51
|
Rajagopal P, Chellappan DR, Sridharan S, Pemiah B, Krishnaswamy S, Sethuraman S, Sekar K, Krishnan UM. Microarray analysis of genes from animals treated with a traditional formulation ChandraprabhaVati reveals its therapeutic targets. J Tradit Complement Med 2019; 10:36-44. [PMID: 31956556 PMCID: PMC6957807 DOI: 10.1016/j.jtcme.2019.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 07/02/2019] [Accepted: 08/01/2019] [Indexed: 02/01/2023] Open
Abstract
Background Traditional medicinal preparations have not received global acceptance, and their therapeutic benefits remain disputed due to lack of scientific evidence on their mechanism of action. Microarray analysis has emerged as a powerful technique that can aid in understanding the complex signaling networks activated by these formulations and thereby assess their beneficial as well as adverse effects. Aim The present work aims to investigate the differential influence of ChandraprabhaVati, Ayurvedic formulation used in the treatment of diabetes, anemia, urinary, respiratory, skin and liver disorders. Materials and methods The RNA from the liver of rats treated with different doses of ChandraprabhaVati for 28 days was isolated and studied for the genome-wide changes in the expression. Results The results revealed several molecular targets that could contribute to the therapeutic effects of ChandraprabhaVati. Several genes have been differentially expressed, among those miRNAs miR-434, miR877, and miRlet7e contribute to the anti-diabetic, anti-fibrotic and anti-inflammatory of CPV. The rejuvenative activity of CPV may be due to the MeOX1 and Upf3b genes. Up-regulation of Hbaa2 gene facilitates the anti-anemic effect. Interestingly gender-specific differential expressions of genes were also observed. Rab3d were found to be altered in female when compared to male animals. Conclusion Thus the microarray data for the CPV treated animals has revealed molecular targets that may be responsible for the various known therapeutic effects and also identified new beneficial effects of CPV.
Collapse
Affiliation(s)
- Pratheppa Rajagopal
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - David Raj Chellappan
- Centre for Advanced Research in Indian Systems of Medicine, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - Sriram Sridharan
- Centre for Advanced Research in Indian Systems of Medicine, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - Brindha Pemiah
- Centre for Advanced Research in Indian Systems of Medicine, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - Sridharan Krishnaswamy
- School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - KalpoondiRajan Sekar
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India.,School of Chemical & Biotechnology, SASTRA Deemed-to-be University, Thanjavur, 613 401, Tamil Nadu, India
| |
Collapse
|
52
|
Liu S, Zhan Y, Luo J, Feng J, Lu J, Zheng H, Wen Q, Fan S. Roles of exosomes in the carcinogenesis and clinical therapy of non-small cell lung cancer. Biomed Pharmacother 2019; 111:338-346. [DOI: 10.1016/j.biopha.2018.12.088] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/06/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
|
53
|
Weston WW, Ganey T, Temple HT. The Relationship between Exosomes and Cancer: Implications for Diagnostics and Therapeutics. BioDrugs 2019; 33:137-158. [DOI: 10.1007/s40259-019-00338-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
54
|
Ren Y, Shi G, Jiang P, Meng Q. MicroRNA-761 is downregulated in colorectal cancer and regulates tumor progression by targeting Rab3D. Exp Ther Med 2018; 17:1841-1846. [PMID: 30783458 DOI: 10.3892/etm.2018.7126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 11/11/2018] [Indexed: 01/07/2023] Open
Abstract
The purpose of the present study was to investigate the biological role of microRNA-761 (miR-761) in colorectal cancer (CRC) and the underlying mechanisms by which miR-761 regulates CRC cell proliferation and migration. Quantitative polymerase chain reaction was performed to measure miR-761 expression in CRC tumor tissues and cell lines. It was demonstrated that miR-761 expression was dramatically reduced in CRC tumor tissues and cell lines compared with in normal tissues and cell lines. Overexpression of miR-761 significantly decreased CRC cell growth and migration. Using bioinformatics analysis and luciferase reporter assays, Rab3D was identified as a novel target of miR-761. In addition, it was demonstrated that Rab3D expression was negatively correlated with miR-761. Furthermore, overexpression of Rab3D could reverse the inhibitory effects of miR-761 on cell proliferation and migration. Collectively, the present study demonstrated that miR-761 overexpression could inhibit the proliferation and migration of CRC cell lines, partly at least, via directly targeting Rab3D.
Collapse
Affiliation(s)
- Yupeng Ren
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Gang Shi
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Peng Jiang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Qingkai Meng
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
55
|
Role of tumor-derived exosomes in cancer metastasis. Biochim Biophys Acta Rev Cancer 2018; 1871:12-19. [PMID: 30419312 DOI: 10.1016/j.bbcan.2018.10.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
The highlights of cancer research include the discovery of exosomes, which are small (30-100 nm) sized vesicular nanoparticles released virtually by all cells. Tumor-derived exosomes (TDEs) are notoriously known for orchestrating the invasion-metastasis cascade via systemic pathways that we have previously proposed (1), resulting in a paradigm shift of our understanding about the pathobiology of metastases. In principle, exosomes serve as transport medium for proteins, mRNAs and miRNAs to transmit targeted cues from the primary cell to distant sites via horizontal transfer or cell-receptor interaction. In this chapter, we seek to explore in-depth the mechanisms engendering TDE in the metastatic cascade, along with experimental models to augment our understanding. The aforementioned has also paved way for parallel advancements in the therapeutic armamentarium, as evident from pronounced efforts to exploit the metastatic process for therapeutic targeting. In this light, we aim to examine potential anti-metastatic therapeutic opportunities derived from exosomal research. Lastly, exosomes may play a crucial role in the contemporary era of "liquid biopsies", given the array of molecular information with diagnostic and predictive indications. We thus intend to end this chapter off by exploring future applications of exosomes that could illuminate shortcomings and propel advancements in biomarker research.
