201
|
Aggarwal R, Jha M, Shrivastava A, Jha AK. Natural Compounds: Role in Reversal of Epigenetic Changes. BIOCHEMISTRY (MOSCOW) 2016; 80:972-89. [PMID: 26547065 DOI: 10.1134/s0006297915080027] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The hallmarks of carcinogenesis are characterized by alterations in the expression of multiple genes that occur via genetic and epigenetic alterations, leading to genome rearrangements and instability. The reversible process of epigenetic regulation, which includes changes in DNA methylation, histone modifications, and alteration in microRNA (miRNA) expression that alter phenotype without any change in the DNA sequence, is recognized as a key mechanism in cancer cell metabolism. Recent advancements in the rapidly evolving field of cancer epigenetics have shown the anticarcinogenic potential of natural compounds targeting epigenetic mechanism as a common molecular approach for cancer treatment. This review summarizes the potential of natural chemopreventive agents to reverse cancer-related epigenetic aberrations by regulating the activity of histone deacetylases, histone acetyltransferases, DNA methyltransferase I, and miRNAs. Furthermore, there is impetus for determining novel and effective chemopreventive strategies, either alone or in combination with other anticancer agents that exhibit similar properties, for improving the therapeutic aspects of cancer.
Collapse
Affiliation(s)
- Ruchi Aggarwal
- Department of Biotechnology, IMS Engineering College, U. P. 201009, India.
| | | | | | | |
Collapse
|
202
|
Zhu H, Qin H, Li DM, Liu J, Zhao Q. Effect of PPM1H on malignant phenotype of human pancreatic cancer cells. Oncol Rep 2016; 36:2926-2934. [DOI: 10.3892/or.2016.5065] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/17/2016] [Indexed: 11/05/2022] Open
|
203
|
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1:12-34. [PMID: 30159408 PMCID: PMC6096431 DOI: 10.1016/j.ncrna.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The epigenetic regulation of cancer cells by small non-coding RNA molecules, the microRNAs (miRNAs), has raised particular interest in the field of oncology. These miRNAs play crucial roles concerning pathogenic properties of cancer cells and the sensitivity of cancer cells towards anticancer drugs. Certain miRNAs are responsible for an enhanced activity of drugs, while others lead to the formation of tumor resistance. In addition, miRNAs regulate survival and proliferation of cancer cells, in particular of cancer stem-like cells (CSCs), that are especially drug-resistant and, thus, cause tumor relapse in many cases. Various small molecule compounds were discovered that target miRNAs that are known to modulate tumor aggressiveness and drug resistance. This review comprises the effects of naturally occurring small molecules (phenolic compounds and terpenoids) on miRNAs involved in cancer diseases.
Collapse
Key Words
- 1,25-D, 1,25-dihydroxyvitamin D3
- 18-AGA, 18α-glycyrrhetinic acid
- 3,6-DHF, 3,6-dihydroxyflavone
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- CAPE, caffeic acid phenethyl ester
- CDODA-Me, methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate
- Dox, doxorubicin
- EGCG, (−)-epigallocatechin-3-O-gallate
- MicroRNA
- PEG, polyethylene glycol
- PPAP, polycyclic polyprenylated acylphloroglucinol
- Polyphenols
- RA, retinoic acid
- ROS, reactive oxygen species
- TQ, thymoquinone
- Terpenes
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|
204
|
Suliman MA, Zhang Z, Na H, Ribeiro ALL, Zhang Y, Niang B, Hamid AS, Zhang H, Xu L, Zuo Y. Niclosamide inhibits colon cancer progression through downregulation of the Notch pathway and upregulation of the tumor suppressor miR-200 family. Int J Mol Med 2016; 38:776-84. [PMID: 27460529 PMCID: PMC4990307 DOI: 10.3892/ijmm.2016.2689] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 06/23/2016] [Indexed: 01/16/2023] Open
Abstract
Colorectal cancer (CRC) is among the most frequent causes of cancer-related deaths worldwide. Thus, there is a need for the development of new therapeutic approaches for the treatment of CRC. Accumulating evidence has revealed that niclosamide, an anthelminthic drug, exerts antitumor activity in several types of cancer, including colon cancer. However, the underlying molecular mechanisms responsible for the effects of this drug remain elusive. Previous studies have shown that the aberrant Notch signaling pathway contributes to the carcinogenesis of colon cancer. Herein, we examined the effects of niclosamide on the growth, migration and apoptosis of colon cancer cells, and the role of the Notch signaling pathway. By performing MTT, wound-healing and Transwell migration assays, we observed that niclosamide suppressed the growth and migration of colon cancer cells, and flow cytometry demonstrated that cell apoptosis was induced. This was associated with the decreased protein expression of Notch1, Notch2, Notch3 and Hey1, and the increased expression of the tumor suppressor microRNA (miR or miRNA)-200 family members (miR-200a, miR-200b, miR-200c, miR-141 and miR-429) that are typically downregulated in colon cancer. Collectively, these findings demonstrate that niclosamide potentially inhibits the progression of colon cancer by downregulating Notch signaling and by upregulating the miR-200 family members.
