1
|
Qin N, Liu H, Wang X, Liu Y, Chang H, Xia X. Sargassum fusiforme polysaccharides protect mice against Citrobacter rodentium infection via intestinal microbiota-driven microRNA-92a-3p-induced Muc2 production. Int J Biol Macromol 2025; 300:140271. [PMID: 39863236 DOI: 10.1016/j.ijbiomac.2025.140271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Sargassum fusiforme, widely consumed in Asian countries, has been proven to have various biological activities. However, the impacts and mechanisms of Sargassum fusiforme polysaccharides (SFPs) on intestinal bacterial infection are not yet fully understood. Our findings indicate that SFPs pretreatment ameliorates intestinal inflammation by reducing C. rodentium colonization, increasing colon length and levels of IL-10 and IL-22, decreasing IL-1β, IL-6, TNF-α, and IL-17 levels, inhibiting colonic crypt elongation and hyperplasia, and enhancing the intestinal mucosal barrier. The protective effects against intestinal bacterial infection are linked to enhanced clearance of C. rodentium and improvements in the intestinal mucosal barrier and C. rodentium-induced intestinal microbiota dysbiosis. Fecal microbiota transplantation experiments were conducted to evaluate the functional impact of microbiota induced by SFPs. The results suggest that intestinal microbiota modified by SFPs effectively countered C. rodentium infection. In addition, our study identified that miRNA-92a-3p is partially complementary to the 3'-UTR of the Notch1 gene, thereby repressing the Notch1-Hes1 signaling pathway and enhancing Muc2 secretion. Taken together, these findings reveal that SFPs protect mice from C. rodentium infection by activating the miR-92a-3p/Notch1-Hes1 regulatory axis driven by the intestinal microbiota, which stimulates Muc2 production to maintain intestinal barrier homeostasis.
Collapse
Affiliation(s)
- Ningbo Qin
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Hongxu Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xinru Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Yi Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Hong Chang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xiaodong Xia
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
2
|
Shi X, Li H, Dan Z, Shu C, Zhu R, Yang Q, Wang Y, Zhu H. Melatonin Potentiates Sensitivity to 5-Fluorouracil in Gastric Cancer Cells by Upregulating Autophagy and Downregulating Myosin Light-Chain Kinase. J Cancer 2023; 14:2608-2618. [PMID: 37779875 PMCID: PMC10539390 DOI: 10.7150/jca.85353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/23/2023] [Indexed: 10/03/2023] Open
Abstract
5-Fluorouracil is an effective chemotherapeutic drug for gastric cancer. However, the acquisition of chemotherapeutic resistance remains a challenge in treatment. Melatonin can enhance the therapeutic effect of 5-fluorouracil; however, the underlying mechanisms are not well understood. We investigated the effects of combinations of melatonin and 5-fluorouracil on the proliferation, migration and invasion of gastric cancer cells. Melatonin significantly potentiated the 5-fluorouracil-mediated inhibition of proliferation, migration and invasion in gastric cancer cells, which potentiates sensitivity to 5-FU by promoting the activation of Beclin-1-dependent autophagy and targeting the myosin light-chain kinase (MLCK) signaling pathway. Previous studies have shown that autophagy might be associated with the MLCK signaling pathway. The autophagy inhibitor, 3-methyladenine, effectively rescued the migratory and invasive capabilities of gastric cancer cells, while also reducing expression level of MLCK and the phosphorylation level of MLC. This indicates that autophagy is involved in tumor metastasis, which may be related to inhibition of the MLCK signaling pathway. Our findings indicate that melatonin can improve the effectiveness of 5-fluorouracil in gastric cancer and could be used as a supplemental agent in the treatment of gastric cancer with 5-fluorouracil.