Collapse
|
56
|
Li D, Qian X, Xu P, Wang X, Li Z, Qian J, Yao J. Identification of lncRNAs and Their Functional Network Associated with Chemoresistance in SW1990/GZ Pancreatic Cancer Cells by RNA Sequencing. DNA Cell Biol 2018; 37:839-849. [PMID: 30113217 DOI: 10.1089/dna.2018.4312] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Duguang Li
- The Second Clinical College of Dalian Medical University, Dalian, China
| | - Xiaowei Qian
- Yangzhou University Medical College, Yangzhou, China
| | - Peng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Xiaodong Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Zhennan Li
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Jianjun Qian
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| |
Collapse
|
57
|
Zheng H, Zhan Y, Liu S, Lu J, Luo J, Feng J, Fan S. The roles of tumor-derived exosomes in non-small cell lung cancer and their clinical implications. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:226. [PMID: 30217217 PMCID: PMC6137883 DOI: 10.1186/s13046-018-0901-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 85% of lung cancer cases, and it is one of the leading causes of cancer death in both men and women worldwide due to diagnosis in the advanced stage, rapid metastasis, and recurrence. At present, precision molecular targeted therapeutics directed toward NSCLC driven genes has made great progress and significantly improved the overall survival of patients with NSCLC, but can easily lead to acquired drug resistance. New methods are needed to develop real-time monitoring of drug efficacy and drug resistance, such as new molecular markers for more effective early detection and prediction of prognosis. Exosomes are nano-sized extracellular vesicles, containing proteins, nucleic acids and lipids, which are secreted by various cells, and they play an important role in the development of lung cancer by controlling a wide range of pathways. Tumor-derived exosomes are of great significance for guiding the targeted therapy of NSCLC and exosomes themselves can be a target for treatment. In this review, we describe the potential roles of tumor-derived exosomes and their clinical significance in NSCLC.
Collapse
Affiliation(s)
- Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Sile Liu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Junmi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Juan Feng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
58
|
Cho SH, Kuo IY, Lu PJF, Tzeng HT, Lai WW, Su WC, Wang YC. Rab37 mediates exocytosis of secreted frizzled-related protein 1 to inhibit Wnt signaling and thus suppress lung cancer stemness. Cell Death Dis 2018; 9:868. [PMID: 30158579 PMCID: PMC6115395 DOI: 10.1038/s41419-018-0915-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/27/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023]
Abstract
Recent studies have revealed that dysregulated Rab small GTPase-mediated vesicle trafficking pathways are associated with cancer progression. However, whether any of the Rabs plays a suppressor role in cancer stemness is least explored. Rab37 has been postulated as a tumor suppressive small GTPase for trafficking anti-tumor cargos. Here, we report a previously uncharacterized mechanism by which Rab37 mediates exocytosis of secreted frizzled-related protein-1 (SFRP1), an extracellular antagonist of Wnt, to suppress Wnt signaling and cancer stemness in vitro and in vivo. Reconstitution experiments indicate that SFRP1 secretion is crucial for Rab37-mediated cancer stemness suppression and treatment with SRPP1 recombinant protein reduces xenograft tumor initiation ability. Clinical results confirm that concordantly low Rab37, low SFRP1, and high Oct4 stemness protein expression profile can be used as a biomarker to predict poor prognosis in lung cancer patients. Our findings reveal that Rab37-mediated SFRP1 secretion suppresses cancer stemness, and dysregulated Rab37-SFRP1 pathway confers cancer stemness via the activation of Wnt signaling. Rab37-SFRP1-Wnt axis could be a potential therapeutic target for attenuating lung cancer stemness.
Collapse
Affiliation(s)
- Shu-Huei Cho
- Department of Pharmacology, National Cheng Kung University, Tainan 701, Tainan City, Taiwan
| | - I-Ying Kuo
- Department of Pharmacology, National Cheng Kung University, Tainan 701, Tainan City, Taiwan
| | - Pei-Jung Frank Lu
- Institute of Clinical Medicine, National Cheng Kung University, Tainan 701, Tainan City, Taiwan
| | - Hong-Tai Tzeng
- Department of Pharmacology, National Cheng Kung University, Tainan 701, Tainan City, Taiwan
| | - Wu-Wei Lai
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University, Tainan 701, Tainan City, Taiwan
| | - Wu-Chou Su
- Division of Oncology, Department of Internal Medicine, National Cheng Kung University, Tainan 701, Tainan City, Taiwan
| | - Yi-Ching Wang
- Department of Pharmacology, National Cheng Kung University, Tainan 701, Tainan City, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Tainan City, Taiwan.