Collapse
Affiliation(s)
- Mohammed A Suliman
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Zhenxing Zhang
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Heya Na
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ailton L L Ribeiro
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yu Zhang
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Bachir Niang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Abdu Salim Hamid
- Department of Clinical Laboratory Science, Asmara College of Health Sciences, Asmara, Eritrea
| | - Hua Zhang
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lijie Xu
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yunfei Zuo
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
205
|
Kim SM. Cellular and Molecular Mechanisms of 3,3'-Diindolylmethane in Gastrointestinal Cancer. Int J Mol Sci 2016; 17:ijms17071155. [PMID: 27447608 PMCID: PMC4964527 DOI: 10.3390/ijms17071155] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 12/18/2022] Open
Abstract
Studies in humans have shown that 3,3′-diindolylmethane (DIM), which is found in cruciferous vegetables, such as cabbage and broccoli, is effective in the attenuation of gastrointestinal cancers. This review presents the latest findings on the use, targets, and modes of action of DIM for the treatment of human gastrointestinal cancers. DIM acts upon several cellular and molecular processes in gastrointestinal cancer cells, including apoptosis, autophagy, invasion, cell cycle regulation, metastasis, angiogenesis, and endoplasmic reticulum (ER) stress. In addition, DIM increases the efficacy of other drugs or therapeutic chemicals when used in combinatorial treatment for gastrointestinal cancer. The studies to date offer strong evidence to support the use of DIM as an anticancer and therapeutic agent for gastrointestinal cancer. Therefore, this review provides a comprehensive understanding of the preventive and therapeutic properties of DIM in addition to its different perspective on the safety of DIM in clinical applications for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Soo Mi Kim
- Department of Physiology, Chonbuk National University Medical School, Jeonju 561-180, Korea.
| |
Collapse
|
206
|
Vinall RL, Tepper CG, Ripoll AAZ, Gandour-Edwards RF, Durbin-Johnson BP, Yap SA, Ghosh PM, deVere White RW. Decreased expression of let-7c is associated with non-response of muscle-invasive bladder cancer patients to neoadjuvant chemotherapy. Genes Cancer 2016; 7:86-97. [PMID: 27382433 PMCID: PMC4918947 DOI: 10.18632/genesandcancer.103] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The identification and development of biomarkers which predict response of muscle invasive bladder cancer (MIBC) patients to neoadjuvant chemotherapy would likely increase usage of this treatment option and thereby improve patient survival rates. MiRNA array and qRT-PCR validation was used to identify miRNA which are associated with response to neoadjuvant chemotherapy. RNA was extracted from a total of 41 archival, fully annotated, MIBC patient diagnostic biopsies (20 chemo-responders and 21 non-responders (response is defined as > 5 year survival rate and being pT0 post-chemotherapy)). Microarray and qPCR identified let-7c as being differentially expressed in chemo-responder versus non-responder patients. Patients with higher let-7c expression levels had significantly higher odds of responding to chemotherapy (p = 0.023, OR 2.493, 95% CI 1.121, 5.546), and assessment of let-7c levels allowed for prediction of patient response (AUC 0.72, positive predictive value 59%). Decreased let-7c was associated with MIBC incidence (p < 0.001), and significantly correlated with other related miRNA including those that were not differentially expressed between responders and non-responders. The combined data indicate let-7c plays a role in mediating chemoresistance to neoadjuvant chemotherapy in MIBC patients, and is a modest, yet clinically meaningful, predictor of patient response.
Collapse
Affiliation(s)
- Ruth L Vinall
- Department of Urology, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA; Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA; California Northstate University College of Pharmacy, Elk Grove, CA, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA
| | - Alexandra A Z Ripoll
- Department of Urology, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA
| | - Regina F Gandour-Edwards
- Department of Pathology, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA
| | - Blythe P Durbin-Johnson
- Department of Public Health Sciences, University of California Davis, Davis, California, USA
| | - Stanley A Yap
- Department of Urology, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA
| | - Paramita M Ghosh
- Department of Urology, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA; Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA; VA Northern California Health Care System, Mather, CA, USA
| | - Ralph W deVere White
- Department of Urology, University of California, Davis, School of Medicine and Comprehensive Cancer Center, Sacramento, California, USA
| |
Collapse
|
207
|
Diab M, Muqbil I, Mohammad RM, Azmi AS, Philip PA. The Role of microRNAs in the Diagnosis and Treatment of Pancreatic Adenocarcinoma. J Clin Med 2016; 5:59. [PMID: 27322337 PMCID: PMC4929414 DOI: 10.3390/jcm5060059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a very challenging malignancy. Disease is diagnosed in an advanced stage in the vast majority of patients, and PDAC cells are often resistant to conventional cytotoxic drugs. Targeted therapies have made no progress in the management of this disease, unlike other cancers. microRNAs (miRs) are small non-coding RNAs that regulate the expression of multitude number of genes by targeting their 3'-UTR mRNA region. Aberrant expression of miRNAs has been linked to the development of various malignancies, including PDAC. In PDAC, a series of miRs have been defined as holding promise for early diagnostics, as indicators of therapy resistance, and even as markers for therapeutic response in patients. In this mini-review, we present an update on the various different miRs that have been defined in PDAC biology.
