1
|
Liu Y, Ma J, Ma Y, Wang BZ, Wang Y, Yuan J, Zhang F, Zhao X, Chen K, Zhang X, Wang H. Neutrophil extracellular traps impede cancer metastatic seeding via protease-activated receptor 2-mediated downregulation of phagocytic checkpoint CD24. J Immunother Cancer 2025; 13:e010813. [PMID: 40010762 PMCID: PMC11865804 DOI: 10.1136/jitc-2024-010813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Phagocytic clearance by macrophages represents a critical immune surveillance mechanism in cancer liver metastasis. Neutrophils, the most abundant immune cells encountered by cancer cells in circulation, play key roles in metastasis through neutrophil extracellular traps (NETs). Although NETs promote macrophage phagocytosis during infection, whether they regulate phagocytosis during cancer metastasis is unknown. The present study aimed to explore the roles of NETs in regulating macrophage phagocytosis during the seeding process of liver metastasis and the mechanisms underlying the roles. METHODS A lipopolysaccharide-induced NET model was applied to study the role of NETs on colorectal cancer (CRC) liver metastasis. The neutrophils isolated from human peripheral blood were stimulated with PMA to release NETs, which were collected and added to the cultures of different CRC cell lines for in vitro studies. Macrophage phagocytosis was assessed with flow cytometry in vitro and in vivo. RNA-seq and microRNA array analyses were performed to identify key pathways regulated by NETs and downstream key molecules. The macrophage phenotypes were evaluated using immunohistochemistry, flow cytometry, and cytokine and chemokine arrays. RESULTS NETs promote macrophage phagocytosis both in vitro and in vivo. Neutrophil elastase (NE), which was able to inactivate the canonical signal of protease-activated receptor 2 (PAR2), downregulated the phagocytotic checkpoint CD24. Notably, PAR2 deficiency imitated the effect of NETs on phagocytosis and CD24. Mechanistic studies indicated that inhibiting PAR2 expression upregulated miR-34a and miR-146a and downregulated CD24 in cancer cells. In addition, PAR2 depletion enhanced the recruitment and M1 polarization of macrophages by upregulating CSF-1 and CXCL1. The correlation of NETs/NE and CD24 was corroborated using human CRC specimens. Furthermore, PAR2 blockade combined with an anti-EGFR antibody (cetuximab (CTX)) synergistically enhanced the phagocytic ability of macrophages and suppressed liver metastasis in vivo. CONCLUSIONS NET-derived elastase inactivated PAR2 canonical signaling and promoted phagocytosis by downregulating CD24, which functions as a phagocytotic checkpoint in CRC liver metastasis. Thus, PAR2 inhibitors combined with CTX may serve as a novel therapeutic strategy against advanced CRC.
Collapse
Affiliation(s)
- Yu Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Jianhui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Bing-Zhi Wang
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Yinong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Junhu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Fanyu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Kun Chen
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Liaoning, China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Liaoning, China
| | - Xiaoli Zhang
- Department of Injury and Repair, Beijing Neurosurgical Institute, Capital Medical University, Beijing, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| |
Collapse
|
2
|
Peach CJ, Edgington-Mitchell LE, Bunnett NW, Schmidt BL. Protease-activated receptors in health and disease. Physiol Rev 2023; 103:717-785. [PMID: 35901239 PMCID: PMC9662810 DOI: 10.1152/physrev.00044.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022] Open
Abstract
Proteases are signaling molecules that specifically control cellular functions by cleaving protease-activated receptors (PARs). The four known PARs are members of the large family of G protein-coupled receptors. These transmembrane receptors control most physiological and pathological processes and are the target of a large proportion of therapeutic drugs. Signaling proteases include enzymes from the circulation; from immune, inflammatory epithelial, and cancer cells; as well as from commensal and pathogenic bacteria. Advances in our understanding of the structure and function of PARs provide insights into how diverse proteases activate these receptors to regulate physiological and pathological processes in most tissues and organ systems. The realization that proteases and PARs are key mediators of disease, coupled with advances in understanding the atomic level structure of PARs and their mechanisms of signaling in subcellular microdomains, has spurred the development of antagonists, some of which have advanced to the clinic. Herein we review the discovery, structure, and function of this receptor system, highlight the contribution of PARs to homeostatic control, and discuss the potential of PAR antagonists for the treatment of major diseases.
Collapse
Affiliation(s)
- Chloe J Peach
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| |
Collapse
|
3
|
Qiao X, Zhao F. Long non-coding RNA Opa interacting protein 5-antisense RNA 1 binds to micorRNA-34a to upregulate oncogenic PD-L1 in non-small cell lung cancer. Bioengineered 2022; 13:9264-9273. [PMID: 35411833 PMCID: PMC9161958 DOI: 10.1080/21655979.2022.2036904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNA (lncRNA) OPA-interacting protein 5 antisense transcript 1 (OIP5-AS1) plays an oncogenic role in several types of cancer, but whether it is involved in non-small-cell lung cancer (NSCLC) is unclear. Our preliminary sequencing analysis revealed the upregulation of OIP5-AS1 in NSCLC. In this study, gene expression levels were analyzed by RT-qPCR. RNA-RNA pull-down assay was applied to detect direct interactions between RNAs. Overexpression assays were performed to explore the relationship between miR-34a and OIP5-AS1. CCK-8 assay and colony formation assay were applied to evaluate cell proliferation. In NSCLC cells (H23), overexpression of OIP5-AS1 increased the expression levels of programmed death-ligand 1 (PD-L1). In addition, inhibition of OIP5-AS1 and overexpression of miR-34a decreased the expression levels of PD-L1, and miR-34a significantly blocked the role of overexpression of OIP5-AS1. Overexpression of OIP5-AS1 and PD-L1 promoted H23 and H22 cells proliferation, while silencing of miR-34a and OIP5-AS1 played opposite roles and eliminated the effects of overexpression of OIP5-AS1 on cell proliferation. Therefore, OIP5-AS1 was upregulated to enhance the expression of oncogenic PD-L1 by sponging miR-34a in NSCLC, leading to promoted NSCLC cell proliferation. Our study also demonstrated that OIP5-AS1 was upregulated while miR-34a was downregulated in NSCLC.
Collapse
Affiliation(s)
- Xinwei Qiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, China
| | - Feng Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, China
| |
Collapse
|
4
|
He S, Xu M, Xiong Z, Hu Y, Huo Q, Lu J, Lin Y, Yang L. Predictive value of protease-activated receptor-2 (PAR 2 ) in cervical cancer metastasis. J Cell Mol Med 2020; 25:1415-1424. [PMID: 33369107 PMCID: PMC7875903 DOI: 10.1111/jcmm.16227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
Metastasis is the primary cause of an unfavourable prognosis in patients with malignant cancer. Over the last decade, the role of proteinases in the tumour microenvironment has attracted increasing attention. As a sensor of proteinases, proteinase-activated receptor 2 (PAR2 ) plays crucial roles in the metastatic progression of cervical cancer. In the present study, the expression of PAR2 in multiple types of cancer was analysed by Gene Expression Profiling Interactive Analysis (GEPIA). Kaplan-Meier plotter was used to calculate the correlation between survival and the levels of PAR2 , Grb-associated binding protein 2(Gab2) and miR-125b. Immunohistochemistry (IHC) was performed to examine PAR2 expression in a tissue microarray (TMA) of CESCs. Empower Stats was used to assess the predictive value of PAR2 in the metastatic potential of CESC. We found that PAR2 up-regulation was observed in multiple types of cancer. Moreover, PAR2 expression was positively correlated with the clinicopathologic characteristics of CESC. miR-125b and its target Gab2, which are strongly associated with PAR2 -induced cell migration, are well-characterized as predictors of the prognostic value of CESC. Most importantly, the Cancer Genome Atlas (TCGA) data set analysis showed that the area under the curve (AUC) of the PAR2 model was significantly greater than that of the traditional model (0.833 vs 0.790, P < .05), demonstrating the predictive value of PAR2 in CESC metastasis. Our results suggest that PAR2 may serve as a prognostic factor for metastasis in CESC patients.