Collapse
Affiliation(s)
- Xiaorui Shi
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Hongxia Li
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhangyong Dan
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Chuanlin Shu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Rumeng Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Qingling Yang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China
| | - Yi Wang
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
- Department of Biological Engineering, School of Life Sciences, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China
| | - Huaqing Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
3
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
4
|
Xiao X, Mao X, Chen D, Yu B, He J, Yan H, Wang J. miRNAs Can Affect Intestinal Epithelial Barrier in Inflammatory Bowel Disease. Front Immunol 2022; 13:868229. [PMID: 35493445 PMCID: PMC9043318 DOI: 10.3389/fimmu.2022.868229] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022] Open
Abstract
The most obvious pathological characterization of inflammatory bowel disease (IBD) is intestinal epithelium erosion and severe inflammation invasion. Micro-ribonucleic acids (miRNA or microRNA), single-stranded noncoding RNAs of ~22 nucleotides, have been considered as the potential therapeutic targets in the pathogenesis of IBD. Many previous studies have focused on the mechanisms that miRNAs use to regulate inflammation, immunity, and microorganisms in IBD. The review highlights in detail the findings of miRNAs in the intestinal epithelial barrier of IBD, and focuses on their gene targets, signaling pathways associated with IBD, and some potential therapies. It will be beneficial for the elucidation of the interaction between miRNAs and the intestinal epithelial barrier in IBD and provide a theoretical reference for preventing and treating IBD in the future.
Collapse
Affiliation(s)
- Xiangjun Xiao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| |
Collapse
|
5
|
IGF2BP2 promotes gastric cancer progression by regulating the IGF1R-RhoA-ROCK signaling pathway. Cell Signal 2022; 94:110313. [DOI: 10.1016/j.cellsig.2022.110313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
|
6
|
Yuan Y, Wang Z, Chen M, Jing Y, Shu W, Xie Z, Li Z, Xu J, He F, Jiao P, Wang J, Xu J, Xia Y, Liu S, Du H, Li H, Dai L, Dai Y, Zhang Y. Macrophage-Derived Exosomal miR-31-5p Promotes Oral Squamous Cell Carcinoma Tumourigenesis Through the Large Tumor Suppressor 2-Mediated Hippo Signalling Pathway. J Biomed Nanotechnol 2021; 17:822-837. [PMID: 34082869 DOI: 10.1166/jbn.2021.3066] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tumour-associated macrophages (TAMs) are thought to contribute to oral squamous cell carcinoma (OSCC) initiation and progression. However, the underlying mechanism through which TAMs foster OSCC progression is still unclear. This study intended to determine whether there are exclusively exosomal miRNAs-derived macrophages that are functionally necessary for OSCC progression. The phenotype of TAM recruitment in OSCC tissue samples was assessed, subsequently identifying the influence of M2 macrophages and exosomes derived from M2 macrophages on OSCC proliferation and tumorigenesis in vitro and in vivo. CD68 and CD163, the specific markers of M2 type macrophages, were upregulated in TAMs presented in intra-cancer tissues. M2 macrophages and M2 macrophage-derived exosomes (M2 exos) both can promote OSCC growth and tumorigenicity. An exosomal RNA-seq analysis was conducted to predict regulatory exosomal miRNAs related to OSCC growth, which determined miR-31-5p and LATS2 for subsequent experiments. Mechanistically, miR-31-5p was delivered to recipient OSCC cells through M2 exos and complementary pairing with the large tumor suppressor 2 (LATS2) coding sequence, thus suppressing the expression of LATS2 and inactivation the Hippo signaling pathway to support OSCC growth. Collectively, our findings demonstrate that M2 macrophage-derived exosomal miR- 31-5p can make tumor suppressor LATS2 gene inhibited and facilitate the progression of OSCC via inhibiting the Hippo signaling pathway, which possibly provides new targets for the molecular therapy of OSCC.