Collapse
Affiliation(s)
- Maria Diab
- Department of Internal Medicine, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer institute, Wayne State University, Detroit, MI 48201, USA.
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer institute, Wayne State University, Detroit, MI 48201, USA.
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer institute, Wayne State University, Detroit, MI 48201, USA.
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer institute, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
208
|
MicroRNA in pancreatic cancer. J Hum Genet 2016; 62:33-40. [DOI: 10.1038/jhg.2016.59] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 02/07/2023]
|
209
|
Mondal SK, Jinka S, Pal K, Nelli S, Dutta SK, Wang E, Ahmad A, AlKharfy KM, Mukhopadhyay D, Banerjee R. Glucocorticoid Receptor-Targeted Liposomal Codelivery of Lipophilic Drug and Anti-Hsp90 Gene: Strategy to Induce Drug-Sensitivity, EMT-Reversal, and Reduced Malignancy in Aggressive Tumors. Mol Pharm 2016; 13:2507-23. [PMID: 27184196 DOI: 10.1021/acs.molpharmaceut.6b00230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many cancers including the late stage ones become drug-resistant and undergo epithelial-to-mesenchymal transition (EMT). These lead to enhanced invasion, migration, and metastasis toward manifesting its aggressiveness and malignancy. One of the key hallmarks of cancer is its overdependence on glycolysis as its preferred energy metabolism pathway. The strict avoidance of alternate energy pathway gluconeogenesis by cancer cells points to a yet-to-be hoisted role of glucocorticoid receptor (GR) especially in tumor microenvironment, where cells are known to become drug-sensitive through induction of gluconeogenesis. However, since GR is involved in metabolism, anti-inflammatory reactions, immunity besides inducing gluconeogenesis, a greater role of GR in tumor microenvironment is envisaged. We have shown previously that GR, although ubiquitously expressed in all cells; afford to be an effective cytoplasmic target for killing cancer cells selectively. Herein, we report the therapeutic use of a newly developed GR-targeted liposomal concoction (DXE) coformulating a lipophilic drug (ESC8) and an anti-Hsp90 anticancer gene against aggressive tumor models. This induced drug-sensitivity and apoptosis while reversing EMT in tumor cells toward effective retardation of aggressive growth in pancreas and skin tumor models. Additionally, the ESC8-free lipid formulation upon cotreatment with hydrophilic drugs, gemcitabine and doxorubicin, could effectively sensitize and kill pancreatic cancer and melanoma cells, respectively. The formulation-triggered EMT-reversal was GR-dependent. Overall, we found a new strategy for drug sensitization that led to the advent of new GR-targeted anticancer therapeutics.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India.,Academy of Scientific & Innovative Research (AcSIR) , 2 Rafi Marg, New Delhi 110001, India
| | - Sudhakar Jinka
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India.,Academy of Scientific & Innovative Research (AcSIR) , 2 Rafi Marg, New Delhi 110001, India
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Swetha Nelli
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, King Saud University , Riyadh 11451, Saudi Arabia
| | - Khalid M AlKharfy
- Department of Clinical Pharmacy, King Saud University , Riyadh 11451, Saudi Arabia
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Jacksonville, Florida 32224, United States
| | - Rajkumar Banerjee
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology , Hyderabad 500 007, India.,Academy of Scientific & Innovative Research (AcSIR) , 2 Rafi Marg, New Delhi 110001, India
| |
Collapse
|
210
|
Ma J, Zeng F, Ma C, Pang H, Fang B, Lian C, Yin B, Zhang X, Wang Z, Xia J. Synergistic reversal effect of epithelial-to-mesenchymal transition by miR-223 inhibitor and genistein in gemcitabine-resistant pancreatic cancer cells. Am J Cancer Res 2016; 6:1384-95. [PMID: 27429851 PMCID: PMC4937740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/02/2016] [Indexed: 06/06/2023] Open
Abstract