Collapse
Affiliation(s)
- Shengnan He
- Biobank of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Meiquan Xu
- Pathology Department of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhen Xiong
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Ye Hu
- Biobank of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Graduate School, University of South China, Hengyang, China
| | - Qin Huo
- Biobank of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jingxiao Lu
- Biobank of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yuntao Lin
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lan Yang
- Biobank of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Gastroenterology of Shenzhen Second People's Hospital, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
5
|
Zhao R, Chen X, Song H, Bie Q, Zhang B. Dual Role of MSC-Derived Exosomes in Tumor Development. Stem Cells Int 2020; 2020:8844730. [PMID: 32963552 PMCID: PMC7499322 DOI: 10.1155/2020/8844730] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/08/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a class of adult stem cells derived from the mesoderm. They can self-renew, have multidirectional differentiation potential, and can differentiate into a variety of mesenchymal tissues. MSCs can produce a large number of exosomes, which can mediate information exchange and transmission between cells in the tumor microenvironment under conditions of rest or stress. Recent studies have reported conflicting findings regarding the effect of MSC-derived exosomes on tumors. Some studies have suggested that MSC-derived exosomes can promote tumor growth and metastasis, but others have reported that they can inhibit tumor cell growth. Here, we investigate the two sides of the debate regarding the effect of MSC-derived exosomes on tumors and analyze the reasons for the divergent findings.
Collapse
Affiliation(s)
- Rou Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- Department of Microbiology, Qingdao University Life Science College, Qingdao, Shandong, China
| | - Xinke Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Hui Song
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Qingli Bie
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, Shandong, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
6
|
Kawaguchi M, Yamamoto K, Kataoka H, Izumi A, Yamashita F, Kiwaki T, Nishida T, Camerer E, Fukushima T. Protease-activated receptor-2 accelerates intestinal tumor formation through activation of nuclear factor-κB signaling and tumor angiogenesis in Apc Min/+ mice. Cancer Sci 2020; 111:1193-1202. [PMID: 31997435 PMCID: PMC7156842 DOI: 10.1111/cas.14335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte growth factor activator inhibitor‐1 (HAI‐1), encoded by the SPINT1 gene, is a membrane‐bound protease inhibitor expressed on the surface of epithelial cells. Hepatocyte growth factor activator inhibitor‐1 regulates type II transmembrane serine proteases that activate protease‐activated receptor‐2 (PAR‐2). We previously reported that deletion of Spint1 in ApcMin/+ mice resulted in accelerated formation of intestinal tumors, possibly through enhanced nuclear factor‐κB signaling. In this study, we examined the role of PAR‐2 in accelerating tumor formation in the ApcMin/+ model in the presence or absence of Spint1. We observed that knockout of the F2rl1 gene, encoding PAR‐2, not only eliminated the enhanced formation of intestinal tumors caused by Spint1 deletion, but also reduced tumor formation in the presence of Spint1. Exacerbation of anemia and weight loss associated with HAI‐1 deficiency was also normalized by compound deficiency of PAR‐2. Mechanistically, signaling triggered by deregulated protease activities increased nuclear translocation of RelA/p65, vascular endothelial growth factor expression, and vascular density in ApcMin/+‐induced intestinal tumors. These results suggest that serine proteases promote intestinal carcinogenesis through activation of PAR‐2, and that HAI‐1 plays a critical tumor suppressor role as an inhibitor of matriptase, kallikreins, and other PAR‐2 activating proteases.
Collapse
Affiliation(s)
- Makiko Kawaguchi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Koji Yamamoto
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Kataoka
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Aya Izumi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Fumiki Yamashita
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takumi Kiwaki
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takahiro Nishida
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Eric Camerer
- Inserm U970, Paris Cardiovascular Research Center, Université de Paris, Paris, France
| | | |
Collapse
|
7
|
Moradi-Marjaneh R, Khazaei M, Ferns GA, Aghaee-Bakhtiari SH. The Role of TGF-β Signaling Regulatory MicroRNAs in the Pathogenesis of Colorectal Cancer. Curr Pharm Des 2019; 24:4611-4618. [PMID: 30636580 DOI: 10.2174/1381612825666190110150705] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers globally and is associated with a high mortality rate. The transforming growth factor beta (TGF-β) signaling pathway plays an important role in normal intestinal tissue function, but has also been implicated in the development of CRC. MicroRNAs (miRNAs) have also recently emerged as important regulators of cancer development and progression. They act by targeting multiple signaling pathways including the TGF-β signaling pathway. There is growing evidence demonstrating that miRNAs target various components of the TGF-β signaling pathway, including TGF-β1, TGF-β2, regulatory SMADs (SMAD1, 2, 3, 5 and 9), co-mediator SMAD4, inhibitory SMADs (SMAD6 and 7) and the TGF-β receptors, and thereby alter the proliferation and migration of CRC cells. In this review, we summarize the data concerning the interaction between TGF-β signaling pathway and miRNAs with the aim to better understanding the CRC molecular mechanisms and hence better management of this disease.
Collapse
Affiliation(s)
- Reyhaneh Moradi-Marjaneh
- Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, United Kingdom
| | - Seyed H Aghaee-Bakhtiari
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Zhang X, Fang Q, Ma Y, Zou S, Liu Q, Wang H. Protease activated receptor 2 mediates tryptase-induced cell migration through MYO10 in colorectal cancer. Am J Cancer Res 2019; 9:1995-2006. [PMID: 31598400 PMCID: PMC6780659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/18/2019] [Indexed: 06/10/2023] Open
Abstract
MYO10 is an actin-based motor protein and correlates with cancer metastasis. However, the regulation of MYO10 by tumor microenvironment is unknown. In the current study, we found that the expression of protease activated receptor 2 (PAR2) was highly correlated with that of MYO10 in colorectal carcinoma (CRC) specimens. Both MYO10 and PAR2 were up-regulated in lymph node metastasis group compared with non-metastasis group. Activation of PAR2 significantly induced cell migration through the up-regulation of MYO10, which was mediated by repression of miR-204 in multiple cell lines. Interestingly, it was observed that tryptase was highly expressed in adjacent tissue around primary tumor of CRC. Furthermore, tryptase stimulated cell migration and up-regulated MYO10 expression through a PAR2-dependent manner. Taken together, our findings showed that PAR2 enhanced the expression of MYO10 through the repression of miR-204. PAR2 mediated tryptase-induced cell migration and might contribute to the invasion of cancer cells at the edge of tumor.
Collapse
Affiliation(s)
- Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| | - Qing Fang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| | - Qian Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021, China
| |
Collapse
|
9
|
Galieva LR, James V, Mukhamedshina YO, Rizvanov AA. Therapeutic Potential of Extracellular Vesicles for the Treatment of Nerve Disorders. Front Neurosci 2019; 13:163. [PMID: 30890911 PMCID: PMC6411850 DOI: 10.3389/fnins.2019.00163] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/12/2019] [Indexed: 12/22/2022] Open
Abstract
The use of extracellular vesicles (EVs) as cell free therapy is a promising approach to stimulate tissue regeneration including that of the nervous system. EVs transfer bioactive proteins and lipids, RNA and microRNAs, which play a relevant role in EV-mediated intercellular communication. The immunomodulatory, anti-inflammatory, and neuroprotective effects of mesenchymal stem cells-derived EVs have been well studied, knowledge of this paracrine mechanism and the availability of these cells, positions mesenchymal stem cells as a potential source of EVs for cell free therapy for a variety of regenerative and nervous system disorders. In this review, we focus on the immunomodulatory and neuroprotective effects of stem cells-derived EVs within in vitro and in vivo models of nerve disorders.