Collapse
Affiliation(s)
- Yi Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Zeyu Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Mengqi Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Yang Jing
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Wei Shu
- Department of Stomatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, Jiangsu, PR China
| | - Zhuoying Xie
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China
| | - Zhiyang Li
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008
| | - Juanyong Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Feng He
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Pengfei Jiao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Jiaqing Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Jiamin Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Yan Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Siyu Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Hongming Du
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Hongwei Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Lu Dai
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu, PR China
| | - Youjin Dai
- Key Laboratory of Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| |
Collapse
|
7
|
Catharmus tinctorius volatile oil promote the migration of mesenchymal stem cells via ROCK2/Myosin light chain signaling. Chin J Nat Med 2020; 17:506-516. [PMID: 31514982 DOI: 10.1016/s1875-5364(19)30072-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Indexed: 12/24/2022]
Abstract
MSC transplantation has been explored as a new clinical approach to stem cell-based therapies for bone diseases in regenerative medicine due to their osteogenic capability. However, only a small population of implanted MSC could successfully reach the injured areas. Therefore, enhancing MSC migration could be a beneficial strategy to improve the therapeutic potential of cell transplantation. Catharmus tinctorius volatile oil (CTVO) was found to facilitate MSC migration. Further exploration of the underlying molecular mechanism participating in the pro-migratory ability may provide a novel strategy to improve MSC transplantation efficacy. This study indicated that CTVO promotes MSC migration through enhancing ROCK2 mRNA and protein expressions. MSC migration induced by CTVO was blunted by ROCK2 inhibitor, which also decreased myosin light chain (MLC) phosphorylation. Meanwhile, the siRNA for ROCK2 inhibited the effect of CTVO on MSC migration ability and attenuated MLC phosphorylation, suggesting that CTVO may promote BMSC migration via the ROCK2/MLC signaling. Taken together, this study indicates that C. tinctorius volatile oil could enhance MSC migration via ROCK2/MLC signaling in vitro. C. tinctorius volatile oil-targeted therapy could be a beneficial strategy to improve the therapeutic potential of cell transplantation for bone diseases in regenerative medicine.
Collapse
|
8
|
Li L, Jing L, Zhao J, Lv J, Yang W, Li W, Zhou L. Valsartan inhibits RhoA-ROCK2-MYL pathway in rat model of alcoholic cardiomyopathy. Exp Ther Med 2019; 18:4313-4321. [PMID: 31777538 PMCID: PMC6862588 DOI: 10.3892/etm.2019.8079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/01/2018] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate variations in the Ras homolog gene family, member A (RhoA)-Rho-associated protein kinase 2 (ROCK2)-myosin light chain (MYL) pathway in a rat model of alcoholic cardiomyopathy (ACM) and the role of angiotensin-converting enzyme inhibitor drugs. Rat models of ACM were established via alcoholic gavage + free access to alcohol. The structural and functional changes of the heart were analyzed by hematoxylin-eosin staining, Masson's trichrome staining, immunohistochemistry staining, western blotting and fluorescence quantitative polymerase chain reaction. A total of 16 weeks later, a decreased ejection fraction and left ventricular fractional shortening in the alcohol group compared with the control group were demonstrated resulting in an increased left ventricular end diastolic diameter. These adverse effects were ameliorated following treatment with valsartan. In addition, the alcohol group revealed a disorganized arrangement of myocardial filaments, which was improved upon treatment with valsartan. RhoA and ROCK2 protein expression significantly increased in myocardial cells in the alcohol compared with the control group. Following drug intervention with valsartan, expression of RhoA and ROCK2 proteins were inhibited in the alcohol group. Furthermore, significantly elevated RhoA and ROCK2 and decreased MYL protein and mRNA expression in the alcohol group was demonstrated compared with the control group. Administration of valsartan reversed the expression profile of RhoA, ROCK and MYL in ACM. Expression of RhoA and ROCK were elevated with downregulation of MYL resulting in heart failure. However, the angiotensin receptor antagonist diminished the expression of RhoA and ROCK and enhanced the expression of MYL. The results of the present study suggest a curative effect of valsartan in ACM.
Collapse
Affiliation(s)
- Luyifei Li
- Department of Internal Critical Illness, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Ling Jing
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jiyi Zhao
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jiachen Lv
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Wen Yang
- Department of The First Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Weimin Li
- Department of The Fifth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Lijun Zhou
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
9
|
Nam S, Kim JH, Lee DH. RHOA in Gastric Cancer: Functional Roles and Therapeutic Potential. Front Genet 2019; 10:438. [PMID: 31156701 PMCID: PMC6529512 DOI: 10.3389/fgene.2019.00438] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 04/29/2019] [Indexed: 12/23/2022] Open
Abstract
The well-known signal mediator and small GTPase family member, RHOA, has now been associated with the progression of specific malignancies. In this review, we appraise the biomedical literature regarding the role of this enzyme in gastric cancer (GC) signaling, suggesting potential clinical significance. To that end, we examined RHOA activity, with regard to second-generation hallmarks of cancer, finding particular association with the hallmark "activation of invasion and metastasis." Moreover, an abundance of studies show RHOA association with Lauren classification diffuse subtype, in addition to poorly differentiated GC. With regard to therapeutic value, we found RHOA signaling to influence the activity of specific widely used chemotherapeutics, and its possible antagonism by various dietary constituents. We also review currently available targeted therapies for GC. The latter, however, showed a paucity of such agents, underscoring the urgent need for further investigation into treatments for this highly lethal malignancy.
