1
|
Chen T, Zhang B, He G, Wang N, Cao M, Shen C, Chen X, Chen L, Liu K, Luo Y, huang Y, Yuan C, Zhou X, Li C. Gut-Derived Exosomes Mediate the Microbiota Dysbiosis-Induced Spermatogenesis Impairment by Targeting Meioc in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310110. [PMID: 38526201 PMCID: PMC11165515 DOI: 10.1002/advs.202310110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/05/2024] [Indexed: 03/26/2024]
Abstract
Diseases like obesity and intestinal inflammation diseases are accompanied by dysbiosis of the gut microbiota (DSGM), which leads to various complications, including systemic metabolic disorders. DSGM reportedly impairs the fertility of male mice; however, the regulatory mechanism is unclear. Exosomes are molecular mediators of intercellular communication, but the regulation of spermatogenesis by non-reproductive tissue-originated exosomes remains unknown. The present study shows that DSGM altered the miRNA expression profile of mouse circulating exosomes and impaired spermatogenesis. Moreover, the single-cell sequencing results indicate that circulating exosomes from mice with DSGM impaired spermatogenesis, while circulating exosomes from wild mice improved spermatogenesis by promoting meiosis. Further study demonstrates that DSGM leads to abnormal upregulation of miR-211-5p in gut-derived circulating exosomes, which inhibited the expression of meiosis-specific with coiled-coil domain (Meioc) in the testes and impaired spermatogenesis by disturbing meiosis process. In summary, this study defines the important role of gut-derived exosomes in connecting the "gut-testis" axis.
Collapse
Affiliation(s)
- Tong Chen
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Boqi Zhang
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Guitian He
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Nan Wang
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Maosheng Cao
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Caomeihui Shen
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Xue Chen
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Lu Chen
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Kening Liu
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Yuxin Luo
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Yiqiu huang
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Chenfeng Yuan
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Xu Zhou
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| | - Chunjin Li
- College of Animal SciencesJilin University5333 Xian RoadChangchunJilin130062China
| |
Collapse
|
2
|
Cheng K, Pan J, Liu Q, Ji Y, Liu L, Guo X, Wang Q, Li S, Sun J, Gong M, Zhang Y, Yuan Y. Exosomal lncRNA XIST promotes perineural invasion of pancreatic cancer cells via miR-211-5p/GDNF. Oncogene 2024; 43:1341-1352. [PMID: 38454138 DOI: 10.1038/s41388-024-02994-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Perineural invasion (PNI) is an essential form of tumor metastasis in multiple malignant cancers, such as pancreatic cancer, prostate cancer, and head and neck cancer. Growing evidence has revealed that pancreatic cancer recurrence and neuropathic pain positively correlate with PNI. Therefore, targeting PNI is a proper strategy for pancreatic cancer treatment. Exosomal lncRNA derived from pancreatic cancer cells is an essential component of the tumor microenvironment. However, whether exosomal lncXIST derived from pancreatic cancer cells can promote PNI and its exact mechanism remains to be elucidated. We show that lncXIST mediates nerve-tumor crosstalk via exosomal delivery. Our data reveal that exosomal lncXIST derived from pancreatic cancer cells is delivered to neural cells and promotes their release of glial-cell-line-derived neurotrophic factor (GDNF), essential in facilitating the PNI of pancreatic cancer. Mechanistically, microRNA-211-5p negatively regulates GDNF, and lncXIST serves as a miR-211-5p sponge. The function of exosomes in the dynamic interplay between nerves and cancer is confirmed in both in vivo and in vitro PNI models. Therefore, targeting pancreatic cancer cell-derived exosomal lncXIST may provide clues for a promising approach for developing a new strategy to combat PNI of pancreatic cancer.
Collapse
Affiliation(s)
- Ke Cheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Qinlong Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Yuke Ji
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Liang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Xiangqian Guo
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 47500, China
| | - Qiang Wang
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 47500, China
| | - Shao Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jinyue Sun
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Miaomiao Gong
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| | - Ying Zhang
- Sixth Department of liver disease, Dalian Public Health Clinical Center, Dalian, 116044, China.