| |
Collapse
|
59
|
Tzeng H, Su C, Chang C, Lai W, Su W, Wang Y. Rab37 in lung cancer mediates exocytosis of soluble ST2 and thus skews macrophages toward tumor‐suppressing phenotype. Int J Cancer 2018; 143:1753-1763. [DOI: 10.1002/ijc.31569] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/03/2018] [Accepted: 04/24/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Hong‐Tai Tzeng
- Department of PharmacologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan Taiwan
| | - Ching‐Chin Su
- Department of PharmacologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan Taiwan
| | - Chih‐Peng Chang
- Department of Microbiology and ImmunologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan Taiwan
| | - Wu‐Wei Lai
- Division of Thoracic Surgery, Department of SurgeryNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan Taiwan
| | - Wu‐Chou Su
- Division of Oncology, Department of Internal MedicineNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan Taiwan
| | - Yi‐Ching Wang
- Department of PharmacologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan Taiwan
| |
Collapse
|
60
|
Sun W, Luo JD, Jiang H, Duan DD. Tumor exosomes: a double-edged sword in cancer therapy. Acta Pharmacol Sin 2018; 39:534-541. [PMID: 29542685 PMCID: PMC5888693 DOI: 10.1038/aps.2018.17] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 02/24/2018] [Indexed: 12/15/2022]
Abstract
Tumor cells produce and secrete more nucleic acids, proteins and lipids than normal cells. These molecules are transported in the blood or around the cells in membrane-encapsulated exosomes. Tumor-derived or tumor-associated exosomes (usually 30-100 nm in diameter) contain abundant biological contents resembling those of the parent cells along with signaling messengers for intercellular communication involved in the pathogenesis, development, progression, and metastasis of cancer. As these exosomes can be detected and isolated from various body fluids, they have become attractive new biomarkers for the diagnosis and prognosis of cancer. Furthermore, tumor exosomes have also attracted increasing attention due to their potential as novel therapeutic strategies for the treatment of cancers. On the one hand, the lipid bilayer membrane-encapsulated vesicles are promising carriers of drugs and other therapeutic materials targeting specific cancer cells. On the other hand, tumor exosomes are important mediators for modulation of the microenvironment that orchestrates events critical to the growth and metastasis of cancer cells as well as chemoresistance. Here, we summarize the advances in our understanding of tumor-associated or tumor-derived exosomes in recent years, and discuss their roles in cancer development, progression, invasion, and metastasis of cancers and, more importantly, their potential in strategies for precision therapy of various cancers as well as important caveats.
Collapse
Affiliation(s)
- Wei Sun
- Department of Oncology, Changzhou Second People's Hospital, Changzhou 213003, China
| | - Ju-dong Luo
- Department of Oncology, Changzhou Second People's Hospital, Changzhou 213003, China
| | - Hua Jiang
- Department of Oncology, Changzhou Second People's Hospital, Changzhou 213003, China
| | - Dayue Darrel Duan
- Laboratory of Cardiovascular Phenomics, Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| |
Collapse
|
61
|
Banworth MJ, Li G. Consequences of Rab GTPase dysfunction in genetic or acquired human diseases. Small GTPases 2018. [PMID: 29239692 DOI: 10.1080/215412481397833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023] Open
Abstract
Rab GTPases are important regulators of intracellular membrane trafficking in eukaryotes. Both activating and inactivating mutations in Rab genes have been identified and implicated in human diseases ranging from neurological disorders to cancer. In addition, altered Rab expression is often associated with disease prognosis. As such, the study of diseases associated with Rabs or Rab-interacting proteins has shed light on the important role of intracellular membrane trafficking in disease etiology. In this review, we cover recent advances in the field with an emphasis on cellular mechanisms.
Collapse
Affiliation(s)
- Marcellus J Banworth
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Guangpu Li
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| |
Collapse
|
62
|
Wang N, Chen S, Zhang B, Li S, Jin F, Gao D, Liu H, Jiang Y. 8u, a pro-apoptosis/cell cycle arrest compound, suppresses invasion and metastasis through HSP90α downregulating and PI3K/Akt inactivation in hepatocellular carcinoma cells. Sci Rep 2018; 8:309. [PMID: 29321577 PMCID: PMC5762664 DOI: 10.1038/s41598-017-18701-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/15/2017] [Indexed: 12/15/2022] Open
Abstract
8u, an acridine derivative, has been proved effective anti-hepatocarcinoma effect, while the underlying mechanism remains unclear. Here, metabolomics and proteomics approaches were applied to study its anti-cancer mechanism and explore its effect on HepG2 cells' invasion and metastasis abilities. The results showed that 8u significantly suppressed HepG2 cells migration and enhanced cell-to-cell junctions. The inhibition effect of 8u on invasion and metastasis disappeared after HSP90α gene silencing, and was reversed after HSP90α overexpression. The biological experimental results indicated that 8u also blocked PI3K/Akt pathway, thereby reducing fatty acid synthase (FASN) protein expression and disordering intracellular lipid metabolism to inhibit cell invasion and metastasis. In addition, HSP90α protein and PI3K/Akt pathway could co-adjust to each other. These findings demonstrated that 8u could efficiently suppress the invasion and metastasis of HepG2 cells by decreasing the expression of HSP90α protein and inhibiting the PI3K/Akt signaling pathway, which could be used as a potential candidate for the treatment of HCC.