Collapse
Affiliation(s)
- Luisa R Galieva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Yana O Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Histology, Cytology, and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
10
|
Hibner G, Kimsa-Furdzik M, Francuz T. Relevance of MicroRNAs as Potential Diagnostic and Prognostic Markers in Colorectal Cancer. Int J Mol Sci 2018; 19:ijms19102944. [PMID: 30262723 PMCID: PMC6213499 DOI: 10.3390/ijms19102944] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is currently the third and the second most common cancer in men and in women, respectively. Every year, more than one million new CRC cases and more than half a million deaths are reported worldwide. The majority of new cases occur in developed countries. Current screening methods have significant limitations. Therefore, a lot of scientific effort is put into the development of new diagnostic biomarkers of CRC. Currently used prognostic markers are also limited in assessing the effectiveness of CRC therapy. MicroRNAs (miRNAs) are a promising subject of research especially since single miRNA can recognize a variety of different mRNA transcripts. MiRNAs have important roles in epigenetic regulation of basic cellular processes, such as proliferation, apoptosis, differentiation, and migration, and may serve as potential oncogenes or tumor suppressors during cancer development. Indeed, in a large variety of human tumors, including CRC, significant distortions in miRNA expression profiles have been observed. Thus, the use of miRNAs as diagnostic and prognostic biomarkers in cancer, particularly in CRC, appears to be an inevitable consequence of the advancement in oncology and gastroenterology. Here, we review the literature to discuss the potential usefulness of selected miRNAs as diagnostic and prognostic biomarkers in CRC.
Collapse
Affiliation(s)
- Grzegorz Hibner
- Department of Biochemistry, School of Medicine in Katowice, Medical University of Silesia in Katowice, St. Medyków 18, 40-752 Katowice, Poland.
| | - Małgorzata Kimsa-Furdzik
- Department of Biochemistry, School of Medicine in Katowice, Medical University of Silesia in Katowice, St. Medyków 18, 40-752 Katowice, Poland.
| | - Tomasz Francuz
- Department of Biochemistry, School of Medicine in Katowice, Medical University of Silesia in Katowice, St. Medyków 18, 40-752 Katowice, Poland.
| |
Collapse
|
11
|
He L, Ma Y, Li W, Han W, Zhao X, Wang H. Protease-activated receptor 2 signaling modulates susceptibility of colonic epithelium to injury through stabilization of YAP in vivo. Cell Death Dis 2018; 9:949. [PMID: 30237580 PMCID: PMC6148223 DOI: 10.1038/s41419-018-0995-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/01/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022]
Abstract
Hippo signaling plays critical roles in intestinal regeneration. However, the mechanisms which regulate its activity in vivo are largely unknown. We hypothesize that protease-activated receptor 2 (PAR2) signaling, which could be activated by trypsin, might affect YAP activity in the setting of tissue damage and regeneration. It is found that knockout of PAR2 severely aggravates the mucosal damage induced by dextran sodium sulfate (DSS) in mouse, which correlated with notable repression of YAP protein in colonic epithelial cells. Although the cytokine expression is reduced, the damage of colonic crypt is more severe after DSS-induced colitis in PAR2-/- mouse. In vitro, PAR2 activation causes the accumulation of YAP, while knockdown of PAR2 with shRNA dramatically represses the expression of YAP protein in different intestinal epithelial cell lines. Moreover, forced expression of YAP significantly reduces the production of reactive oxygen species (ROS) and the sensitivity to nitric oxide-induced apoptosis in PAR2-deficient condition. Further studies show that PAR2 signaling stabilizes YAP protein but independent of Lats. Nevertheless PAR2 activation increased the binding of YAP with protein phosphatase PP1. Inhibition of PP1 with specific siRNA blocked PAR2-induced dephosphorylation of YAP. Taken together, PAR2 signaling might modulate susceptibility of colonic epithelium to injury through stabilization of YAP.
Collapse
Affiliation(s)
- Longmei He
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China, 100021
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China, 100021
| | - Weiwei Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China, 100021
| | - Wenxiao Han
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China, 100021
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China, 100021
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China, 100021.
| |
Collapse
|
12
|
Signaling Crosstalk of TGF-β/ALK5 and PAR2/PAR1: A Complex Regulatory Network Controlling Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19061568. [PMID: 29795022 PMCID: PMC6032192 DOI: 10.3390/ijms19061568] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Both signaling by transforming growth factor-β (TGF-β) and agonists of the G Protein-coupled receptors proteinase-activated receptor-1 (PAR1) and -2 (PAR2) have been linked to tissue fibrosis and cancer. Intriguingly, TGF-β and PAR signaling either converge on the regulation of certain matrix genes overexpressed in these pathologies or display mutual regulation of their signaling components, which is mediated in part through sphingosine kinases and sphingosine-1-phosphate and indicative of an intimate signaling crosstalk between the two pathways. In the first part of this review, we summarize the various regulatory interactions that have been discovered so far according to the organ/tissue in which they were described. In the second part, we highlight the types of signaling crosstalk between TGF-β on the one hand and PAR2/PAR1 on the other hand. Both ligand–receptor systems interact at various levels and by several mechanisms including mutual regulation of ligand–ligand, ligand–receptor, and receptor–receptor at the transcriptional, post-transcriptional, and receptor transactivation levels. These mutual interactions between PAR2/PAR1 and TGF-β signaling components eventually result in feed-forward loops/vicious cycles of matrix deposition and malignant traits that exacerbate fibrosis and oncogenesis, respectively. Given the crucial role of PAR2 and PAR1 in controlling TGF-β receptor activation, signaling, TGF-β synthesis and bioactivation, combining PAR inhibitors with TGF-β blocking agents may turn out to be more efficient than targeting TGF-β alone in alleviating unwanted TGF-β-dependent responses but retaining the beneficial ones.
Collapse
|
13
|
Strubberg AM, Madison BB. MicroRNAs in the etiology of colorectal cancer: pathways and clinical implications. Dis Model Mech 2017; 10:197-214. [PMID: 28250048 PMCID: PMC5374322 DOI: 10.1242/dmm.027441] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are small single-stranded RNAs that repress mRNA translation
and trigger mRNA degradation. Of the ∼1900 miRNA-encoding genes present
in the human genome, ∼250 miRNAs are reported to have changes in
abundance or altered functions in colorectal cancer. Thousands of studies have
documented aberrant miRNA levels in colorectal cancer, with some miRNAs reported
to actively regulate tumorigenesis. A recurrent phenomenon with miRNAs is their
frequent participation in feedback loops, which probably serve to reinforce or
magnify biological outcomes to manifest a particular cellular phenotype. Here,
we review the roles of oncogenic miRNAs (oncomiRs), tumor suppressive miRNAs
(anti-oncomiRs) and miRNA regulators in colorectal cancer. Given their stability
in patient-derived samples and ease of detection with standard and novel
techniques, we also discuss the potential use of miRNAs as biomarkers in the
diagnosis of colorectal cancer and as prognostic indicators of this disease.
MiRNAs also represent attractive candidates for targeted therapies because their
function can be manipulated through the use of synthetic antagonists and miRNA
mimics. Summary: This Review provides an overview of some important
microRNAs and their roles in colorectal cancer.