Emerging studies have demonstrated that EMT phenotype is closely related with tumor progression and drug resistance in a variety of human cancers. Recently, it has been extensively demonstrated that microRNAs (miRNAs) play a pivotal role in regulating EMT. In our previously reports, we have reported that inhibition of miR-223 could reverse EMT phenotype and improve chemotherapeutic drug sensitivity. We also reported that genistein down-regulated miR-223 expression in gemcitabine-resistant (GR) pancreatic cancer cells. Here, we explored whether there was the synergistic effect between miR-223 inhibitor and genistein on cell growth, migration, invasion and reversal of EMT in GR pancreatic cancer. We found that the combination of miR-223 inhibitor and genistein synergistically reduced cell motility and invasion and enhanced gemcitabine sensitivity in GR cells. In addition, we further observed that miR-223 inhibitor and genistein reversed EMT features in GR cells. This study suggests that the combination of miR-223 inhibitor and genistein may be a potential therapeutic strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jia Ma
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow UniversitySuzhou 215123, China
- Department of Biochemistry and Molecular Biology, Bengbu Medical CollegeAnhui 233030, China
| | - Fanpeng Zeng
- Research Center of Clinical Laboratory Science, Bengbu Medical CollegeAnhui 233030, China
| | - Cong Ma
- Research Center of Clinical Laboratory Science, Bengbu Medical CollegeAnhui 233030, China
| | - Haijie Pang
- Research Center of Clinical Laboratory Science, Bengbu Medical CollegeAnhui 233030, China
| | - Binbin Fang
- Research Center of Clinical Laboratory Science, Bengbu Medical CollegeAnhui 233030, China
| | - Chaoqun Lian
- Department of Biochemistry and Molecular Biology, Bengbu Medical CollegeAnhui 233030, China
| | - Bin Yin
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow UniversitySuzhou 215123, China
| | - Xueping Zhang
- Department of Anesthesiology, Shenzhen People’s Hospital, 2nd Clinical Medical College of Jinan UniversityShenzhen 518020, China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow UniversitySuzhou 215123, China
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA 02215, USA
| | - Jun Xia
- Department of Biochemistry and Molecular Biology, Bengbu Medical CollegeAnhui 233030, China
| |
Collapse
|
211
|
Hawa Z, Haque I, Ghosh A, Banerjee S, Harris L, Banerjee SK. The miRacle in Pancreatic Cancer by miRNAs: Tiny Angels or Devils in Disease Progression. Int J Mol Sci 2016; 17:E809. [PMID: 27240340 PMCID: PMC4926343 DOI: 10.3390/ijms17060809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/04/2016] [Accepted: 05/19/2016] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with increasing incidence and high mortality. Surgical resection is the only potentially curative treatment of patients with PDAC. Because of the late presentation of the disease, about 20 percent of patients are candidates for this treatment. The average survival of resected patients is between 12 and 20 months, with a high probability of relapse. Standard chemo and radiation therapies do not offer significant improvement of the survival of these patients. Furthermore, novel treatment options aimed at targeting oncogenes or growth factors in pancreatic cancer have proved unsuccessful. Thereby, identifying new biomarkers that can detect early stages of this disease is of critical importance. Among these biomarkers, microRNAs (miRNAs) have supplied a profitable recourse and become an attractive focus of research in PDAC. MiRNAs regulate many genes involved in the development of PDAC through mRNA degradation or translation inhibition. The possibility of intervention in the molecular mechanisms of miRNAs regulation could begin a new generation of PDAC therapies. This review summarizes the reports describing miRNAs involvement in cellular processes involving pancreatic carcinogenesis and their utility in diagnosis, survival and therapeutic potential in pancreatic cancer.
Collapse
Affiliation(s)
- Zuhair Hawa
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
| | - Inamul Haque
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| | - LaCoiya Harris
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO 64128, USA.
- Division of Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66205, USA.
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66205, USA.
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS 66205, USA.
