) can deliver miRNAs to target cells and regulate recipient cell functions. Herein, we tested the hypothesis that miRNA delivery by serum sEVs after cerebral ischemia/reperfusion (I/R) injury promote microglial M1 activation, demonstrating that serum sEVs from middle cerebral artery occlusion (MCAO) mice promoted proliferation and M1 activation of BV2 microglia. To explore the underlying mechanism of serum sEVs-mediated microglial activation in the early phase of cerebral I/R injury, we examined the effects of ischemic brain injury on the serum sEVs miRNAs profile in a mouse MCAO model using small RNAseq. Of the 1257 detected miRNA replications, the levels of 72 were significantly modulated. Bioinformatics analysis revealed that a panel of miRNAs was closely associated with inflammation, and in vitro experiments demonstrated that serum sEVs from MCAO mice could effectively transfer inflammatory miRNAs to BV2 microglia. Collectively, our data suggested that miRNAs delivered by serum sEVs after cerebral I/R injury promoted microglial M1 activation. The identification of microglial activation regulators in future studies will give rise to more effective treatments for stroke.
Collapse
Affiliation(s)
- Xin Zhou
- Institute of Immunology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
| | - Shuyuan Li
- Institute of Immunology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dachong Chao
- Institute of Immunology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zixin Chen
- Institute of Immunology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Junyu Zhang
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Jianhang Lin
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Yuhua Ji
- Institute of Immunology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Qiuhong Ji
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226019, China.
| |
Collapse
|
10
|
The Cohesin Complex and Its Interplay with Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040067. [PMID: 34707078 PMCID: PMC8552073 DOI: 10.3390/ncrna7040067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
The cohesin complex is a multi-subunit protein complex initially discovered for its role in sister chromatid cohesion. However, cohesin also has several other functions and plays important roles in transcriptional regulation, DNA double strand break repair, and chromosome architecture thereby influencing gene expression and development in organisms from yeast to man. While most of these functions rely on protein–protein interactions, post-translational protein, as well as DNA modifications, non-coding RNAs are emerging as additional players that facilitate and modulate the function or expression of cohesin and its individual components. This review provides a condensed overview about the architecture as well as the function of the cohesin complex and highlights its multifaceted interplay with both short and long non-coding RNAs.
Collapse
|
11
|
Wei F, Ren B, Han W, Guan H, Jing G, Wang M. Investigate the Effect of miR-22 on the Apoptosis of Coronary Heart Disease Cells Through the Wnt-1 Pathway Based on Nano-Silica-Induced Rat Models. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:1338-1344. [PMID: 33183481 DOI: 10.1166/jnn.2021.18637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this paper, by examining the toxicity of nano-silica to coronary heart disease cells, we explored the apoptosis of rat myocardial cells induced by nano-silica, and explored the effect of apoptosis on cells during the process of myocardial cytotoxicity induced by nano-silica. This article selects rat cardiomyocytes as the research object and conducts a group control experiment. A control group is set up with cells that are not stained with nano-silica. Different concentrations of nanosilica suspensions are applied to rat cells and detected by CCK-8 method. Cell survival rate after exposure to different concentrations of cells is used to determine the most stable exposure time and concentration. We used flow cytometry to detect intracellular reactive oxygen species and apoptotic rates, and used Western Blot to detect the expression of proteins that affect apoptosis. Finally, we investigated the effect of the Wnt signaling pathway on coronary heart disease. The Wnt signaling pathway regulates the development of the heart and blood vessels. In the treatment of cardiovascular disease, this pathway will be activated again to play a regulatory role. We conclude that nano-silica can induce cytotoxicity in rat myocardial cells through the Wnt-1 pathway, and nanosilica can induce myocardial cell apoptosis through the Wnt-1 pathway.