Collapse
Affiliation(s)
- Ashlee M Strubberg
- Division of Gastroenterology, Washington University School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Blair B Madison
- Division of Gastroenterology, Washington University School of Medicine, Washington University, Saint Louis, MO 63110, USA
| |
Collapse
|
14
|
Law IKM, Padua DM, Iliopoulos D, Pothoulakis C. Role of G protein-coupled receptors-microRNA interactions in gastrointestinal pathophysiology. Am J Physiol Gastrointest Liver Physiol 2017; 313:G361-G372. [PMID: 28774868 PMCID: PMC5792214 DOI: 10.1152/ajpgi.00144.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 01/31/2023]
Abstract
G protein-coupled receptors (GPCRs) make up the largest transmembrane receptor superfamily in the human genome and are expressed in nearly all gastrointestinal cell types. Coupling of GPCRs and their respective ligands activates various phosphotransferases in the cytoplasm, and, thus, activation of GPCR signaling in intestine regulates many cellular and physiological processes. Studies in microRNAs (miRNAs) demonstrate that they represent critical epigenetic regulators of different pathophysiological responses in different organs and cell types in humans and animals. Here, we reviewed recent research on GPCR-miRNA interactions related to gastrointestinal pathophysiology, such as inflammatory bowel diseases, irritable bowel syndrome, and gastrointestinal cancers. Given that the presence of different types of cells in the gastrointestinal tract suggests the importance of cell-cell interactions in maintaining gastrointestinal homeostasis, we also discuss how GPCR-miRNA interactions regulate gene expression at the cellular level and subsequently modulate gastrointestinal pathophysiology through molecular regulatory circuits and cell-cell interactions. These studies helped identify novel molecular pathways leading to the discovery of potential biomarkers for gastrointestinal diseases.
Collapse
Affiliation(s)
- Ivy Ka Man Law
- 1Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - David Miguel Padua
- 1Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - Dimitrios Iliopoulos
- 1Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and ,2Center for Systems Biomedicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Charalabos Pothoulakis
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and
| |
Collapse
|
15
|
Yang X, Yang L, Ma Y, Zhao X, Wang H. MicroRNA-205 Mediates Proteinase-Activated Receptor 2 (PAR 2) -Promoted Cancer Cell Migration. Cancer Invest 2017; 35:601-609. [PMID: 28990808 DOI: 10.1080/07357907.2017.1378671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Activation of proteinase-activated receptor 2 (PAR2) promotes cell migration in cancers, but the exact mechanism underlying this process remains largely unknown. Here we report that activation of PAR2 reduced miR-205 expression, whereas inhibition of miR-205 promoted cell migration in cancer cells. Overexpression of miR-205 blocked PAR2-mediated stimulation of cell migration. BMPR1B was identified as a downstream target gene of miR-205. In colorectal carcinoma specimens from patients, the level of PAR2 was negatively correlated with that of miR-205, but it was positively associated with BMPR1B expression. Taken together, our findings indicate that PAR2 signaling promotes cancer cell migration through miR-205/BMPR1B pathway in human colorectal carcinoma.
Collapse
Affiliation(s)
- Xu Yang
- a State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences , Peking Union Medical College , Beijing , China
| | - Lan Yang
- a State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences , Peking Union Medical College , Beijing , China
| | - Yiming Ma
- a State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences , Peking Union Medical College , Beijing , China
| | - Xinhua Zhao
- a State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences , Peking Union Medical College , Beijing , China
| | - Hongying Wang
- a State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences , Peking Union Medical College , Beijing , China
| |
Collapse
|
16
|
Aman M, Ohishi Y, Imamura H, Shinozaki T, Yasutake N, Kato K, Oda Y. Expression of protease-activated receptor-2 (PAR-2) is related to advanced clinical stage and adverse prognosis in ovarian clear cell carcinoma. Hum Pathol 2017; 64:156-163. [DOI: 10.1016/j.humpath.2017.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 01/17/2023]
|
17
|
Zhang L, Fang Y, Feng JY, Cai QY, Wei LH, Lin S, Peng J. Chloroform fraction of Scutellaria barbata D. Don inhibits the growth of colorectal cancer cells by activating miR‑34a. Oncol Rep 2017; 37:3695-3701. [PMID: 28498458 DOI: 10.3892/or.2017.5625] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/19/2017] [Indexed: 11/06/2022] Open
Abstract
Scutellaria barbata D. Don (SB) is a well known formula in traditional Chinese medicine, which exhibits potent anticancer effects on various cancers. Many miRNAs play crucial roles in the regulation of cancer, for instance, miR‑34a functions as a tumor suppressor, and is often downregulated during cancer. In this study, we investigated the role of ECSB in suppressing the growth of human colon cancer HCT‑8 cells, and whether this is mediated by regulation of miR‑34a and its downstream target genes, using real-time PCR and western blot analysis. ECSB treatment significantly inhibited the proliferation of HCT‑8 cells and promoted apoptosis in a dose-dependent manner. In addition, ECSB treatment significantly increased the level of miR‑34a expression and decreased the levels of Bcl-2, Notch1/2 and Jagged1 expression. Furthermore, knockdown of miR‑34a expression through transfection of anti-miR‑34a oligonucleotide was significantly reversed by ECSB treatment. Likewise, knockdown of miR‑34a resulted in significant upregulation of Bcl-2, Notch1/2 and Jagged1 expression, which was reversed following ECSB treatment. Therefore, this study reveals that ECSB inhibited cancer cell growth via promoting apoptosis and inhibiting proliferation, through regulation of miR‑34a. These findings further support the use of ECSB as an effective therapeutic agent against colon cancer.
Collapse
Affiliation(s)
- Ling Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yi Fang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jian-Yu Feng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Qiao-Yan Cai
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Li-Hui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
18
|
Yi R, Li Y, Wang FL, Miao G, Qi RM, Zhao YY. MicroRNAs as diagnostic and prognostic biomarkers in colorectal cancer. World J Gastrointest Oncol 2016; 8:330-40. [PMID: 27096028 PMCID: PMC4824711 DOI: 10.4251/wjgo.v8.i4.330] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 01/13/2016] [Accepted: 01/28/2016] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs (miRNAs) are key regulators involved in various tumors. They regulate cell cycle, apoptosis and cancer stemness, metastasis and chemoresistance by controlling their target gene expressions. Here, we mainly discuss the potential uses of miRNAs in colorectal cancer (CRC) diagnosis. We also shed light on the important corresponding miRNA targets and on the major regulators of miRNAs. Furthermore, we discuss miRNA activity in assessing the prognosis and recurrence of CRC as well as in modulating responsiveness to chemotherapy. Based on the various pro-oncogenic/anti-oncogenic roles of miRNAs, the advantages of a therapeutic strategy based on the delivery of miRNA mimics are also mentioned. Together, miRNA seems to be an excellent tool for effectively monitoring and targeting CRC.
Collapse
|
19
|
Chi Y, Zhou D. MicroRNAs in colorectal carcinoma--from pathogenesis to therapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:43. [PMID: 26964533 PMCID: PMC4787051 DOI: 10.1186/s13046-016-0320-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/07/2016] [Indexed: 12/12/2022]
Abstract
Background Acting as inflammatory mediators, tumor oncogenes or suppressors, microRNAs are involved in cell survival, death, epithelial–mesenchymal transition and metastasis, etc. Investigating the communication between microRNAs and tumorigenesis is critical to our understanding of the pathogenesis of multiple disease states. Main body Currently, colorectal carcinoma (CRC), one of the most common malignancies worldwide, has a poor prognosis due to lack of an effective therapeutic option. Increasing evidence has identified altered profiles and regulatory potential of microRNAs in conditions related to environmentally-caused colorectal inflammation and colitis-associated cancer. Many studies have shed light on a more thorough understanding of the function and distribution of microRNAs in CRC initiation and emergence. However, the molecular mechanisms by which microRNAs modulate cellular processes still need to be further elucidated and may offer a foundation for evaluating microRNA-based therapeutic potential for CRC in both animal models and clinical trials. Conclusion In this review, the roles and mechanisms of microRNAs involved in CRC from pathogenesis to therapy are summarized and discussed, which may provide more useful hints for CRC prevention and therapy.