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
3
|
Wang J, Gu S, Qin B. Eosinophil and mast cell-derived exosomes promote integrity of intestinal mucosa via the NEAT1/miR-211-5p/glial cell line-derived neurotrophic factor axis in duodenum. ENVIRONMENTAL TOXICOLOGY 2023; 38:2595-2607. [PMID: 37466184 DOI: 10.1002/tox.23895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/31/2023] [Accepted: 07/01/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Exosomes are applied as biomarkers in several diseases according to their disease-specific profiles. However, the exosomes effects in functional dyspepsia (FD) are still fragmentary. Here we examined the role of Eosinophil and mast cell derived-exosomes in FD progression. METHODS Fifty FD subjects and age- and sex-matched healthy controls were included in this retrospective cohort study. Duodenal mucosa and gastric juice were collected to analyze molecular difference. Eosinophil and mast cell were evaluated by immunofluorescence and microarray was subjected to examine the expression levels of NEAT1, miR-211-5p, and glial cell line-derived neurotrophic factor (GDNF), which were subsequently were tested by quantitative reverse transcription PCR (RT-qPCR) validation cohorts. CCK-8 assays, and wound healing assays were used to evaluate integrity of intestinal mucosal barrier in vitro. Rats' weights and gastric emptying rates were used as evaluation of FD severity in vivo. RESULTS Eosinophil and mast cell were enriched and secreted more exosomes in duodenal mucosa of FD patients. We identified differential lncRNAs that were consistently and significantly up regulated in FD cases. Of these, NEAT1 was further validated by RT-qPCR and had closely relationship with GDNF. MiR-211-5p level was found to be reduced in FD and negatively related with NEAT1 and GDNF. Furthermore, NEAT1and GDNF relived FD while miR-211-5p made symptoms worse. The NEAT1/miR-211-5p/GDNF axis had a good predictive ability for FD. CONCLUSIONS The NEAT1/miR-211-5p/GDNF could be a potential FD biomarker.
Collapse
Affiliation(s)
- Jue Wang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sai Gu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Qin
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Overexpression of microRNA-211 in Functional Dyspepsia via Downregulation of the Glial Cell Line-Derived Neurotrophic Factor (GDNF) by Increasing Phosphorylation of p38 MAPK Pathway. Can J Gastroenterol Hepatol 2022; 2022:9394381. [PMID: 36569394 PMCID: PMC9771656 DOI: 10.1155/2022/9394381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Overexpression of miRNA-211 suppresses the differentiation of bone marrow stem cells into intestinal ganglion cells via downregulation of GDNF, a regulator of intestine barrier function. The study aimed to investigate the interaction between miR-211 and GDNF on intestinal epithelial cells. METHODS The expression levels of miR-211 and GDNF in duodenal biopsy specimens from FD patients and healthy controls were compared. Enteric glia cell (EGCs) cell line transfected with miR-211 mimics and inhibitors were used to clarify the expression levels of GDNF were analyzed by qRT-PCR and ELISA. Intestine epithelial cell (IECs) cell line cultured in medium from ECGs in different transfection conditions were used in wound healing assay, cell proliferation assay, and western blotting for evaluation of p38 MAPK phosphorylation level. RESULTS MiR-211 expression was significantly upregulated in the duodenal tissue of patients with FD compared to healthy subjects, whereas GDNF expression was significantly downregulated (both p < 0.05). Transfection with miR-211 mimics significantly decreased GDNF mRNA expression and protein secretion (p < 0.001). An inhibited intestinal epithelial cell wound healing (p < 0.05) and increased expression levels of phosphorylated p38 MAPK (p < 0.05) were found in IECs cultured with medium from EGCs transfected with miR-211 mimics. CONCLUSIONS MiR-211 may downregulates GDNF mRNA and protein expression via activation of the pp38 MAPK signaling pathway. Targeting miR-211 or the MAPK pathway may be a potential intervention for FD.