Collapse
Affiliation(s)
- Seungyoon Nam
- Department of Genome Medicine and Science, College of Medicine, Gachon University, Incheon, South Korea.,Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, South Korea.,Gachon Advanced Institute of Health Sciences and Technology, Gachon University, Incheon, South Korea.,Department of Life Sciences, Gachon University, Seongnam, South Korea
| | - Jung Ho Kim
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, School of Medicine, Gachon University, Incheon, South Korea.,Gachon Medical Research Institute, Gachon University Gil Medical Center, Incheon, South Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, South Korea.,Department of Internal Medicine, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
10
|
Bardua M, Haftmann C, Durek P, Westendorf K, Buttgereit A, Tran CL, McGrath M, Weber M, Lehmann K, Addo RK, Heinz GA, Stittrich AB, Maschmeyer P, Radbruch H, Lohoff M, Chang HD, Radbruch A, Mashreghi MF. MicroRNA-31 Reduces the Motility of Proinflammatory T Helper 1 Lymphocytes. Front Immunol 2018; 9:2813. [PMID: 30574141 PMCID: PMC6291424 DOI: 10.3389/fimmu.2018.02813] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/14/2018] [Indexed: 12/27/2022] Open
Abstract
Proinflammatory type 1 T helper (Th1) cells are enriched in inflamed tissues and contribute to the maintenance of chronic inflammation in rheumatic diseases. Here we show that the microRNA- (miR-) 31 is upregulated in murine Th1 cells with a history of repeated reactivation and in memory Th cells isolated from the synovial fluid of patients with rheumatic joint disease. Knock-down of miR-31 resulted in the upregulation of genes associated with cytoskeletal rearrangement and motility and induced the expression of target genes involved in T cell activation, chemokine receptor- and integrin-signaling. Accordingly, inhibition of miR-31 resulted in increased migratory activity of repeatedly activated Th1 cells. The transcription factors T-bet and FOXO1 act as positive and negative regulators of T cell receptor (TCR)-mediated miR-31 expression, respectively. Taken together, our data show that a gene regulatory network involving miR-31, T-bet, and FOXO1 controls the migratory behavior of proinflammatory Th1 cells.
Collapse
Affiliation(s)
- Markus Bardua
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | | | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | | | | | - Cam Loan Tran
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | - Mairi McGrath
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | - Melanie Weber
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | - Katrin Lehmann
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | | | | | | | | | - Helena Radbruch
- Department of Neuropathology, Charité-Universitätsmedizin, Berlin, Germany
| | - Michael Lohoff
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | | | | | | |
Collapse
|
11
|
Yu T, Ma P, Wu D, Shu Y, Gao W. Functions and mechanisms of microRNA-31 in human cancers. Biomed Pharmacother 2018; 108:1162-1169. [PMID: 30372817 DOI: 10.1016/j.biopha.2018.09.132] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs can exhibit opposite functions in different tumors. MiR-31 is a representative example as it can not only enhance tumor development and progression in pancreatic cancer, colorectal cancer and so on, but also inhibit tumorigenesis and induce apoptosis in ovarian cancer, prostate cancer and etc. The mechanism underlying its' pleiotropy remains unknown. Several recent studies that focused on the global gene expression changes caused by aberrant miR-31 provided information on the upstream and downstream events associated with deregulated miR-31. MiR-31 might interact with a number of signaling pathways including RAS/MARK, PI3K/AKT and RB/E2F to play its opposite functions. This review summarizes the target genes and pathways associated with miR-31 and examines the mechanisms underlying the function of miR-31. The resulting hypothesis is possible that the tissue-specific features of adenocarcinoma and squamous cell cancer and the positive feedback loop consists of miR-31 and its upstream and downstream may account for the diversity of miR-31 functions.