Collapse
Affiliation(s)
- Fangjing Wei
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Baojun Ren
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Wei Han
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Hong Guan
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Guoqiang Jing
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Min Wang
- Department of Cardiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| |
Collapse
|
12
|
The tissue specific regulation of miR22 expression in the lung and brain by ribosomal protein L29. Sci Rep 2020; 10:16242. [PMID: 33004906 PMCID: PMC7530758 DOI: 10.1038/s41598-020-73281-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 09/15/2020] [Indexed: 11/23/2022] Open
Abstract
Endogenous miR22 is associated with a diverse range of biological processes through post-translational modification of gene expression and its deregulation results in various diseases including cancer. Its expression is usually tissue or cell-specific, however, the reasons behind this tissue or cell specificity are not clearly outlined till-date. Therefore, our keen interest was to investigate the mechanisms of tissue or cell-specific expression of miR22. In the current study, miR22 expression showed a tissues-specific difference in the poly(I:C) induced inflammatory mouse lung and brain tissues. The cell-specific different expression of miR22 was also observed in inflammatory glial cells and endothelial cells. The pattern of RPL29 expression was also similar to miR22 in these tissues and cells under the same treatment. Interestingly, the knockdown of RPL29 exerted an inhibitory effect on miR22 and its known transcription factors including Fos-B and c-Fos. Fos-B and c-Fos were also differentially expressed in the two cell lines transfected with poly(I:C). The knockdown of c-Fos also exerted its negative effects on miR22 expression in both cells. These findings suggest that RPL29 might have regulatory roles on tissue or cell-specific expression of miR22 through the transcription activities of c-Fos and also possibly through Fos-B.
Collapse
|
13
|
Li S, Yang Y, Shi MH, Wang JF, Ran XQ. miR-96-5p attenuates malathion-induced apoptosis of human kidney cells by targeting the ER stress marker DDIT3. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2020; 55:1080-1086. [PMID: 32897819 DOI: 10.1080/03601234.2020.1816092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Micro RNAs (miRNAs) are major players in cellular responses to xenobiotic compounds and toxins. However, their functions in organophosphate-induced cytotoxicity remain unclear. This study investigated the involvement of miR-96-5p in the non-cholinergic toxicity of malathion in normal human kidney cells (HK-2 cells). Malathion decreased HK-2 cell viability and the expression of miR-96-5p in a dose- and time-dependent manner. In addition, transfection with miR-96-5p mimics attenuated malathion-induced HK-2 cell apoptosis, whereas transfection with a miR-96-5p inhibitor increased HK-2 cell apoptosis. Luciferase assays indicated that miR-96-5p could bind directly to the 3'-untranslated region of DDIT3, a well-known marker of endoplasmic reticulum stress. Further analyses of the expression of apoptosis-related genes and proteins indicated that miR-96-5p may function to reduce malathion-induced HK-2 cell apoptosis via regulation of the DDIT3/B-cell lymphoma (BCL)-2/caspase-3 signaling pathway. In summary, the results of the present study indicate that miR-96-5p protects HK-2 cells from malathion-induced ER stress-dependent apoptosis by targeting DDIT3.
Collapse
Affiliation(s)
- Sheng Li
- The Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education)/Guizhou Key Lab of Agro-Bioengineering, Institute of Agro-Bioengineering and College of Life Sciences, Guizhou University, Guiyang, China
| | - Yang Yang
- The Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education)/Guizhou Key Lab of Agro-Bioengineering, Institute of Agro-Bioengineering and College of Life Sciences, Guizhou University, Guiyang, China
| | - Ming Hui Shi
- The Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education)/Guizhou Key Lab of Agro-Bioengineering, Institute of Agro-Bioengineering and College of Life Sciences, Guizhou University, Guiyang, China
| | - Jia Fu Wang
- The Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education)/Guizhou Key Lab of Agro-Bioengineering, Institute of Agro-Bioengineering and College of Life Sciences, Guizhou University, Guiyang, China
| | - Xue Qin Ran
- Faculty of Animal Science and Veterinary Medicine, Guizhou University, Guiyang, China
| |
Collapse
|
14
|