Collapse
Affiliation(s)
- Ning Wang
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Shaopeng Chen
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Bin Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, 315211, China
| | - Shangfu Li
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Feng Jin
- Neptunus Pharmaceutical Technology Center, Shenzhen, 518057, China
| | - Dan Gao
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China.
| | - Hongxia Liu
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Yuyang Jiang
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- School of Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
63
|
Banworth MJ, Li G. Consequences of Rab GTPase dysfunction in genetic or acquired human diseases. Small GTPases 2017; 9:158-181. [PMID: 29239692 DOI: 10.1080/21541248.2017.1397833] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rab GTPases are important regulators of intracellular membrane trafficking in eukaryotes. Both activating and inactivating mutations in Rab genes have been identified and implicated in human diseases ranging from neurological disorders to cancer. In addition, altered Rab expression is often associated with disease prognosis. As such, the study of diseases associated with Rabs or Rab-interacting proteins has shed light on the important role of intracellular membrane trafficking in disease etiology. In this review, we cover recent advances in the field with an emphasis on cellular mechanisms.
Collapse
Affiliation(s)
- Marcellus J Banworth
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Guangpu Li
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| |
Collapse
|
64
|
Luo Y, Yu SY, Chen JJ, Qin J, Qiu YE, Zhong M, Chen M. MiR-27b directly targets Rab3D to inhibit the malignant phenotype in colorectal cancer. Oncotarget 2017; 9:3830-3841. [PMID: 29423086 PMCID: PMC5790503 DOI: 10.18632/oncotarget.23237] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/01/2017] [Indexed: 02/05/2023] Open
Abstract
MiRNAs, as oncogenes or as anti-oncogenes, play critically regulated roles in the initiation and progression of colorectal cancer at posttranscriptional level. However, the underlying functions of miR-27b in colorectal cancer remain largely unexplored. Here, we demonstrated miR-27b is significantly down-regulated in colorectal cancer tissues, and decreased miR-27b expression was closely associated with shorter overall survival of patients with colorectal cancer. By gain- and loss-of-function studies, we showed miR-27b remarkably suppressed cell proliferation and invasion of colorectal cancer. Furthermore, luciferase reporter assay identified Rab3D was the direct functional target of miR-27b. And Rab3D partly reversed the suppression of cell proliferation and invasion caused by miR-27b mimics. Finally, the animal experiment showed miR-27b plays a crucial role on colorectal cancer progression by targeting Rab3D. Taken together, our study implied miR-27b inhibits cell growth and invasion by targeting Rab3D, and miR-27b is a potential biomarker for prognosis and therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Yang Luo
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Shi-Yong Yu
- Department of General Surgery, Shanghai Pudong New Area People's Hospital, Shanghai 201200, P.R. China
| | - Jian-Jun Chen
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jun Qin
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yi-Er Qiu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Min Chen
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
65
|
Shah F, Goossens E, Atallah NM, Grimard M, Kelley MR, Fishel ML. APE1/Ref-1 knockdown in pancreatic ductal adenocarcinoma - characterizing gene expression changes and identifying novel pathways using single-cell RNA sequencing. Mol Oncol 2017; 11:1711-1732. [PMID: 28922540 PMCID: PMC5709621 DOI: 10.1002/1878-0261.12138] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/24/2017] [Accepted: 09/02/2017] [Indexed: 12/18/2022] Open
Abstract
Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1 or APE1) is a multifunctional protein that regulates numerous transcription factors associated with cancer-related pathways. Because APE1 is essential for cell viability, generation of APE1-knockout cell lines and determining a comprehensive list of genes regulated by APE1 has not been possible. To circumvent this challenge, we utilized single-cell RNA sequencing to identify differentially expressed genes (DEGs) in relation to APE1 protein levels within the cell. Using a straightforward yet novel statistical design, we identified 2837 genes whose expression is significantly changed following APE1 knockdown. Using this gene expression profile, we identified multiple new pathways not previously linked to APE1, including the EIF2 signaling and mechanistic target of Rapamycin pathways and a number of mitochondrial-related pathways. We demonstrate that APE1 has an effect on modifying gene expression up to a threshold of APE1 expression, demonstrating that it is not necessary to completely knockout APE1 in cells to accurately study APE1 function. We validated the findings using a selection of the DEGs along with siRNA knockdown and qRT-PCR. Testing additional patient-derived pancreatic cancer cells reveals particular genes (ITGA1, TNFAIP2, COMMD7, RAB3D) that respond to APE1 knockdown similarly across all the cell lines. Furthermore, we verified that the redox function of APE1 was responsible for driving gene expression of mitochondrial genes such as PRDX5 and genes that are important for proliferation such as SIPA1 and RAB3D by treating with APE1 redox-specific inhibitor, APX3330. Our study identifies several novel genes and pathways affected by APE1, as well as tumor subtype specificity. These findings will allow for hypothesis-driven approaches to generate combination therapies using, for example, APE1 inhibitor APX3330 with other approved FDA drugs in an innovative manner for pancreatic and other cancer treatments.