Collapse
Affiliation(s)
- Tao Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Deqin Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
12
|
Wang LL, Li HX, Yang YY, Su YL, Lian JS, Li T, Xu J, Wang XN, Jin N, Liu XF. MiR-31 is a potential biomarker for diagnosis of head and neck squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4339-4345. [PMID: 31949830 PMCID: PMC6962945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/13/2018] [Indexed: 06/10/2023]
Abstract
MicroRNAs (miRNAs) play important roles in the development of head and neck squamous cell carcinoma (HNSCC). However, their potential clinical value as biomarkers remains poorly known. The aim of this study was to assess the association between tissue/serum miR-31 expression levels and prognosis of HNSCC. In this clinical study, tumor samples were obtained from 118 patients with HNSCC and 48 patients with oral epithelial dysplasia, and blood samples were collected from all the HNSCC cases and 60 normal controls. The expression levels of tissue/serum miR-31 were measured by real-time PCR. Chi-square test was used to evaluate the correlation between tissue/serum miR-31 and clinical parameters of HNSCC. Survival curves were constructed using the Kaplan-Meier method and log-rank test. Multivariate Cox regression analyses were used to estimate independent predictors of survival for HNSCC. Our findings showed that tissue miR-31 levels in HNSCC tumor specimens exhibited higher than that in oral epithelial dysplasia samples and normal tissues. Oral epithelial dysplasia with higher expression of miR-31 was more prone to progress into HNSCC. Likewise, serum miR-31 expression in HNSCC patients was markedly increased in compared to normal controls. Moreover, serum miR-31 performed well to distinguish HNSCC subjects from controls. In addition, increased tissue/serum miR-31 expression was positively correlated with poor clinical variables and dismal prognosis. Finally, tissue miR-31 was confirmed to be an independent prognostic factor for HNSCC. Taken together, miR-31 had strong potential as a promising biomarker in HNSCC detection.
Collapse
Affiliation(s)
- Long-Long Wang
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Hai-Xia Li
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Yong-Yan Yang
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Ya-La Su
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Jin-Shan Lian
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Teng Li
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Jie Xu
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Xiao-Ning Wang
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Na Jin
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| | - Xian-Feng Liu
- The Second Department of Head and Neck Surgery, Baotou Cancer Hospital, The Affiliated Cancer Hospital of Baotou Medical College Baotou 014030, Inner Mongolia, China
| |
Collapse
|
13
|
MicroRNA-31 Function as a Suppressor Was Regulated by Epigenetic Mechanisms in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5348490. [PMID: 29333444 PMCID: PMC5733238 DOI: 10.1155/2017/5348490] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/21/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023]
Abstract
Gastric cancer is one of the most lethal malignancies worldwide. The aberrant expression of microRNA-31 (miR-31) has been reported in gastric cancer; however, its regulation mechanisms are still unclear. Here, we confirmed that miR-31 expression was significantly decreased in gastric cancer tissue and cell lines. Ectopic expression of miR-31 potentially suppresses proliferation and induced early apoptosis in gastric cancer cells. Furthermore, miR-31 expression was regulated as a result of epigenetic mechanisms. The downregulation of miR-31 was associated with promoter DNA methylation status in gastric cancer and cell lines. Moreover, we found that HDAC2 was the direct target of miR-31 by binding to 3′-UTR from the results of luciferase reporter assays, qRT-PCR, and western blotting. HDAC2 played an activation role in tumor growth, whose expression is upregulated and inversely associated with miR-31 levels. All the results suggested that miR-31 function as a crucial tumor suppressor was regulated by epigenetic mechanisms in gastric cancer. We found an epigenetic pathway loop, DNA methylation-miRNA expression-target gene-tumor progression in gastric cancer, and also provided implications for molecular diagnosis and therapeutics of gastric malignancies by detecting miR-31 as a potential target.