| |
Collapse
|
212
|
Roles of Dietary Phytoestrogens on the Regulation of Epithelial-Mesenchymal Transition in Diverse Cancer Metastasis. Toxins (Basel) 2016; 8:toxins8060162. [PMID: 27231938 PMCID: PMC4926129 DOI: 10.3390/toxins8060162] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 12/31/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays a key role in tumor progression. The cells undergoing EMT upregulate the expression of cell motility-related proteins and show enhanced migration and invasion. The hallmarks of EMT in cancer cells include changed cell morphology and increased metastatic capabilities in cell migration and invasion. Therefore, prevention of EMT is an important tool for the inhibition of tumor metastasis. A novel preventive therapy is needed, such as treatment of natural dietary substances that are nontoxic to normal human cells, but effective in inhibiting cancer cells. Phytoestrogens, such as genistein, resveratrol, kaempferol and 3,3′-diindolylmethane (DIM), can be raised as possible candidates. They are plant-derived dietary estrogens, which are found in tea, vegetables and fruits, and are known to have various biological efficacies, including chemopreventive activity against cancers. Specifically, these phytoestrogens may induce not only anti-proliferation, apoptosis and cell cycle arrest, but also anti-metastasis by inhibiting the EMT process in various cancer cells. There have been several signaling pathways found to be associated with the induction of the EMT process in cancer cells. Phytoestrogens were demonstrated to have chemopreventive effects on cancer metastasis by inhibiting EMT-associated pathways, such as Notch-1 and TGF-beta signaling. As a result, phytoestrogens can inhibit or reverse the EMT process by upregulating the expression of epithelial phenotypes, including E-cadherin, and downregulating the expression of mesenchymal phenotypes, including N-cadherin, Snail, Slug, and vimentin. In this review, we focused on the important roles of phytoestrogens in inhibiting EMT in many types of cancer and suggested phytoestrogens as prominent alternative compounds to chemotherapy.
Collapse
|
213
|
Fujioka N, Fritz V, Upadhyaya P, Kassie F, Hecht SS. Research on cruciferous vegetables, indole-3-carbinol, and cancer prevention: A tribute to Lee W. Wattenberg. Mol Nutr Food Res 2016; 60:1228-38. [DOI: 10.1002/mnfr.201500889] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Naomi Fujioka
- Masonic Cancer Center; University of Minnesota; Minneapolis MN USA
| | - Vincent Fritz
- Southern Research and Outreach Center; University of Minnesota; Minneapolis MN USA
| | - Pramod Upadhyaya
- Masonic Cancer Center; University of Minnesota; Minneapolis MN USA
| | - Fekadu Kassie
- Masonic Cancer Center; University of Minnesota; Minneapolis MN USA
| | - Stephen S. Hecht
- Masonic Cancer Center; University of Minnesota; Minneapolis MN USA
| |
Collapse
|
214
|
Khoogar R, Kim BC, Morris J, Wargovich MJ. Chemoprevention in gastrointestinal physiology and disease. Targeting the progression of cancer with natural products: a focus on gastrointestinal cancer. Am J Physiol Gastrointest Liver Physiol 2016; 310:G629-44. [PMID: 26893159 PMCID: PMC4867331 DOI: 10.1152/ajpgi.00201.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/11/2016] [Indexed: 01/31/2023]
Abstract
The last decade has witnessed remarkable progress in the utilization of natural products for the prevention and treatment of human cancer. Many agents now in the pipeline for clinical trial testing have evolved from our understanding of how human nutritional patterns account for widespread differences in cancer risk. In this review, we have focused on many of these promising agents arguing that they may provide a new strategy for cancer control: natural products once thought to be only preventive in their mode of action now are being explored for efficacy in tandem with cancer therapeutics. Natural products may reduce off-target toxicity of therapeutics while making cancers more amenable to therapy. On the horizon is the use of certain natural products, in their own right, as mitigants of late-stage cancer, a new frontier for small-molecule natural product drug discovery.
Collapse
Affiliation(s)
- Roxane Khoogar
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Byung-Chang Kim
- 2Center for Colorectal Center, Center for Cancer Prevention and Detection, Research Institute and Hospital, National Cancer Center, Ilsan-ro, Illsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea
| | - Jay Morris
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Michael J. Wargovich
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| |
Collapse
|
215
|
Shankar E, Kanwal R, Candamo M, Gupta S. Dietary phytochemicals as epigenetic modifiers in cancer: Promise and challenges. Semin Cancer Biol 2016; 40-41:82-99. [PMID: 27117759 DOI: 10.1016/j.semcancer.2016.04.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 04/08/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022]
Abstract
The influence of diet and environment on human health has been known since ages. Plant-derived natural bioactive compounds (phytochemicals) have acquired an important role in human diet as potent antioxidants and cancer chemopreventive agents. In past few decades, the role of epigenetic alterations such as DNA methylation, histone modifications and non-coding RNAs in the regulation of mammalian genome have been comprehensively addressed. Although the effects of dietary phytochemicals on gene expression and signaling pathways have been widely studied in cancer, the impact of these dietary compounds on mammalian epigenome is rapidly emerging. The present review outlines the role of different epigenetic mechanisms in the regulation and maintenance of mammalian genome and focuses on the role of dietary phytochemicals as epigenetic modifiers in cancer. Above all, the review focuses on summarizing the progress made thus far in cancer chemoprevention with dietary phytochemicals, the heightened interest and challenges in the future.