Wei J, Liu J, Zhang L, Zhu Y, Li X, Zhou G, Zhao Y, Sun Z, Zhou X. Endosulfan induces cardiotoxicity through apoptosis via unbalance of pro-survival and mitochondrial-mediated apoptotic pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 727:138790. [PMID: 32344260 DOI: 10.1016/j.scitotenv.2020.138790] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/31/2020] [Accepted: 04/16/2020] [Indexed: 06/11/2023]
Abstract
Although the associations between endosulfan and adverse cardiovascular health have been reported, the toxic effects and underlying mechanism of endosulfan on the heart are not well understood. In this study, we examined the cardiotoxicity induced by endosulfan using Wistar rats and human cardiomyocytes (AC16) cells. Wistar rats were divided into control group (received corn oil alone) and three concentrations of endosulfan groups (1, 5 and 10 mg/kg·bw) by gavage. The AC16 cells were treated with three various concentrations (0, 1.25, 5, and 20 μg/mL) of endosulfan. The results showed that endosulfan induced cytotoxicity through damaging myocardial structure, decreasing the viability of cardiomyocytes, and elevating the serum levels of cardiac troponin I, heart fatty acid binding protein, aspartate aminotransferase, and reactive oxygen species (p < 0.05). Moreover, measurement of mitochondrial function showed that endosulfan could significantly decrease adenosine triphosphate levels and cytochrome c oxidase IV expression in AC16 cells (p < 0.05). In addition, endosulfan obviously inhibited Bcl-2 expression, activated the expressions of cytochrome c/Caspase-9/Caspase-3 signaling pathway, and induced the apoptosis of AC16 cells (p < 0.05). Furthermore, endosulfan significantly increased the expression of Bim, and inhibited the expressions of PI3K/Akt/FoxO3a signaling pathways in cardiomyocytes (p < 0.05). These results suggest that endosulfan may induce cardiotoxicity by inducing myocardial apoptosis resulting from activation of mitochondria-mediated apoptosis pathway and inhibition of pro-survival signaling pathways, which might be helpful in elucidating the mechanism of cardiac dysfunction induced by endosulfan.
Collapse
Affiliation(s)
- Jialiu Wei
- Key Laboratory of Cardiovascular Epidemiology & Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Jianhui Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Lianshuang Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yupeng Zhu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiangyang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Guiqing Zhou
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yanzhi Zhao
- Yanjing Medical College, Capital Medical University, Beijing, China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xianqing Zhou
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Jing QB, Tong HX, Tang WJ, Tian SD. Clinical Significance and Potential Regulatory Mechanisms of Serum Response Factor in 1118 Cases of Thyroid Cancer Based on Gene Chip and RNA-Sequencing Data. Med Sci Monit 2020; 26:e919302. [PMID: 31967986 PMCID: PMC6995247 DOI: 10.12659/msm.919302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Thyroid cancer (TC) is one of the most prevalent endocrine malignancies and there may be many unclarified molecular events and gene types involved in TC. The objective of this study was to assess the clinical implications and potential mechanisms of serum response factor (SRF) in TC. Material/Methods RNA-sequencing and gene chip data with TC expression were collected from The Cancer Genome Atlas/Genotype-Tissue Expression, Gene Expression Omnibus, ArrayExpress, Sequence Read Archive, and Oncomine. SRF expression of all TC and adjacent non-cancerous tissue were calculated using the t test, STATA, and Meta-DiSc. The related pathways of the potential SRF target genes and target miRNAs were explored. Dual-luciferase reporter assay was performed to validate the association between SRF and its putative miRNA. Results One RNA-sequencing and 15 gene chips were collected, and the pooled standardized mean difference of SRF was −1.00. Furthermore, the area under the curve of sROC of SRF in TC was 0.8251, indicating a dramatic decreased expression of SRF in TC tissues based on 1118 cases. The intersection of differentially expressed genes in TC, SRF co-expressed genes, and SRF potential target genes achieved from Cistrome Cancer led to 169 overlapped genes. miR-330-5p was predicted to target SRF, which was further confirmed by dual-luciferase reporter assay. Conclusions The reduction of SRF appears to play a crucial role in the origin of TC. These properties are accomplished by the target genes of SRF, as a transcription factor, or by the axes with the associated miRNAs.