Collapse
|
5
|
Gürünlüoğlu S, Ceran C, Gürünlüoğlu K, Koçbiyik A, Gül M, Yıldız T, Bağ HG, Gül S, Taşçi A, Bayrakçi E, Akpinar N, Çin ES, Ateş H, Demircan M. Glial Cell Line-Derived Neurotrophic Factor, S-100 Protein and Synaptophysin Expression in Biliary Atresia Gallbladder Tissue. Pediatr Gastroenterol Hepatol Nutr 2021; 24:173-186. [PMID: 33833973 PMCID: PMC8007845 DOI: 10.5223/pghn.2021.24.2.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 11/14/2022] Open
Abstract
PURPOSE Biliary atresia (BA) is a disease that manifests as jaundice after birth and leads to progressive destruction of the ductal system in the liver. The aim of this study was to investigate histopathological changes and immunohistochemically examine the expression of glial cell line-derived neurotrophic factor (GDNF), synaptophysin, and S-100 protein in the gallbladder of BA patients. METHODS The study included a BA group of 29 patients and a control group of 41 children with cholecystectomy. Gallbladder tissue removed during surgery was obtained and examined immunohistochemically and histopathologically. Tissue samples of both groups were immunohistochemically assessed in terms of GDNF, S-100 protein, and synaptophysin expression. Expression was classified as present or absent. Inflammatory activity assessment with hematoxylin and eosin staining and fibrosis assessment with Masson's trichrome staining were performed for tissue sample sections of both groups. RESULTS Ganglion cells were not present in gallbladder tissue samples of the BA group. Immunohistochemically, GDNF, synaptophysin, and S-100 expression was not detected in the BA group. Histopathological examination revealed more frequent fibrosis and slightly higher inflammatory activity in the BA than in the control group. CONCLUSION We speculate that GDNF expression will no longer continue in this region, when the damage caused by inflammation of the extrahepatic bile ducts reaches a critical threshold. The study's findings may represent a missing link in the chain of events forming the etiology of BA and may be helpful in its diagnosis.
Collapse
Affiliation(s)
- Semra Gürünlüoğlu
- Department of Pathology Malatya Education and Research Hospital, Pathology Laboratory, Malatya, Turkey
| | - Canan Ceran
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Kubilay Gürünlüoğlu
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Alper Koçbiyik
- Department of Pathology, Istanbul Bakırköy Dr Sadi Konuk Education and Research Hospital, Pathology Laboratory, Istanbul, Turkey
| | - Mehmet Gül
- Department of Histology and Embryology, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Turan Yıldız
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Harika Gözükara Bağ
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Semir Gül
- Department of Histology and Embryology, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Aytaç Taşçi
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Ercan Bayrakçi
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Necmettin Akpinar
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Ecem Serbest Çin
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Hasan Ateş
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Mehmet Demircan
- Department of Pediatric Surgery, Faculty of Medicine, İnönü University, Malatya, Turkey
| |
Collapse
|
6
|
Enterría-Morales D, Del Rey NLG, Blesa J, López-López I, Gallet S, Prévot V, López-Barneo J, d'Anglemont de Tassigny X. Molecular targets for endogenous glial cell line-derived neurotrophic factor modulation in striatal parvalbumin interneurons. Brain Commun 2020; 2:fcaa105. [PMID: 32954345 PMCID: PMC7472905 DOI: 10.1093/braincomms/fcaa105] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/05/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Administration of recombinant glial cell line-derived neurotrophic factor into the putamen has been tested in preclinical and clinical studies to evaluate its neuroprotective effects on the progressive dopaminergic neuronal degeneration that characterizes Parkinson’s disease. However, intracerebral glial cell line-derived neurotrophic factor infusion is a challenging therapeutic strategy, with numerous potential technical and medical limitations. Most of these limitations could be avoided if the production of endogenous glial cell line-derived neurotrophic factor could be increased. Glial cell line-derived neurotrophic factor is naturally produced in the striatum from where it exerts a trophic action on the nigrostriatal dopaminergic pathway. Most of striatal glial cell line-derived neurotrophic factor is synthesized by a subset of GABAergic interneurons characterized by the expression of parvalbumin. We sought to identify molecular targets specific to those neurons and which are putatively associated with glial cell line-derived neurotrophic factor synthesis. To this end, the transcriptomic