Collapse
Affiliation(s)
- Yudan Chi
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Dongming Zhou
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
20
|
Vallabhaneni KC, Penfornis P, Dhule S, Guillonneau F, Adams KV, Mo YY, Xu R, Liu Y, Watabe K, Vemuri MC, Pochampally R. Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites. Oncotarget 2016; 6:4953-67. [PMID: 25669974 PMCID: PMC4467126 DOI: 10.18632/oncotarget.3211] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/27/2014] [Indexed: 12/20/2022] Open
Abstract
Human mesenchymal stem/stromal cells (hMSCs) have been shown to support breast cancer cell proliferation and metastasis, partly through their secretome. hMSCs have a remarkable ability to survive for long periods under stress, and their secretome is tumor supportive. In this study, we have characterized the cargo of extracellular vesicular (EV) fraction (that is in the size range of 40-150nm) of serum deprived hMSCs (SD-MSCs). Next Generation Sequencing assays were used to identify small RNA secreted in the EVs, which indicated presence of tumor supportive miRNA. Further assays demonstrated the role of miRNA-21 and 34a as tumor supportive miRNAs. Next, proteomic assays revealed the presence of ≈150 different proteins, most of which are known tumor supportive factors such as PDGFR-β, TIMP-1, and TIMP-2. Lipidomic assays verified presence of bioactive lipids such as sphingomyelin. Furthermore, metabolite assays identified the presence of lactic acid and glutamic acid in EVs. The co-injection xenograft assays using MCF-7 breast cancer cells demonstrated the tumor supportive function of these EVs. To our knowledge this is the first comprehensive -omics based study that characterized the complex cargo of extracellular vesicles secreted by hMSCs and their role in supporting breast cancers.
Collapse
Affiliation(s)
| | - Patrice Penfornis
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Santosh Dhule
- Department of Chemical and Biomolecular Engineering, New Orleans, LA, USA
| | - Francois Guillonneau
- 3P5 Proteomic Platform of the Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Kristen V Adams
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yin Yuan Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Rui Xu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS, USA
| | - Yiming Liu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS, USA
| | - Kounosuke Watabe
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mohan C Vemuri
- Stem Cell Biology, Thermo Fisher Scientific, Frederick, MD, USA
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
21
|
Yang Q, Diamond MP, Al-Hendy A. The emerging role of extracellular vesicle-derived miRNAs: implication in cancer progression and stem cell related diseases. JOURNAL OF CLINICAL EPIGENETICS 2016; 2:13. [PMID: 27099870 PMCID: PMC4834835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cells release into the extracellular environment, diverse types of membrane vesicles of endosomal and plasma membrane origin called exosomes and microvesicles. A number of studies indicate that these extracellular vehicles (EVs) mediate the interaction between cancer cells and their microenvironment; and thereby, play a critical role in the development of cancers. EVs contain cargo which consist of proteins, lipids, mRNAs, and miRNAs that can be delivered to different types of cells in nascent as well as distal locations. Discovery of this latter cargo has drawn an increasing amount of attention, due to their altering effects on the transcriptome, proteins, and subsequent cellular characteristics in recipient cells. Cancer cell derived exosomes (CCEs) have been identified in body fluids of cancer patients including urine, plasma and saliva. Because CCE content largely depends on tumor type and stage, they invariably lend great potential in serving as prognostic and diagnostic markers. Notably, accumulating evidence demonstrates that EV-derived miRNAs have key roles in regulating various aspects of cellular homeostasis, including proliferation, survival, migration, metastasis, and the immune system etc. More recently, diagnostic and therapeutic exploitation of stem cells derived EVs are under investigation. This review aims to summarize recent advances in EV-derived miRNAs in a variety of tumor types, and suggests that these cancer-derived exosomal miRNAs play a critical role in regulating cellular functions in surrounding and distant locations. It also discusses the role of adverse environmental exposure in altering stem cell exosomal miRNA profiling, which we believe leads to changes in the extracellular environment as well as a diverse range of biological processes.
Collapse
|
22
|
Yang L, Ma Y, Han W, Li W, Cui L, Zhao X, Tian Y, Zhou Z, Wang W, Wang H. Proteinase-activated receptor 2 promotes cancer cell migration through RNA methylation-mediated repression of miR-125b. J Biol Chem 2015; 290:26627-26637. [PMID: 26354435 PMCID: PMC4646319 DOI: 10.1074/jbc.m115.667717] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/05/2015] [Indexed: 01/08/2023] Open
Abstract
Proteinase activated-receptor 2 (PAR2) participates in cancer metastasis promoted by serine proteinases. The current study aimed to test the molecular mechanism by which PAR2 promotes cancer cell migration. In different cancer cells, activation of PAR2 by activating peptide (PAR2-AP) dramatically increased cell migration, whereas knock down of PAR2 inhibited cellular motility. The PAR2 activation also repressed miR-125b expression while miR-125b mimic successfully blocked PAR2-induced cell migration. Moreover, Grb associated-binding protein 2 (Gab2) was identified as a novel target gene of miR-125b and it mediated PAR2-induced cell migration. The correlation of PAR2 with miR-125b and Gab2 was further supported by the findings obtained from human colorectal carcinoma specimens. Remarkably, knock down of NOP2/Sun domain family, member 2 (NSun2), a RNA methyltransferase, blocked the reduction in miR-125b induced by PAR2. Furthermore, PAR2 activation increased the level of N(6)-methyladenosine (m(6)A)-containing pre-miR-125b in NSun2-dependent manner. Taken together, our results demonstrated that miR-125b mediates PAR2-induced cancer cell migration by targeting Gab2 and that NSun2-dependent RNA methylation contributes to the down-regulation of miR-125b by PAR2 signaling. These findings suggest a novel epigenetic mechanism by which microenvironment regulates cancer cell migration by altering miRNA expression.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Binding Sites
- Cell Line, Tumor
- Cell Movement/drug effects
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- HCT116 Cells
- HT29 Cells
- Humans
- Methylation/drug effects
- Methyltransferases/antagonists & inhibitors
- Methyltransferases/genetics
- Methyltransferases/metabolism
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Oligopeptides/pharmacology
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Protein Binding
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor, PAR-2/genetics
- Receptor, PAR-2/metabolism
- Signal Transduction
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Lan Yang
- From the State Key Laboratory of Molecular Oncology
| | - Yiming Ma
- From the State Key Laboratory of Molecular Oncology
| | - Wenxiao Han
- From the State Key Laboratory of Molecular Oncology
| | - Weiwei Li
- From the State Key Laboratory of Molecular Oncology
| | - Liang Cui
- Department of Abdominal Surgical Oncology, and
| | - Xinhua Zhao
- From the State Key Laboratory of Molecular Oncology
| | - Yantao Tian
- Department of Abdominal Surgical Oncology, and
| | - Zhixiang Zhou
- Department of Gastrointestinal Surgical Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 100021 Beijing, PR China and
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191 Beijing, PR China
| | - Hongying Wang
- From the State Key Laboratory of Molecular Oncology,
| |
Collapse
|
23
|
Li C, Wang Y, Lu S, Zhang Z, Meng H, Liang L, Zhang Y, Song B. MiR-34a inhibits colon cancer proliferation and metastasis by inhibiting platelet-derived growth factor receptor α. Mol Med Rep 2015; 12:7072-8. [PMID: 26324236 DOI: 10.3892/mmr.2015.4263] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 06/22/2015] [Indexed: 12/20/2022] Open
Abstract
The microRNA (miRNA), miR‑34a is significant in colon cancer progression. In the present study, the role of miR‑34a in colon cancer cell proliferation and metastasis was investigated. It was found that the expression of miR‑34a in colon cancer tissues and cell lines was lower when compared with that of normal tissues and cells. Further research demonstrated that miR‑34a inhibited cell proliferation, induced G1 phase arrest, and suppressed metastasis and epithelial mesenchymal transition in colon cancer cells. Bioinformatic prediction indicated that platelet‑derived growth factor receptor α (PDGFRA) was a potential target gene of miR‑34a and a luciferase assay identified that PDGFRA was a novel direct target gene of miR‑34a. In addition, assays of western blot analyses and quantitative reverse‑transcription polymerase chain reaction confirmed that miR‑34a decreased PDGFRA mRNA expression and protein levels in colon cancer cells. Assessment of cellular function indicated that miR‑34a inhibited colon cancer progression via PDGFRA. These findings demonstrate that miR‑34a may act as a negative regulator in colon cancer by targeting PDGFRA.