Collapse
|
14
|
Korourian A, Roudi R, Shariftabrizi A, Madjd Z. MicroRNA-31 inhibits RhoA-mediated tumor invasion and chemotherapy resistance in MKN-45 gastric adenocarcinoma cells. Exp Biol Med (Maywood) 2017; 242:1842-1847. [PMID: 28836853 DOI: 10.1177/1535370217728460] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
microRNAs are small single-stranded non-coding RNA molecules which modify gene expression by silencing potential target genes. The aberrant expression of RhoA, a small GTPase protein of Rho family, is involved in gastric cancer tumorigenesis. Since miR-31 is a pleomorphic molecule, we evaluated the miR-31/RhoA axis in inducing the malignant phenotype of gastric cancer cells MKN-45. Also, the clinicopathological significance of RhoA was investigated in a well-defined collection of gastric carcinomas which were embedded in tissue microarray blocks. Induction of miR-31 in MKN-45 followed by suppression of RhoA expression resulted in increased sensitivity to 5-fluorouracil, inhibition of cell proliferation, and invasion compared to the control groups. Immunohistochemical analysis in gastric adenocarcinoma patients' samples showed significantly higher expression of RhoA in diffuse versus intestinal subtype tumors ( P = 0.009), poorly differentiated versus well and moderately differentiated tumors ( P = 0.03) and the presence of vascular invasion versus the absence of vascular invasion ( P = 0.04). Our findings suggest a critical role for miR-31, as a tumor suppressor gene, in gastric cancer tumorigenesis by targeting the RhoA. Impact statement Gastric cancer ranks as the third leading cause of cancer-associated deaths worldwide. The RhoA gene encodes a small GTPase protein of Rho family (RhoA) that its dysregulation is associated with cell motility and invasion. A strong line of evidence supports the regulation of RhoA by a number of miRs, including miR-31 in tumors. Our findings revealed that miR-31 is involved in gastric cancer tumorigenesis as a tumor suppressor gene. Through down-regulation of RhoA, miR-31 decreased cell proliferation, migration, and invasion in gastric cancer cells. In addition, induction of miR-31 increased sensitivity to 5-FU; thus, increasing its tissue concentrations could be a potential target for treatment of gastric cancer in the future.
Collapse
Affiliation(s)
- Alireza Korourian
- 1 Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14530, Iran
| | - Raheleh Roudi
- 2 Oncopathology Research Center, Iran University of Medical Sciences, Tehran 14496-14530, Iran
| | - Ahmad Shariftabrizi
- 3 Department of Nuclear Medicine and Molecular Imaging, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zahra Madjd
- 1 Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14530, Iran.,2 Oncopathology Research Center, Iran University of Medical Sciences, Tehran 14496-14530, Iran
| |
Collapse
|
15
|
Xiong Y, Wang C, Shi L, Wang L, Zhou Z, Chen D, Wang J, Guo H. Myosin Light Chain Kinase: A Potential Target for Treatment of Inflammatory Diseases. Front Pharmacol 2017; 8:292. [PMID: 28588494 PMCID: PMC5440522 DOI: 10.3389/fphar.2017.00292] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 05/08/2017] [Indexed: 01/30/2023] Open
Abstract
Myosin light chain kinase (MLCK) induces contraction of the perijunctional apical actomyosin ring in response to phosphorylation of the myosin light chain. Abnormal expression of MLCK has been observed in respiratory diseases, pancreatitis, cardiovascular diseases, cancer, and inflammatory bowel disease. The signaling pathways involved in MLCK activation and triggering of endothelial barrier dysfunction are discussed in this review. The pharmacological effects of regulating MLCK expression by inhibitors such as ML-9, ML-7, microbial products, naturally occurring products, and microRNAs are also discussed. The influence of MLCK in inflammatory diseases starts with endothelial barrier dysfunction. The effectiveness of anti-MLCK treatment may depend on alleviation of that primary pathological mechanism. This review summarizes evidence for the potential benefits of anti-MLCK agents in the treatment of inflammatory disease and the importance of avoiding treatment-related side effects, as MLCK is widely expressed in many different tissues.
Collapse
Affiliation(s)
- Yongjian Xiong
- Central Laboratory, The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Chenou Wang
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Liqiang Shi
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Liang Wang
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Zijuan Zhou
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Dapeng Chen
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Jingyu Wang
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Huishu Guo
- Central Laboratory, The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| |
Collapse
|