Collapse
Affiliation(s)
- Fenil Shah
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Emery Goossens
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - Nadia M Atallah
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Michelle Grimard
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark R Kelley
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa L Fishel
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
66
|
Tzeng HT, Li TH, Tang YA, Tsai CH, Frank Lu PJ, Lai WW, Chiang CW, Wang YC. Phosphorylation of Rab37 by protein kinase C alpha inhibits the exocytosis function and metastasis suppression activity of Rab37. Oncotarget 2017; 8:108556-108570. [PMID: 29312551 PMCID: PMC5752464 DOI: 10.18632/oncotarget.20998] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/05/2017] [Indexed: 01/31/2023] Open
Abstract
We previously identified a novel Rab small GTPase protein, Rab37, which plays a critical role in regulating exocytosis of secreted glycoproteins, tissue inhibitor of metalloproteinases 1 (TIMP1) to suppress lung cancer metastasis. Patients with preserved Rab37 protein expression were associated with better prognosis. However, a significant number of the patients with preserved Rab37 expression showed poor survival. In addition, the molecular mechanism for the regulation of Rab37-mediated exocytosis remained to be further identified. Therefore, we investigated the molecular mechanism underlying the dysregulation of Rab37-mediated exocytosis and metastasis suppression. Here, we report a novel mechanism for Rab37 inactivation by phosphorylation. Lung cancer patients with preserved Rab37, low TIMP1, and high PKCα expression profile correlate with worse progression-free survival examined by Kaplan-Meier survival, suggesting that PKCα overexpression leads to dysfunction of Rab37. This PKCα-Rab37-TIMP1 expression profile predicts the poor outcome by multivariate Cox regression analysis. We also show that Rab37 is phosphorylated by protein kinase Cα (PKCα) at threonine 172 (T172), leading to attenuation of its GTP-bound state, and impairment of the Rab37-mediated exocytosis of TIMP1, and thus reduces its suppression activity on lung cancer cell motility. We further demonstrate that PKCα reduces vesicle colocalization of Rab37 and TIMP1, and therefore inhibits Rab37-mediated TIMP1 trafficking. Moreover, Phospho-mimetic aspartate substitution mutant T172D of Rab37 significantly promotes tumor metastasis in vivo. Our findings reveal a novel regulation of Rab37 activity by PKCα-mediated phosphorylation which inhibits exocytic transport of TIMP1 and thereby enhances lung tumor metastasis.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Hsin Li
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-An Tang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology and Research (ASTAR), Singapore
| | - Chung-Han Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jung Frank Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Wei Lai
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Wu Chiang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
67
|
Lian Q, Ma DM, Chen MG, Chen K, Li XJ. Silencing Rab14 represses the proliferation and migration of oral squamous cell carcinoma, and enhances cisplatin sensitivity. Am J Transl Res 2017; 9:4195-4205. [PMID: 28979693 PMCID: PMC5622262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/17/2017] [Indexed: 06/07/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is a subtype of head and neck cancer with a relatively poor prognosis. The mechanisms underlying the initiation and progression of OSCC are complex and not yet fully understood; however, this information is critical for developing novel therapeutic targets and improving patient outcome. Rab14, a Ras related protein, has been implicated in multiple forms of cancer. In the present study, we confirmed that Rab14 is overexpression in human OSCC tissue, compared with normal oral mucosa samples. In addition, knockdown of Rab14 exerted potent anti-tumor effects by repressing the proliferation and migration of OSCC cell lines. Moreover, knockdown of Rab14 reduced the expression of Cyclin D1 and CXCR4, at the level of protein and mRNA, both in vitro and in vivo. Additionally, abrogation of Rab14 enhanced cisplatin sensitivity in OSCC cells in vitro and in vivo. Taken together, our data provides evidence for Rab14 as a potential therapeutic target in OSCC treatment.
Collapse
Affiliation(s)
- Qing Lian
- Department of Stomatology, The People’s Hospital of ZhenhaiNingbo 315202, Zhejiang Province, PR China
| | - Dong-Mei Ma
- Department of Stomatology, Zhejiang Xiaoshan HospitalHangzhou 311200, Zhejiang Province, PR China
| | - Ming-Gang Chen
- Department of Stomatology, The People’s Hospital of ZhenhaiNingbo 315202, Zhejiang Province, PR China
| | - Ke Chen
- Department of Stomatology, The People’s Hospital of ZhenhaiNingbo 315202, Zhejiang Province, PR China
| | - Xiang-Jun Li
- Department of Stomatology, Zhejiang Xiaoshan HospitalHangzhou 311200, Zhejiang Province, PR China
| |
Collapse
|
68
|
Silencing of Rab3D suppresses the proliferation and invasion of esophageal squamous cell carcinoma cells. Biomed Pharmacother 2017; 91:402-407. [DOI: 10.1016/j.biopha.2017.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 12/27/2022] Open
|
69
|
Xie K, Chen M, Zhu M, Wang C, Qin N, Liang C, Song C, Dai J, Jin G, Shen H, Lin D, Ma H, Hu Z. A polymorphism in miR-1262 regulatory region confers the risk of lung cancer in Chinese population. Int J Cancer 2017; 141:958-966. [PMID: 28510306 DOI: 10.1002/ijc.30788] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 04/09/2017] [Accepted: 05/05/2017] [Indexed: 12/27/2022]
Abstract
It has been proposed that the majority of disease-associated loci identified by genome-wide association studies (GWAS) are enriched in non-coding regions, such as the promoter, enhancer or non-coding RNA genes. Thus, we performed a two-stage case-control study to systematically evaluate the association of genetic variants in miRNA regulatory regions (promoter and enhancer) with lung cancer risk in 7,763 subjects (discovery stage: 2,331 cases and 3,077 controls; validation stage: 1,065 cases and 1,290 controls). As a result, we identified that rs12740674 (C > T) in miR-1262 enhancer was significantly associated with the increased risk of lung cancer (additive model in discovery stage: adjusted OR = 1.31, 95%CI = 1.13-1.53, p = 3.846 × 10-4 in Nanjing GWAS; adjusted OR = 1.20, 95%CI = 1.00-1.44, p = 0.041 in Beijing GWAS; validation stage: adjusted OR = 1.20, 95%CI = 1.03-1.41, p = 0.024). In meta-analysis, the p value for the association between rs12740674 and lung cancer risk reached 6.204 × 10-6 (adjusted OR = 1.24, 95%CI = 1.13-1.36). Using 3DSNP database, The Cancer Genome Atlas (TCGA) data and functional assays, we observed that the risk T allele of rs12740674 reduced the expression level of miR-1262 in lung tissue through chromosomal looping, and overexpression of miR-1262 inhibited lung cancer cell proliferation probably through targeting the expression levels of ULK1 and RAB3D. Our findings confirmed the important role that genetic variants of noncoding sequence play in lung cancer susceptibility and indicated that rs12740674 in miR-1262 may be biologically relevant to lung carcinogenesis.