Collapse
Affiliation(s)
- Chunyan Li
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yulin Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Shuming Lu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhuqing Zhang
- Department of Pathology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Hua Meng
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lina Liang
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Bo Song
- Department of Pathology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
24
|
Zhang H, Li S, Yang J, Liu S, Gong X, Yu X. The prognostic value of miR-34a expression in completely resected gastric cancer: tumor recurrence and overall survival. Int J Clin Exp Med 2015; 8:2635-2641. [PMID: 25932212 PMCID: PMC4402859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 01/29/2015] [Indexed: 06/04/2023]
Abstract
The prognosis of gastric cancer is mainly linked to tumor recurrence. MicroRNA-34a (miR-34a) is a direct transcriptional target of p53 and links tumor suppressor function and the oncogenic pathways in some cancers. However, the role of miR-34a in predicting prognosis of gastric cancer has not been fully elucidated. In this study, we aimed to investigate the expression level of miR-34a and its prognostic value in gastric cancer. A total of 137 consecutive gastric cancer patients who underwent gastrectomy with D2 lymph node dissection were included in this study. Quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to detect miR-34a expression in gastric cancer tissues and adjacent normal tissues. The results showed that the levels of miR-34a expression were significantly decreased in the tumor tissues compared with the adjacent normal tissues (P<0.001). Low miR-34a expression level was associated with lymph node involvement (P=0.004), advanced TNM stage (P=0.006), poor tumor differentiation (P=0.024), high tumor recurrence rate (P=0.008), and poor five-year survival (P<0.001). The median time to recurrence and median overall survival time were significantly shorter in patients with low miR-34a levels compared with those with high miR-34a levels (P=0.028 and P=0.021, respectively). Furthermore, when analyzed with a multivariate Cox regression model, a low miR-34a level was significantly correlated with high recurrence rate and poor overall survival. Taken together, our results suggest that downregulation of miR-34a in gastric cancer is associated with high recurrence rate and poor overall survival and that miR-34a may be served as a prognostic marker for gastric cancer.
Collapse
Affiliation(s)
- Huaping Zhang
- Department of Gastrointestinal Surgery, Hepatobiliary Surgery and Thoracic Surgery, Yuhuangding HospitalYantai 264000, Shandong, China
| | - Shaojun Li
- Department of Gastrointestinal Surgery, Hepatobiliary Surgery and Thoracic Surgery, Yuhuangding HospitalYantai 264000, Shandong, China
| | - Junqi Yang
- Qingdao University School of MedicineQingdao 266021, Shandong, China
| | - Shuai Liu
- Qingdao University School of MedicineQingdao 266021, Shandong, China
| | - Xiangqian Gong
- Department of Gastrointestinal Surgery, Hepatobiliary Surgery and Thoracic Surgery, Yuhuangding HospitalYantai 264000, Shandong, China
| | - Xiaofeng Yu
- Department of Gastrointestinal Surgery, Hepatobiliary Surgery and Thoracic Surgery, Yuhuangding HospitalYantai 264000, Shandong, China
| |
Collapse
|
25
|
Montella M, D'Arena G, Crispo A, Capunzo M, Nocerino F, Grimaldi M, Barbieri A, D'Ursi AM, Tecce MF, Amore A, Galdiero M, Ciliberto G, Giudice A. Role of Sex Hormones in the Development and Progression of Hepatitis B Virus-Associated Hepatocellular Carcinoma. Int J Endocrinol 2015; 2015:854530. [PMID: 26491442 PMCID: PMC4600563 DOI: 10.1155/2015/854530] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 12/25/2022] Open
Abstract
Infection with hepatitis B virus (HBV) is a major risk factor for hepatocellular carcinoma (HCC) in developed countries. Epidemiological reports indicate that the incidence of HBV-related HCC is higher in males and postmenopausal females than other females. Increasing evidence suggests that sex hormones such as androgens and estrogens play an important role in the progression of an HBV infection and in the development of HBV-related HCC. While androgen is supposed to stimulate the androgen signaling pathway and cooperate to the increased transcription and replication of HBV genes, estrogen may play a protecting role against the progression of HBV infections and in the development of HBV-related HCC through decreasing HBV RNA transcription and inflammatory cytokines levels. Additionally, sex hormones can also affect HBV-related hepatocarcinogenesis by inducing epigenetic changes such as the regulation of mRNA levels by microRNAs (miRNAs), DNA methylation, and histone modification in liver tissue. This review describes the molecular mechanisms underlying the gender disparity in HBV-related HCC with the aim of improving the understanding of key factors underneath the sex disparity often observed in HBV infections. Furthermore, the review will propose more effective prevention strategies and treatments of HBV-derived diseases.
Collapse
Affiliation(s)
- Maurizio Montella
- Epidemiology Unit, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
- *Maurizio Montella:
| | - Giovanni D'Arena
- Department of Onco-Hematology, IRCCS, Cancer Referral Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Anna Crispo
- Epidemiology Unit, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| | - Mario Capunzo
- Department of Medicine and Surgery, University of Salerno, 84081 Fisciano, Italy
| | - Flavia Nocerino
- Epidemiology Unit, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| | - Maria Grimaldi
- Epidemiology Unit, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| | - Antonio Barbieri
- Animal Facility, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| | - Anna Maria D'Ursi
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Salerno, Italy
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Salerno, Italy
| | - Alfonso Amore
- Department of Surgery, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| | | | - Gennaro Ciliberto
- National Cancer Institute “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| | - Aldo Giudice
- Epidemiology Unit, National Cancer Institute of Naples “G. Pascale Foundation”, IRCCS, 80131 Naples, Italy
| |
Collapse
|
26
|
Kularathna PK, Pagel CN, Mackie EJ. Tumour progression and cancer-induced pain: a role for protease-activated receptor-2? Int J Biochem Cell Biol 2014; 57:149-56. [PMID: 25448411 DOI: 10.1016/j.biocel.2014.10.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/23/2014] [Indexed: 02/08/2023]
Abstract
The role of proteases in modifying the microenvironment of tumour cells has long been recognised. With the discovery of the protease-activated receptor family of G protein-coupled receptors a mechanism for cells to sense and respond directly to proteases in their microenvironment was revealed. Many early studies described the roles of protease-activated receptors in the cellular events that occur during blood coagulation and inflammation. More recently, studies have begun to focus on the roles of protease-activated receptors in the establishment, progression and metastasis of a variety of tumours. This review will focus on the expression of protease-activated receptor-2 and its activators by normal and neoplastic tissues, and describe current evidence that activation of protease-activated receptor-2 is an important event at multiple stages of tumour progression and in pain associated with cancer.