Collapse
Affiliation(s)
- Eswar Shankar
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA; Department of Urology, Case Western Reserve University, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Rajnee Kanwal
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA; Department of Urology, Case Western Reserve University, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Mario Candamo
- Department of Biology, School of Undergraduate Studies, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sanjay Gupta
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA; Department of Urology, Case Western Reserve University, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.
| |
Collapse
|
216
|
miR-200c: a versatile watchdog in cancer progression, EMT, and drug resistance. J Mol Med (Berl) 2016; 94:629-44. [PMID: 27094812 DOI: 10.1007/s00109-016-1420-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are 20-22-nucleotide small endogenous non-coding RNAs which regulate gene expression at post-transcriptional level. In the last two decades, identification of almost 2600 miRNAs in human and their potential to be modulated opened a new avenue to target almost all hallmarks of cancer. miRNAs have been classified as tumor suppressors or oncogenes depending on the phenotype they induce, the targets they modulate, and the tissue where they function. miR-200c, an illustrious tumor suppressor, is one of the highly studied miRNAs in terms of development, stemness, proliferation, epithelial-mesenchymal transition (EMT), therapy resistance, and metastasis. In this review, we first focus on the regulation of miR-200c expression and its role in regulating EMT in a ZEB1/E-cadherin axis-dependent and ZEB1/E-cadherin axis-independent manner. We then describe the role of miR-200c in therapy resistance in terms of multidrug resistance, chemoresistance, targeted therapy resistance, and radiotherapy resistance in various cancer types. We highlight the importance of miR-200c at the intersection of EMT and chemoresistance. Furthermore, we show how miR-200c coordinates several important signaling cascades such as TGF-β signaling, PI3K/Akt signaling, Notch signaling, VEGF signaling, and NF-κB signaling. Finally, we discuss miR-200c as a potential prognostic/diagnostic biomarker in several diseases, but mainly focusing on cancer and its potential application in future therapeutics.
Collapse
|
217
|
Taucher V, Mangge H, Haybaeck J. Non-coding RNAs in pancreatic cancer: challenges and opportunities for clinical application. Cell Oncol (Dordr) 2016; 39:295-318. [DOI: 10.1007/s13402-016-0275-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 01/17/2023] Open
|
218
|
Fan P, Liu L, Yin Y, Zhao Z, Zhang Y, Amponsah PS, Xiao X, Bauer N, Abukiwan A, Nwaeburu CC, Gladkich J, Gao C, Schemmer P, Gross W, Herr I. MicroRNA-101-3p reverses gemcitabine resistance by inhibition of ribonucleotide reductase M1 in pancreatic cancer. Cancer Lett 2016; 373:130-137. [PMID: 26828016 DOI: 10.1016/j.canlet.2016.01.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/20/2016] [Accepted: 01/22/2016] [Indexed: 01/25/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the most lethal malignancies and resistance to chemotherapy prevents the therapeutic outcome. MicroRNAs provide a novel therapeutic strategy. Here, the established and primary human PDA cell lines PANC-1, AsPC-1, MIA-PaCa2, AsanPaCa, BxPC-3 and three gemcitabine-resistant subclones were examined. A gene expression profiling revealed that the ribonucleotide reductase M1 (RRM1) was upregulated in gemcitabine-resistant cells, which was confirmed by qRT-PCR, Western blot analysis and immunostaining. Inhibition of RRM1 by lipotransfection of siRNA reduced its expression and reversed gemcitabine resistance. The expression of RRM1 correlated to gemcitabine resistance in vitro and was higher in malignant patient pancreas tissue compared to non-malignant pancreas tissue. By microRNA expression profiling, we identified microRNA-101-3p as top-downregulated candidate. Lipotransfection of microRNA-101-3p mimics inhibited the expression of RRM1, reduced the luciferase activity of its 3'UTR and sensitized for gemcitabine-induced cytotoxicity. These results underline the relevance of microRNA-101-3p-driven regulation of RRM1 in drug resistance and suggest the co-delivery of microRNA-101-3p and gemcitabine for more effective therapy outcome.