Collapse
Affiliation(s)
- Kaipeng Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.,Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Mengxi Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Meng Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Na Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Ci Song
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology and Department of Etiology and Carcinogenesis, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongxia Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
70
|
Qin X, Wang J, Wang X, Liu F, Jiang B, Zhang Y. Targeting Rabs as a novel therapeutic strategy for cancer therapy. Drug Discov Today 2017; 22:1139-1147. [PMID: 28390930 DOI: 10.1016/j.drudis.2017.03.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 02/18/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
Rab GTPases constitute the largest family of small GTPases. Rabs regulate not only membrane trafficking but also cell signaling, growth and survival, and development. Increasingly, Rabs and their effectors are shown to be overexpressed or subject to loss-of-function mutations in a variety of disease settings, including cancer progression. This review provides an overview of dysregulated Rab proteins in cancer, and highlights the signaling and secretory pathways in which they operate, with the aim of identifying potential avenues for therapeutic intervention. Recent progress and perspectives for direct and/or indirect targeting of Rabs are also summarized.
Collapse
Affiliation(s)
- Xiaoyu Qin
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China
| | - Jiongyi Wang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China
| | - Xinxin Wang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China
| | - Feng Liu
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China
| | - Bin Jiang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China.
| | - Yanjie Zhang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201900, China.
| |
Collapse
|
71
|
Simon M, Mesmar F, Helguero L, Williams C. Genome-wide effects of MELK-inhibitor in triple-negative breast cancer cells indicate context-dependent response with p53 as a key determinant. PLoS One 2017; 12:e0172832. [PMID: 28235006 PMCID: PMC5325553 DOI: 10.1371/journal.pone.0172832] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 02/10/2017] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive, highly recurrent breast cancer subtype, affecting approximately one-fifth of all breast cancer patients. Subpopulations of treatment-resistant cancer stem cells within the tumors are considered to contribute to disease recurrence. A potential druggable target for such cells is the maternal embryonic leucine-zipper kinase (MELK). MELK expression is upregulated in mammary stem cells and in undifferentiated cancers, where it correlates with poor prognosis and potentially mediates treatment resistance. Several MELK inhibitors have been developed, of which one, OTSSP167, is currently in clinical trials. In order to better understand how MELK and its inhibition influence TNBC, we verified its anti-proliferative and apoptotic effects in claudin-low TNBC cell lines MDA-MB-231 and SUM-159 using MTS assays and/or trypan blue viability assays together with analysis of PARP cleavage. Then, using microarrays, we explored which genes were affected by OTSSP167. We demonstrate that different sets of genes are regulated in MDA-MB-231 and SUM-159, but in both cell lines genes involved in cell cycle, mitosis and protein metabolism and folding were regulated. We identified p53 (TP53) as a potential upstream regulator of the regulated genes. Using western blot we found that OTSSP167 downregulates mutant p53 in all tested TNBC cell lines (MDA-MB-231, SUM-159, and BT-549), but upregulates wild-type p53 in the luminal A subtype MCF-7 cell line. We propose that OTSSP167 might have context-dependent or off-target effects, but that one consistent mechanism of action could involve the destabilization of mutant p53.
Collapse
Affiliation(s)
- Marisa Simon
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Texas, United States of America
| | - Fahmi Mesmar
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Texas, United States of America
| | - Luisa Helguero
- Institute for Research in Biomedicine, Department of Biosciences, University of Aveiro, Aveiro, Portugal
| | - Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Texas, United States of America
- Division of Proteomics, SciLifeLab, School of Biotechnology, KTH – Royal Institute of Technology, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
72
|
Steinbichler TB, Dudás J, Riechelmann H, Skvortsova II. The role of exosomes in cancer metastasis. Semin Cancer Biol 2017; 44:170-181. [PMID: 28215970 DOI: 10.1016/j.semcancer.2017.02.006] [Citation(s) in RCA: 304] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/09/2017] [Indexed: 12/21/2022]
Abstract
Exosomes are small membrane vesicles with a size ranging from 40 to 100nm. They can serve as functional mediators in cell interaction leading to cancer metastasis. Metastasis is a complex multistep process of cancer cell invasion, survival in blood vessels, attachment to and colonization of the host organ. Exosomes influence every step of this cascade and can be targeted by oncological treatment. This review highlights the role of exosomes in the various steps of the metastatic cascade and how exosome dependent pathways can be targeted as therapeutic approach or used for liquid biopsies.