differences between glial cell line-derived neurotrophic factor-positive parvalbumin neurons in the striatum and parvalbumin neurons located in the nearby cortex, which do not express glial cell line-derived neurotrophic factor, were analysed. Using mouse reporter models, we have defined the genomic signature of striatal parvalbumin interneurons obtained by fluorescence-activated cell sorting followed by microarray comparison. Short-listed genes were validated by additional histological and molecular analyses. These genes code for membrane receptors (Kit, Gpr83, Tacr1, Tacr3, Mc3r), cytosolic proteins (Pde3a, Crabp1, Rarres2, Moxd1) and a transcription factor (Lhx8). We also found the proto-oncogene cKit to be highly specific of parvalbumin interneurons in the non-human primate striatum, thus highlighting a conserved expression between species and suggesting that specific genes identified in mouse parvalbumin neurons could be putative targets in the human brain. Pharmacological stimulation of four G-protein-coupled receptors enriched in the striatal parvalbumin interneurons inhibited Gdnf expression presumably by decreasing cyclic adenosine monophosphate formation. Additional experiments with pharmacological modulators of adenylyl cyclase and protein kinase A indicated that this pathway is a relevant intracellular route to induce Gdnf gene activation. This preclinical study is an important step in the ongoing development of a specific pro-endo-glial cell line-derived neurotrophic factor pharmacological strategy to treat Parkinson’s disease.
Collapse
Affiliation(s)
- Daniel Enterría-Morales
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | | | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ivette López-López
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Sarah Gallet
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S 1172, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S 1172, Lille, France
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
7
|
Danbaran GR, Aslani S, Sharafkandi N, Hemmatzadeh M, Hosseinzadeh R, Azizi G, Jadidi-Niaragh F, Babaie F, Mohammadi H. How microRNAs affect the PD-L1 and its synthetic pathway in cancer. Int Immunopharmacol 2020; 84:106594. [PMID: 32416456 DOI: 10.1016/j.intimp.2020.106594] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/27/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022]
Abstract
Programmed cell death-ligand 1 (PD-L1) is a glycoprotein that is expressed on the cell surface of both hematopoietic and nonhematopoietic cells. PD-L1 play a role in the immune tolerance and protect self-tissues from immune system attack. Dysfunction of this molecule has been highlighted in the pathogenesis of tumors, autoimmunity, and infectious disorders. MicroRNAs (miRNAs) are endogenous molecules that are classified as small non-coding RNA with approximately 20-22 nucleotides (nt) length. The function of miRNAs is based on complementary interactions with target mRNA via matching completely or incompletely. The result of this function is decay of the target mRNA or preventing mRNA translation. In the past decades, several miRNAs have been discovered which play an important role in the regulation of PD-L1 in various malignancies. In this review, we discuss the effect of miRNAs on PD-L1 expression and consider the effect of miRNAs on the synthetic pathway of PD-L1, especially during cancers.
Collapse
Affiliation(s)
| | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nadia Sharafkandi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Babaie
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
8
|
Ding S, Liu G, Jiang H, Fang J. MicroRNA Determines the Fate of Intestinal Epithelial Cell Differentiation and Regulates Intestinal Diseases. Curr Protein Pept Sci 2019; 20:666-673. [PMID: 30678626 DOI: 10.2174/1389203720666190125110626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
Abstract
The rapid self-renewal of intestinal epithelial cells enhances intestinal function, promotes the nutritional needs of animals and strengthens intestinal barrier function to resist the invasion of foreign pathogens. MicroRNAs (miRNAs) are a class of short-chain, non-coding RNAs that regulate stem cell proliferation and differentiation by down-regulating hundreds of conserved target genes after transcription via seed pairing to the 3' untranslated regions. Numerous studies have shown that miRNAs can improve intestinal function by participating in the proliferation and differentiation of different cell populations in the intestine. In addition, miRNAs also contribute to disease regulation and therefore not only play a vital role in the gastrointestinal disease management but also act as blood or tissue biomarkers of disease. As changes to the levels of miRNAs can change cell fates, miRNA-mediated gene regulation can be used to update therapeutic strategies and approaches to disease treatment.