Collapse
Affiliation(s)
- Pamuditha K Kularathna
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Charles N Pagel
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Eleanor J Mackie
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
27
|
Iablokov V, Hirota CL, Peplowski MA, Ramachandran R, Mihara K, Hollenberg MD, MacNaughton WK. Proteinase-activated receptor 2 (PAR2) decreases apoptosis in colonic epithelial cells. J Biol Chem 2014; 289:34366-77. [PMID: 25331954 DOI: 10.1074/jbc.m114.610485] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mucosal biopsies from inflamed colon of inflammatory bowel disease patients exhibit elevated epithelial apoptosis compared with those from healthy individuals, disrupting mucosal homeostasis and perpetuating disease. Therapies that decrease intestinal epithelial apoptosis may, therefore, ameliorate inflammatory bowel disease, but treatments that specifically target apoptotic pathways are lacking. Proteinase-activated receptor-2 (PAR2), a G protein-coupled receptor activated by trypsin-like serine proteinases, is expressed on intestinal epithelial cells and stimulates mitogenic pathways upon activation. We sought to determine whether PAR2 activation and signaling could rescue colonic epithelial (HT-29) cells from apoptosis induced by proapoptotic cytokines that are increased during inflammatory bowel disease. The PAR2 agonists 2-furoyl-LIGRLO (2f-LI), SLIGKV and trypsin all significantly reduced cleavage of caspase-3, -8, and -9, poly(ADP-ribose) polymerase, and the externalization of phosphatidylserine after treatment of cells with IFN-γ and TNF-α. Knockdown of PAR2 with siRNA eliminated the anti-apoptotic effect of 2f-LI and increased the sensitivity of HT-29 cells to cytokine-induced apoptosis. Concurrent inhibition of both MEK1/2 and PI3K was necessary to inhibit PAR2-induced survival. 2f-LI was found to increase phosphorylation and inactivation of pro-apoptotic BAD at Ser(112) and Ser(136) by MEK1/2 and PI3K-dependent signaling, respectively. PAR2 activation also increased the expression of anti-apoptotic MCL-1. Simultaneous knockdown of both BAD and MCL-1 had minimal effects on PAR2-induced survival, whereas single knockdown had no effect. We conclude that PAR2 activation reduces cytokine-induced epithelial apoptosis via concurrent stimulation of MEK1/2 and PI3K but little involvement of MCL-1 and BAD. Our findings represent a novel mechanism whereby serine proteinases facilitate epithelial cell survival and may be important in the context of colonic healing.
Collapse
Affiliation(s)
- Vadim Iablokov
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| | - Christina L Hirota
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| | - Michael A Peplowski
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| | - Rithwik Ramachandran
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| | - Koichiro Mihara
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| | - Morley D Hollenberg
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| | - Wallace K MacNaughton
- From the Department of Physiology and Pharmacology, the Inflammation Research Network, and the Snyder Institute for Chronic Diseases, University of Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
28
|
Misso G, Di Martino MT, De Rosa G, Farooqi AA, Lombardi A, Campani V, Zarone MR, Gullà A, Tagliaferri P, Tassone P, Caraglia M. Mir-34: a new weapon against cancer? MOLECULAR THERAPY-NUCLEIC ACIDS 2014; 3:e194. [PMID: 25247240 PMCID: PMC4222652 DOI: 10.1038/mtna.2014.47] [Citation(s) in RCA: 390] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 08/11/2014] [Indexed: 02/07/2023]
Abstract
The microRNA(miRNA)-34a is a key regulator of tumor suppression. It controls the
expression of a plethora of target proteins involved in cell cycle, differentiation
and apoptosis, and antagonizes processes that are necessary for basic cancer cell
viability as well as cancer stemness, metastasis, and chemoresistance. In this
review, we focus on the molecular mechanisms of miR-34a-mediated tumor suppression,
giving emphasis on the main miR-34a targets, as well as on the principal regulators
involved in the modulation of this miRNA. Moreover, we shed light on the miR-34a role
in modulating responsiveness to chemotherapy and on the phytonutrients-mediated
regulation of miR-34a expression and activity in cancer cells. Given the broad
anti-oncogenic activity of miR-34a, we also discuss the substantial benefits of a new
therapeutic concept based on nanotechnology delivery of miRNA mimics. In fact, the
replacement of oncosuppressor miRNAs provides an effective strategy against tumor
heterogeneity and the selective RNA-based delivery systems seems to be an excellent
platform for a safe and effective targeting of the tumor.
Collapse
Affiliation(s)
- Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University "Federico II" of Naples, Naples, Italy
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | - Angela Lombardi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Virginia Campani
- Department of Pharmacy, University "Federico II" of Naples, Naples, Italy
| | - Mayra Rachele Zarone
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierfrancesco Tassone
- 1] Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy [2] Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | - Michele Caraglia
- 1] Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy [2] Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Jasinski-Bergner S, Mandelboim O, Seliger B. The role of microRNAs in the control of innate immune response in cancer. J Natl Cancer Inst 2014; 106:dju257. [PMID: 25217579 DOI: 10.1093/jnci/dju257] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ligands for receptors of natural killer (NK) cells and CD8(+) cytotoxic T lymphocytes (CTL), such as the inhibitory nonclassical HLA-G, the activating stress-induced major histocompatibility complex class I-related antigens MICA and MICB, and/or the UL16-binding proteins (ULBPs), are often aberrantly expressed upon viral infection and neoplastic transformation, thereby preventing virus-infected or malignant-transformed cells from elimination by immune effector cells. Recently, it has been shown that ligands of both NK and CD8(+) T cells are regulated by a number of cellular and/or viral microRNAs (miRs). These miRs are involved in shaping the antiviral and/or antitumoral immune responses as well as neoplastic growth properties. This review summarizes the expression pattern and function of miRs directed against selected NK and T cell receptor ligands, their putative role in shaping immune surveillance and tumorigenicity, and their clinical relevance. In addition, the potential role of RNA-binding proteins in the post-transcriptional gene regulation of these ligands will be discussed.
Collapse
Affiliation(s)
- Simon Jasinski-Bergner
- Martin-Luther-University Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany (SJB, BS); The Hebrew University of Jerusalem, Ein Kerem, The Lautenberg Center for General and Tumor Immunology, IMRIC, Jerusalem, Israel (OM)
| | - Ofer Mandelboim
- Martin-Luther-University Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany (SJB, BS); The Hebrew University of Jerusalem, Ein Kerem, The Lautenberg Center for General and Tumor Immunology, IMRIC, Jerusalem, Israel (OM)
| | - Barbara Seliger
- Martin-Luther-University Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany (SJB, BS); The Hebrew University of Jerusalem, Ein Kerem, The Lautenberg Center for General and Tumor Immunology, IMRIC, Jerusalem, Israel (OM).
| |
Collapse
|
30
|
MicroRNA-34A inhibits the growth, invasion and metastasis of gastric cancer by targeting PDGFR and MET expression. Biosci Rep 2014; 34:BSR20140020. [PMID: 24837198 PMCID: PMC4069683 DOI: 10.1042/bsr20140020] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Within the family of RTKs (receptor tyrosine kinases), PDGFR (platelet-derived growth factor receptor) has been implicated in carcinogenesis and tumour development. miRNAs (microRNAs), which can target the mRNAs (messenger RNAs) of cancer-associated genes, are abnormally expressed in various cancers. In this study, our aim was to identify the miRNAs that target PDGFR-α/β and to study the functions of these miRNAs. miR-34a was predicted to target PDGFR, and luciferase reporter assays showed that miR-34a could directly target PDGFR. Meanwhile, we found that miR-34a was down-regulated in gastric cancer tissues and was associated with metastasis. Our findings showed that miR-34a could inhibit gastric cancer cell migration, invasion and proliferation, but these tumourigenic properties were only partially restored when PDGFR-α/β was overexpressed. In subsequent experiments, we found that the overexpression of both PDGFR and MET could completely restore the gastric cancer tumourigenic properties. Moreover, the cancer-associated cell signalling pathway was studied, and we found that miR-34a could inhibit Akt [PKB (protein kinase B)] phosphorylation, which was restored by the overexpression of both PDGFR and MET. In conclusion, miR-34a may act as a potential tumour suppressor in gastric cancer and is associated with the mechanisms of gastric cancer metastasis; miR-34a can inhibit gastric cancer tumourigenesis by targeting PDGFR and MET through the PI3K (phosphoinositide 3-kinase)/Akt pathway.