Collapse
Affiliation(s)
| | - József Dudás
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Riechelmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Ira-Ida Skvortsova
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
73
|
Abstract
Tumor extracellular vesicles (EVs), including exosomes, emerged as key drivers of the pro-tumorigenic dialog between the tumor mass and its microenvironment by mediating long and short distance communication. In vitro studies defined the capacity of tumor EVs to modify the phenotypes of stromal and tumor cells. These studies are now supported by a growing number of functional in vivo experiments. Remarkably, they allowed the identification of a new role for tumor EVs in priming the pre-metastatic niches (PMN). Several molecules transported in tumor EVs (RNAs and proteins) have recently been found to be essential for tumor progression and metastasis in vivo. In parallel, novel EV labeling and tracking strategies have very recently allowed the first descriptions of tumor EVs in vivo and pave the way for a better understanding of their function in realistic pathophysiological contexts. Here, we review the functional approaches and the recent progress in in vivo imaging of EVs, which have refined our understanding of the role played by tumor EVs. Finally, we emphasize the remaining challenges and open questions related to the biology of tumor EVs.
Collapse
Affiliation(s)
- Vincent Hyenne
- a Inserm U1109, MN3T , Strasbourg , France.,b Université de Strasbourg , Strasbourg , France.,c LabEx Medalis, Université de Strasbourg , Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) , Strasbourg , France.,e CNRS SNC5055 , Strasbourg , France
| | - Olivier Lefebvre
- a Inserm U1109, MN3T , Strasbourg , France.,b Université de Strasbourg , Strasbourg , France.,c LabEx Medalis, Université de Strasbourg , Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) , Strasbourg , France
| | - Jacky G Goetz
- a Inserm U1109, MN3T , Strasbourg , France.,b Université de Strasbourg , Strasbourg , France.,c LabEx Medalis, Université de Strasbourg , Strasbourg , France.,d Fédération de Médecine Translationnelle de Strasbourg (FMTS) , Strasbourg , France
| |
Collapse
|
74
|
邹 洪, 邬 红, 许 川. [Research Progress of Exosomes in Lung Cancer Diagnosis and Treatment]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:778-783. [PMID: 27866522 PMCID: PMC5999640 DOI: 10.3779/j.issn.1009-3419.2016.11.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/20/2016] [Accepted: 08/20/2016] [Indexed: 12/15/2022]
Abstract
As the leading cause of morbidity and cancer related-death worldwide, lung cancer has a serious threat to human health. Exosomes are nanoscale lipid membrane vesicles derived from multivesicles, which containing active biomolecules including proteins, lipids, nucleic acids and etc. Exosomes play important roles in lung cancer initiation and progression by promoting the formation of tumor microenvironment, enhancing tumor invasive and metastasis capability, leading to immunosuppression and resistance to chemoradiotherapy, and also have the application value in early diagnosis and treatment. This review summarizes the research progress of exosomes in tumor initiation and progression, and its roles in diagnosis and treatment of lung cancer.
Collapse
Affiliation(s)
- 洪波 邹
- 646000 泸州,西南医科大学附属医院The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- 610083 成都,成都军区总医院肿瘤诊治中心Department of Oncology, Chengdu Military General Hospital, Chengdu 610083, China
| | - 红 邬
- 646000 泸州,西南医科大学附属医院The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- 530021 南宁,广西医科大学Guangxi Medical University, Nanning 530021, China
| | - 川 许
- 646000 泸州,西南医科大学附属医院The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- 610083 成都,成都军区总医院肿瘤诊治中心Department of Oncology, Chengdu Military General Hospital, Chengdu 610083, China
| |
Collapse
|
75
|
Abstract
A large group of small Rab GTPases which mediate secretory and endosomal membrane transport, as well as autophagosome biogenesis, are essential components of vesicle trafficking machinery. Specific Rab protein together with the cognate effectors coordinates the dynamics of trafficking pathway and determines the cargo proteins destination. Functional impairments of Rab proteins by mutations or post-translational modifications disrupting the regulatory network of vesicle trafficking have been implicated in tumorigenesis. Therefore, the vesicle transport regulators play essential roles in the mediation of cancer cell biology, including uncontrolled cell growth, invasion and metastasis. The context-dependent role of the same Rab to act as either an oncoprotein or tumor suppressor in different cancers is found. Such discrepancies may be due in part to the interaction of specific Rab protein with different effectors or cargos in various tumors. Here, we review recent advances in the roles of Rab GTPases in communicating with other effectors in tumor progression. In this review, we also emphasize dysregulation of Rab-mediated membrane delivery shifting normal cell behaviors toward malignancy. Thus, recovery of the dysregulated vesicle trafficking systems in cancer cells may provide future directions for potential strategy to restrain tumor progression.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Department of Pharmacology, National Cheng Kung University, College of Medicine, No.1, University Road, Tainan, 70101, Taiwan, People's Republic of China
| | - Yi-Ching Wang
- Department of Pharmacology, National Cheng Kung University, College of Medicine, No.1, University Road, Tainan, 70101, Taiwan, People's Republic of China. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan, 70101, Taiwan, People's Republic of China.