Collapse
Affiliation(s)
- Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China.,Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, China.,Academician Workstation of Hunan Baodong Farming Co., Ltd., Hunan 422001, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China.,Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan, China
| | - Hongmei Jiang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| |
Collapse
|
9
|
LncRNA-MEG3 protects against ganglion cell dysplasia in congenital intestinal atresia through directly regulating miR-211-5p/GDNF axis. Biomed Pharmacother 2019; 111:436-442. [DOI: 10.1016/j.biopha.2018.11.089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/07/2018] [Accepted: 11/25/2018] [Indexed: 12/20/2022] Open
|
10
|
Li EY, Zhao PJ, Jian J, Yin BQ, Sun ZY, Xu CX, Tang YC, Wu H. LncRNA MIAT overexpression reduced neuron apoptosis in a neonatal rat model of hypoxic-ischemic injury through miR-211/GDNF. Cell Cycle 2018; 18:156-166. [PMID: 30563429 DOI: 10.1080/15384101.2018.1560202] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate the underlying mechanism of lncRNA myocardial infarction-associated transcript (MIAT) in hypoxic-ischemic (HI)-induced neonatal cerebral palsy. MATERIALS AND METHODS Neonatal rat model of HI injury was established to detect the motor function. LncRNA MIAT, miR-211, glial cell line-derived neurotrophic factor (GDNF) and caspase-3 expressions were measured by qRT-PCR or western blot. The apoptosis of Neuro2A cells was detected by flow cytometry. RNA immunoprecipitation (RIP) and RNA pull-down assays were performed to confirm the interaction between MIAT and miR-211. RESULTS Compared with control group, lncRNA MIAT and GDNF were downregulated in striatal tissues of neonatal rats in HI group and oxygen glucose deprivation (OGD)-induced ischemic injury of Neuro2A cells, whereas miR-211 was up-regulated in striatal tissues of HI group and OGD-induced ischemic injury of Neuro2A cells. LncRNA MIAT interacted with miR-211, and lncRNA MIAT overexpression reduced neuron apoptosis through miR-211. Besides, GDNF expression was positively regulated by lncRNA MIAT and negatively regulated by miR-211 in Neuro2A cells. In vivo experiment proved MIAT promoted motor function and relieved HI injury. CONCLUSION MIAT overexpression reduced apoptosis of Neuro2A cells through miR-211/GDNF, which relieved HI injury of neonatal rats.
Collapse
Affiliation(s)
- En-Yao Li
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Peng-Ju Zhao
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Jie Jian
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Bao-Qi Yin
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Zhen-Yu Sun
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Cui-Xiang Xu
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - You-Cai Tang
- a Department of children rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan , The Fifth Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Hong Wu
- b Central Laboratory , Henan Province Hospital of TCM , Zhengzhou , China
| |
Collapse
|
11
|
Tian T, Zhang Y, Wu T, Yang L, Chen C, Li N, Li Y, Xu S, Fu Z, Cui X, Ji C, Chi X, Tong M, Chen R, Hong Q, Hu Y. miRNA profiling in the hippocampus of attention‐deficit/hyperactivity disorder rats. J Cell Biochem 2018; 120:3621-3629. [PMID: 30270454 DOI: 10.1002/jcb.27639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/15/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Tian Tian
- Department of Child Health Care The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Yan Zhang
- Department of Pediatrics Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, The Affiliated XuZhou Hospital of Medical College of Southeast University Jiangsu China
| | - Tianqi Wu
- Department of Cancer Institute Fudan University Shanghai Cancer Center Fudan University Shanghai China
| | - Lei Yang
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Chunxiao Chen
- Department of Child Health Care Yancheng Maternity and Child Health Care Hospital Yancheng China
| | - Nan Li
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Yue Li
- Department of Child Health Care The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Siliang Xu
- Department of Child Health Care The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Ziyi Fu
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Xianwei Cui
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Chenbo Ji
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Xia Chi
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Meiling Tong
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Ronghua Chen
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Qin Hong
- Department of Child Health Care Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University Nanjing China
| | - Youfang Hu
- Department of Child Health Care The First Affiliated Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|