Collapse
Affiliation(s)
- Qiang-Bin Jing
- Center of Medical Oncology, The First People's Hospital of Huaihua, Huaihua, Hunan, China (mainland)
| | - Hai-Xiao Tong
- Center of Medical Oncology, The First People's Hospital of Huaihua, Huaihua, Hunan, China (mainland)
| | - Wei-Jian Tang
- Center of Medical Oncology, The First People's Hospital of Huaihua, Huaihua, Hunan, China (mainland)
| | - Shao-Dong Tian
- Center of Medical Oncology, The First People's Hospital of Huaihua, Huaihua, Hunan, China (mainland)
| |
Collapse
|
16
|
Qiao J, Du Y, Yu J, Guo J. MicroRNAs as Potential Biomarkers of Insecticide Exposure: A Review. Chem Res Toxicol 2019; 32:2169-2181. [PMID: 31625722 DOI: 10.1021/acs.chemrestox.9b00236] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Insecticides are key weapons for the control of pests. Large scale use of insecticides is harmful to the ecosystem, which is made up of a wide range of species and environments. MicroRNAs (miRNAs) are a class of endogenous single-stranded noncoding small RNAs in length of 20-24 nucleotides (nt), which extensively regulate expression of genes at transcriptional and post-transcriptional levels. The current research on miRNA-induced insecticide resistance reveals that dysregulated miRNAs cause significant changes in detoxification genes, particularly cytochrome P450s. Meanwhile, insecticide-induced changes in miRNAs are related to the decline of honeybees and threatened the development of zebrafish and other animals. Additionally, miRNAs are involved in insecticide-induced cytotoxicity, and dysregulated miRNAs are associated with human occupational and environmental exposure to insecticides. Therefore, miRNAs are valuable novel biomarkers of insecticide exposure, and they are potential factors to explain the toxicological effects of insecticides.
Collapse
Affiliation(s)
- Jiakai Qiao
- College of Life Sciences and Medicine , Zhejiang Sci-Tech University , Hangzhou , Zhejiang 310018 , China
| | - Yuting Du
- College of Life Sciences and Medicine , Zhejiang Sci-Tech University , Hangzhou , Zhejiang 310018 , China
| | - Junjie Yu
- College of Life Sciences and Medicine , Zhejiang Sci-Tech University , Hangzhou , Zhejiang 310018 , China
| | - Jiangfeng Guo
- College of Life Sciences and Medicine , Zhejiang Sci-Tech University , Hangzhou , Zhejiang 310018 , China
| |
Collapse
|
17
|
Müller S, Glaß M, Singh AK, Haase J, Bley N, Fuchs T, Lederer M, Dahl A, Huang H, Chen J, Posern G, Hüttelmaier S. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res 2019; 47:375-390. [PMID: 30371874 PMCID: PMC6326824 DOI: 10.1093/nar/gky1012] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/09/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022] Open
Abstract
The oncofetal mRNA-binding protein IGF2BP1 and the transcriptional regulator SRF modulate gene expression in cancer. In cancer cells, we demonstrate that IGF2BP1 promotes the expression of SRF in a conserved and N6-methyladenosine (m6A)-dependent manner by impairing the miRNA-directed decay of the SRF mRNA. This results in enhanced SRF-dependent transcriptional activity and promotes tumor cell growth and invasion. At the post-transcriptional level, IGF2BP1 sustains the expression of various SRF-target genes. The majority of these SRF/IGF2BP1-enhanced genes, including PDLIM7 and FOXK1, show conserved upregulation with SRF and IGF2BP1 synthesis in cancer. PDLIM7 and FOXK1 promote tumor cell growth and were reported to enhance cell invasion. Consistently, 35 SRF/IGF2BP1-dependent genes showing conserved association with SRF and IGF2BP1 expression indicate a poor overall survival probability in ovarian, liver and lung cancer. In conclusion, these findings identify the SRF/IGF2BP1-, miRNome- and m6A-dependent control of gene expression as a conserved oncogenic driver network in cancer.