Collapse
MESH Headings
- 3' Untranslated Regions
- Antimetabolites, Antineoplastic/pharmacology
- Binding Sites
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/enzymology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/genetics
- Gene Expression Profiling/methods
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/enzymology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- RNA Interference
- Ribonucleoside Diphosphate Reductase
- Time Factors
- Transfection
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Up-Regulation
- Gemcitabine
Collapse
Affiliation(s)
- Pei Fan
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Li Liu
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Yefeng Yin
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Zhefu Zhao
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Yiyao Zhang
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Prince S Amponsah
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Xi Xiao
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Nathalie Bauer
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alia Abukiwan
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Clifford C Nwaeburu
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jury Gladkich
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Chao Gao
- Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Peter Schemmer
- Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Gross
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery, University of Heidelberg, Heidelberg, Germany; Section Surgical Research, University of Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
219
|
Vorvis C, Koutsioumpa M, Iliopoulos D. Developments in miRNA gene signaling pathways in pancreatic cancer. Future Oncol 2016; 12:1135-50. [PMID: 26984178 DOI: 10.2217/fon-2015-0050] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a devastating malignancy that ranks as the fourth leading cause of cancer-related deaths worldwide. Dismal prognosis is mainly attributable to limited knowledge of the molecular pathogenesis of the disease. miRNAs have been found to be deregulated in pancreatic cancer, affecting several steps of initiation and aggressiveness of the disease by regulating important signaling pathways, such as the KRAS and Notch pathways. Moreover, the effect of miRNAs on regulating cell cycle events and expression of transcription factors has gained a lot of attention. Recent studies have highlighted the application of miRNAs as biomarkers and therapeutic tools. The current review focuses on latest advances with respect to the roles of miRNAs in pancreatic ductal adenocarcinoma associated signaling pathways and miRNA-based therapeutics.
Collapse
Affiliation(s)
- Christina Vorvis
- Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Marina Koutsioumpa
- Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| |
Collapse
|
220
|
Zeng F, Xue M, Xiao T, Li Y, Xiao S, Jiang B, Ren C. MiR-200b promotes the cell proliferation and metastasis of cervical cancer by inhibiting FOXG1. Biomed Pharmacother 2016; 79:294-301. [PMID: 27044840 DOI: 10.1016/j.biopha.2016.02.033] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Previous studies have revealed the important role of miR-200b in cancer biology, including its upregulation in cervical cancer. However, miR-200b function in cervical cancer progression remains unclear. Thus, this study aims to explore the functional role of miR-200b in cervical cancer development, involving its potential regulation on FoxG1, one transcriptional repressor. METHODS Thirty paired cervical cancer samples were used to analyze the expression of miR-200b and FoxG1 by real time PCR and western blot analysis. Further gain- and loss-of-function studies were performed to validate FoxG1 as one miR-200b target, in line with luciferase report assays. MiR-200b silence was also conducted to observe its regulation on cell viability, migration and invasion in vitro, while tumor growth in vivo was tracked through the delivery of miR-200b inhibitor. RESULTS MiR-200b upregulation was confirmed in cancer tissues or cells as compared to normal controls, while FoxG1 downregulation was observed and then FoxG1 was definitely validated as one miR-200b target. Further in vitro studies showed that enforced miR-200b downregulation induced the decrease of cell ability, with increased cell apoptosis, and attenuated ability of cell migration and invasion in both HeLa and C33A cells, while further inhibition of FoxG1 expression could reverse all these changes. In addition, miR-200b silence in vivo strongly inhibited tumor growth. CONCLUSION Upregulated miR-200b in cervical cancer was proven to show positive regulation on cervical cancer development by directly targeting FoxG1. Moreover, miR-200b silence was proposed to inhibit tumor growth in vivo, implying its therapeutic value in cervical cancer treatment.
Collapse
Affiliation(s)
- Fei Zeng
- Department of Gynecology, the Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Min Xue
- Department of Gynecology, the Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China.
| | - Ting Xiao
- Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Yueran Li
- Department of Gynecology, the Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Songshu Xiao
- Department of Gynecology, the Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Bin Jiang
- Department of Gynecology, the Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Caiping Ren
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, School of Basic Medical Science, Central South University, Xiangya Road 110, Changsha 410078, Hunan Province, China.
| |
Collapse
|
221
|
Abstract
The hedgehog signaling pathway was first discovered in the 1980s. It is a stem cell-related pathway that plays a crucial role in embryonic development, tissue regeneration, and organogenesis. Aberrant activation of hedgehog signaling leads to pathological consequences, including a variety of human tumors such as pancreatic cancer. Multiple lines of evidence indicate that blockade of this pathway with several small-molecule inhibitors can inhibit the development of pancreatic neoplasm. In addition, activated hedgehog signaling has been reported to be involved in fibrogenesis in many tissues, including the pancreas. Therefore, new therapeutic targets based on hedgehog signaling have attracted a great deal of attention to alleviate pancreatic diseases. In this review, we briefly discuss the recent advances in hedgehog signaling in pancreatic fibrogenesis and carcinogenesis and highlight new insights on their potential relationship with respect to the development of novel targeted therapies.