| |
Collapse
|
76
|
Luo Y, Ye GY, Qin SL, Mu YF, Zhang L, Qi Y, Qiu YE, Yu MH, Zhong M. High expression of Rab3D predicts poor prognosis and associates with tumor progression in colorectal cancer. Int J Biochem Cell Biol 2016; 75:53-62. [DOI: 10.1016/j.biocel.2016.03.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/04/2016] [Accepted: 03/28/2016] [Indexed: 12/18/2022]
|
77
|
Schulten HJ, Hussein D, Al-Adwani F, Karim S, Al-Maghrabi J, Al-Sharif M, Jamal A, Al-Ghamdi F, Baeesa SS, Bangash M, Chaudhary A, Al-Qahtani M. Microarray Expression Data Identify DCC as a Candidate Gene for Early Meningioma Progression. PLoS One 2016; 11:e0153681. [PMID: 27096627 PMCID: PMC4838307 DOI: 10.1371/journal.pone.0153681] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/01/2016] [Indexed: 12/23/2022] Open
Abstract
Meningiomas are the most common primary brain tumors bearing in a minority of cases an aggressive phenotype. Although meningiomas are stratified according to their histology and clinical behavior, the underlying molecular genetics predicting aggressiveness are not thoroughly understood. We performed whole transcript expression profiling in 10 grade I and four grade II meningiomas, three of which invaded the brain. Microarray expression analysis identified deleted in colorectal cancer (DCC) as a differentially expressed gene (DEG) enabling us to cluster meningiomas into DCC low expression (3 grade I and 3 grade II tumors), DCC medium expression (2 grade I and 1 grade II tumors), and DCC high expression (5 grade I tumors) groups. Comparison between the DCC low expression and DCC high expression groups resulted in 416 DEGs (p-value<0.05; fold change>2). The most significantly downregulated genes in the DCC low expression group comprised DCC, phosphodiesterase 1C (PDE1C), calmodulin-dependent 70kDa olfactomedin 2 (OLFM2), glutathione S-transferase mu 5 (GSTM5), phosphotyrosine interaction domain containing 1 (PID1), sema domain, transmembrane domain (TM) and cytoplasmic domain, (semaphorin) 6D (SEMA6D), and indolethylamine N-methyltransferase (INMT). The most significantly upregulated genes comprised chromosome 5 open reading frame 63 (C5orf63), homeodomain interacting protein kinase 2 (HIPK2), and basic helix-loop-helix family, member e40 (BHLHE40). Biofunctional analysis identified as predicted top upstream regulators beta-estradiol, TGFB1, Tgf beta complex, LY294002, and dexamethasone and as predicted top regulator effectors NFkB, PIK3R1, and CREBBP. The microarray expression data served also for a comparison between meningiomas from female and male patients and for a comparison between brain invasive and non-invasive meningiomas resulting in a number of significant DEGs and related biofunctions. In conclusion, based on its expression levels, DCC may constitute a valid biomarker to identify those benign meningiomas at risk for progression.
Collapse
Affiliation(s)
- Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Deema Hussein
- King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatima Al-Adwani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jaudah Al-Maghrabi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Mona Al-Sharif
- Department of Biology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awatif Jamal
- Department of Pathology, Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Fahad Al-Ghamdi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Saleh S. Baeesa
- Division of Neurosurgery, Department of Surgery, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Mohammed Bangash
- Division of Neurosurgery, Department of Surgery, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Adeel Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
78
|
Abstract
The Golgi apparatus-complex is a highly dynamic organelle which is considered the "heart" of intracellular transportation. Since its discovery by Camillo Golgi in 1873, who described it as the "black reaction," and despite the enormous volume of publications about Golgi, this apparatus remains one of the most enigmatic of the cytoplasmic organelles. A typical mammalian Golgi consists of a parallel series of flattened, disk-shaped cisternae which align into stacks. The tremendous volume of Golgi-related incoming and outgoing traffic is mediated by different motor proteins, including members of the dynein, kinesin, and myosin families. Yet in spite of the strenuous work it performs, Golgi contrives to maintain its monolithic morphology and orchestration of matrix and residential proteins. However, in response to stress, alcohol, and treatment with many pharmacological drugs over time, Golgi undergoes a kind of disorganization which ranges from mild enlargement to critical scattering. While fragmentation of the Golgi was confirmed in cancer by electron microscopy almost fifty years ago, it is only in recent years that we have begun to understand the significance of Golgi fragmentation in the biology of tumors. Below author would like to focus on how Golgi fragmentation opens the doors for cascades of fatal pathways which may facilitate cancer progression and metastasis. Among the issues addressed will be the most important cancer-specific hallmarks of Golgi fragmentation, including aberrant glycosylation, abnormal expression of the Ras GTPases, dysregulation of kinases, and hyperactivity of myosin motor proteins.
Collapse
Affiliation(s)
- Armen Petrosyan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|