Collapse
Affiliation(s)
- Simon Müller
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Anurag K Singh
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| | - Jacob Haase
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Nadine Bley
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Tommy Fuchs
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Marcell Lederer
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Andreas Dahl
- Deep Sequencing Group, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden
| | - Huilin Huang
- Department of Systems Biology, City of Hope, Monrovia, CA 91016, USA
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45219, USA
| | - Jianjun Chen
- Department of Systems Biology, City of Hope, Monrovia, CA 91016, USA
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45219, USA
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford protein center, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| |
Collapse
|
18
|
Xu D, Guo YB, Zhang M, Sun YQ. The subsequent biological effects of simulated microgravity on endothelial cell growth in HUVECs. Chin J Traumatol 2018; 21:229-237. [PMID: 30017544 PMCID: PMC6085276 DOI: 10.1016/j.cjtee.2018.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/17/2018] [Accepted: 02/28/2018] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Microgravity is known to cause endothelium dysfunction in astronauts returning from spaceflight. We aimed to reveal the regulatory mechanism in alterations of human endothelial cells after simulated microgravity (SMG). METHODS We utilized the rotary cell culture system (RCCS-1) to explore the subsequent effects of SMG on human umbilical vein endothelial cells (HUVECs). RESULTS SMG-treated HUVECs appeared obvious growth inhibition after return to normal gravity, which might be attributed to a set of responses including alteration of cytoskeleton, decreased cell adhesion capacity and increased apoptosis. Expression levels of mTOR and its downstream Apaf-1 were increased during subsequent culturing after SMG. miR-22 was up-regulated and its target genes SRF and LAMC1 were down-regulated at mRNA levels. LAMC1 siRNAs reduced cell adhesion rate and inhibited stress fiber formation while SRF siRNAs caused apoptosis. CONCLUSION SMG has the subsequent biological effects on HUVECs, resulting in growth inhibition through mTOR signaling and miR-22-mediated mechanism.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Environmental Systems Biology, Dalian Maritime University, Dalian 116026, China
| | - Yu-Bing Guo
- Institute of Environmental Systems Biology, Dalian Maritime University, Dalian 116026, China
| | - Min Zhang
- Institute of Environmental Systems Biology, Dalian Maritime University, Dalian 116026, China,Department of Molecular Physiology and Medical Bioregulation, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Ye-Qing Sun
- Institute of Environmental Systems Biology, Dalian Maritime University, Dalian 116026, China,Corresponding author.
| |
Collapse
|
19
|
Su Z, Zhang M, Xu M, Li X, Tan J, Xu Y, Pan X, Chen N, Chen X, Zhou Q. MicroRNA181c inhibits prostate cancer cell growth and invasion by targeting multiple ERK signaling pathway components. Prostate 2018; 78:343-352. [PMID: 29341215 DOI: 10.1002/pros.23478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/13/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND The ERK signaling pathway is frequently deregulated in tumorigenesis, mostly by classical mechanisms such as gene mutation of its components (eg, RAS and RAF). However, whether and how multiple key components of ERK pathway are regulated by microRNAs are not clear. METHODS We firstly predicted post-transcriptional regulation of multiple key components of the ERK signaling pathway by miR181c through bioinformatics analysis, and then confirmed the post-transcriptional regulation by dual luciferase reporter gene assays and Western blot analysis. The biological effects of miR181c on prostate cancer cell proliferation, apoptosis, migration, and invasion were measured by CCK-8 assay, flow cytometry, wound scratch assay, transwell cell migration, and invasion assays. RESULTS miR181c post-transcriptionally regulated multiple key members of the ERK signaling pathway, including extracellular signal-regulated kinase 2 (ERK2), ribosomal S6 kinase 2 (RSK2), serum response factor (SRF), and FBJ murine osteosarcoma viral oncogene homolog (c-Fos). Ectopic expression of miR181c mimics effectively suppressed prostate cancer cell proliferation, migration, and invasion, but promoted cell apoptosis. Furthermore, miR181c treatment combined with the multi-kinase inhibitor sorafenib significantly enhanced these anti-tumor effects. CONCLUSIONS Downregulation of miR181c results in deregulated ERK signaling and promotes prostate cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Zhengzheng Su
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Miao Xu
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xinglan Li
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Junya Tan
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yunyi Xu
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xiuyi Pan
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xueqin Chen
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|