Collapse
|
31
|
Cai WS, Shen F, Li JL, Feng Z, Wang YC, Xiao HQ, Xu B. Activated protease receptor-2 induces GATA6 expression to promote survival in irradiated colon cancer cells. Arch Biochem Biophys 2014; 555-556:28-32. [PMID: 24887481 DOI: 10.1016/j.abb.2014.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 05/20/2014] [Accepted: 05/21/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND AIMS The resistance to irradiation is common and a great drawback in the treatment of cancer with radiotherapy; the underlying mechanism is unclear. GATA binding protein 6 (GATA6) is associated with the pathogenesis of cancer. This study aims to investigate the role of GATA6 on compromising irradiation effect on HT55 and HT29 cells, 2 colorectal cancer cell lines. METHODS Human colon cancer cell lines, HT55 and HT29 cells, were treated with irradiation in the culture. Apoptosis of HT55 and HT29 cells was determined by flow cytometry. The expression of PAR2 and GATA6 in HT55 and HT29 cells was analyzed by real time RT-PCR and Western blotting. The gene silence and gene over expression were employed to observe the effect of GATA6 on p53 expression in HT55 and HT29 cells. RESULTS The results showed that HT55 and HT29 cells expressed protease-activated receptor-2 (PAR2). Irradiation induced 38.6% HT55 cell and 33.8% HT29 cell apoptosis, which reduced to 4.2% and 5.6%, respectively after activation of PAR2. Exposure to irradiation increased the expression of GATA6; the latter played a critical role in suppression of p53 expression in HT55 and HT29 cells. Inhibition of GATA6 significantly increased the effect of irradiation on HT55 and HT29 cells. CONCLUSIONS Activation of PAR2 compromises the effect of irradiation on inducing colorectal cancer cell apoptosis, which can be prevented by inhibition of GATA6 expression.
Collapse
Affiliation(s)
- Wen-song Cai
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fei Shen
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiang-lin Li
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhe Feng
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ying-cheng Wang
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huan-qing Xiao
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bo Xu
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
32
|
Liu WC, Liu QY. Molecular mechanisms of gender disparity in hepatitis B virus-associated hepatocellular carcinoma. World J Gastroenterol 2014; 20:6252-6261. [PMID: 24876746 PMCID: PMC4033463 DOI: 10.3748/wjg.v20.i20.6252] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 12/22/2013] [Accepted: 01/08/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic hepatitis B virus (HBV) infection is one of the most common causes of hepatocellular carcinoma (HCC), a malignant tumor with high mortality worldwide. One remarkable clinical feature of HBV-related HCC is that its incidence is higher in males and postmenopausal females compared to other females. Increasing evidence indicates that HBV-associated HCC may involve gender disparity and that it may be a type of hormone-responsive malignant tumor. Sex hormones, such as androgen and estrogen, have been shown to play very different roles in the progression of an HBV infection and in the development of HBV-related HCC. Through binding to their specific cellular receptors and affecting the corresponding signaling pathways, sex hormones can regulate the transactivation of HBx, cause the chronic release of inflammatory cytokines in the hepatocellular microenvironment, and participate in epigenetic and genetic alternations in hepatocytes. All of these functions may be related to the initiation and progression of HBV-associated HCC. A thorough investigation of the molecular mechanisms underlying the gender-related disparity in HBV-related HCC should provide a new perspective for better understanding its pathogenesis and exploring more effective methods for the prevention and treatment of this disease.
Collapse
|
33
|
Jiang DS, Wang YW, Jiang J, Li SM, Liang SZ, Fang HY. MicroRNA-26a involved in Toll-like receptor 9‑mediated lung cancer growth and migration. Int J Mol Med 2014; 34:307-12. [PMID: 24788552 DOI: 10.3892/ijmm.2014.1764] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/08/2014] [Indexed: 11/06/2022] Open
Abstract
Toll-like receptor 9 (TLR9) has been shown to have a significant role in cancer. MicroRNAs (miRNAs), a group of small non-coding RNAs that fine tune translation of multiple target mRNAs, are involved in the development and progression of human cancers. The present study was undertaken to determine the roles of TLR9 on lung cancer and whether miR-26a is involved in TLR9‑mediated lung cancer growth and migration. The lung cancer models were elicited by injecting human lung cancer cells into the left ventricle. The expression of TLR9 and miR-26a in lung cancer tissues obtained from lung cancer patients was increased. TLR9 ligand CpG-oligodeoxynucleotides (CpG-ODN) caused an increase in the mean tumor weight and the size of tumor mass in nude mice, and the proliferation and migration of H460 human lung cancer cells. CpG-ODN also induced an increase in the expression of miR-26a in H460 cells. The overexpression of miR-26a increased the weight and size of the tumor mass in the nude mice, and the proliferation and migration of H460 cells. Expression of phosphoinositide 3 kinase (PI3K) and phosphorylation of protein kinase B (Akt) was increased after miR-26a overexpression in the H460 cells. PI3K inhibitor wortmannin (WM) or Akt inhibitor triciribine hydrate (TCN) eliminated the increase in the proliferation and migration induced by the overexpression of miR-26a in H460 cells. These results suggested that miR-26a is involved in the TLR9‑mediated growth and migration of lung cancer through the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- De-Sheng Jiang
- Department of Respiratory Medicine, The 454 Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| | - Yu-Wei Wang
- Department of Respiratory Medicine, The 454 Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| | - Jing Jiang
- Department of Respiratory Medicine, The 454 Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| | - Shu-Meng Li
- Department of Respiratory Medicine, The 454 Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| | - Shun-Zhi Liang
- Department of Respiratory Medicine, The 454 Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| | - Hong-Yan Fang
- Department of Respiratory Medicine, The 454 Hospital of PLA, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
34
|
Wang X, Kuang YY, Hu XT. Advances in epigenetic biomarker research in colorectal cancer. World J Gastroenterol 2014; 20:4276-4287. [PMID: 24764665 PMCID: PMC3989963 DOI: 10.3748/wjg.v20.i15.4276] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 11/05/2013] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) causes approximately 600000 deaths annually and is the third leading cause of cancer mortality worldwide. Despite significant advancements in treatment options, CRC patient survival is still poor owing to a lack of effective tools for early diagnosis and a limited capacity for optimal therapeutic decision making. Since there exists a need to find new biomarkers to improve diagnosis of CRC, the research on epigenetic biomarkers for molecular diagnostics encourages the translation of this field from the bench to clinical practice. Epigenetic alterations are thought to hold great promise as tumor biomarkers. In this review, we will primarily focus on recent advances in the study of epigenetic biomarkers for colorectal cancer and discuss epigenetic biomarkers, including DNA methylation, microRNA expression and histone modification, in cancer tissue, stool, plasma, serum, cell lines and xenografts. These studies have improved the chances that epigenetic biomarkers will find a place in the clinical practices of screening, early diagnosis, prognosis, therapy choice and recurrence surveillance for CRC patients. However, these studies have typically been small in size, and evaluation at a larger scale of well-controlled randomized clinical trials is the next step that is necessary to increase the quality of epigenetic biomarkers and ensure their widespread clinical use.
Collapse
|
35
|
|