Collapse
Affiliation(s)
- Yongyu Bai
- From the Wenzhou Medical University (Yongyu Bai, JD, QL, YJ, MZ); and Wenzhou Key Laboratory of Surgery (Yongheng Bai, BC), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | | | | | | | | | | |
Collapse
|
222
|
Yang H, Lu X, Liu Z, Chen L, Xu Y, Wang Y, Wei G, Chen Y. FBXW7 suppresses epithelial-mesenchymal transition, stemness and metastatic potential of cholangiocarcinoma cells. Oncotarget 2016; 6:6310-25. [PMID: 25749036 PMCID: PMC4467439 DOI: 10.18632/oncotarget.3355] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/17/2015] [Indexed: 12/11/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays a fundamental role in cancer metastasis. The ubiquitin ligase FBXW7, a general tumor suppressor in human cancer, has been implicated in diverse cellular processes, however, its role in cholangiocarcinoma (CCA) metastasis has not been identified. Here, we report a crucial role of FBXW7 in CCA metastasis by regulating EMT. Loss of FBXW7 expression was detected in CCA cells and clinical specimens. Clinicopathological analysis revealed a close correlation between FBXW7 deficiency and metastasis, TNM stage and differentiation in intrahepatic CCA and perihilar CCA. Moreover, FBXW7 silencing in CCA cells dramatically promoted EMT, stem-like capacity and metastasis both in vitro and in vivo. Conversely, FBXW7 overexpression attenuated these processes. Mechanistically, treatment with rapamycin, a mTOR inhibitor, inhibited EMT, stem-like capacity and metastasis induced by FBXW7 silencing both in vitro and in vivo. Furthermore, the expression of EMT regulating transcription factors, snail, slug and ZEB1, were also decreased markedly with rapamycin treatment. In addition, silencing ZEB1 inhibited EMT and metastasis of both CCA cells and FBXW7 deficient CCA cells, which implicated the potential role of ZEB1 in FBXW7/mTOR signaling pathway related CCA metastasis. In conclusion, our findings defined a pivotal function of FBXW7 in CCA metastasis by regulating EMT.
Collapse
Affiliation(s)
- Hui Yang
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaofei Lu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China.,Department of General Surgery, Jinan Central Hospital of Shandong University, Jinan, China
| | - Ziming Liu
- Department of Emergency Medicine, Jinan Fifth People's Hospital, Jinan, China
| | - Lili Chen
- Department of Pathology, Jinan Fourth People's Hospital, Jinan, China
| | - Yunfei Xu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yuli Wang
- Department of Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, China
| | - Guangwei Wei
- Department of Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, China
| | - Yuxin Chen
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
223
|
Meidhof S, Brabletz S, Lehmann W, Preca BT, Mock K, Ruh M, Schüler J, Berthold M, Weber A, Burk U, Lübbert M, Puhr M, Culig Z, Wellner U, Keck T, Bronsert P, Küsters S, Hopt UT, Stemmler MP, Brabletz T. ZEB1-associated drug resistance in cancer cells is reversed by the class I HDAC inhibitor mocetinostat. EMBO Mol Med 2016; 7:831-47. [PMID: 25872941 PMCID: PMC4459821 DOI: 10.15252/emmm.201404396] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Therapy resistance is a major clinical problem in cancer medicine and crucial for disease relapse and progression. Therefore, the clinical need to overcome it, particularly for aggressive tumors such as pancreatic cancer, is very high. Aberrant activation of an epithelial-mesenchymal transition (EMT) and an associated cancer stem cell phenotype are considered a major cause of therapy resistance. Particularly, the EMT-activator ZEB1 was shown to confer stemness and resistance. We applied a systematic, stepwise strategy to interfere with ZEB1 function, aiming to overcome drug resistance. This led to the identification of both its target gene miR-203 as a major drug sensitizer and subsequently the class I HDAC inhibitor mocetinostat as epigenetic drug to interfere with ZEB1 function, restore miR-203 expression, repress stemness properties, and induce sensitivity against chemotherapy. Thereby, mocetinostat turned out to be more effective than other HDAC inhibitors, such as SAHA, indicating the relevance of the screening strategy. Our data encourage the application of mechanism-based combinations of selected epigenetic drugs with standard chemotherapy for the rational treatment of aggressive solid tumors, such as pancreatic cancer.
Collapse
Affiliation(s)
- Simone Meidhof
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Freiburg, Germany Faculty of Biology, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Simone Brabletz
- Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Waltraut Lehmann
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany Faculty of Biology, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Bogdan-Tiberius Preca
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany Faculty of Biology, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Kerstin Mock
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany Faculty of Biology, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Manuel Ruh
- Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Schüler
- Oncotest GmbH, Institute for Experimental Oncology, Freiburg, Germany
| | - Maria Berthold
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - Anika Weber
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - Ulrike Burk
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - Michael Lübbert
- Department of Hematology and Oncology, University of Freiburg Medical Center, Freiburg, Germany German Cancer Consortium (DKTK), Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Puhr
- Division of Experimental Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Zoran Culig
- Division of Experimental Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Ulrich Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany
| | - Peter Bronsert
- Tumorbank Comprehensive Cancer Center Freiburg and Institute of Surgical Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Simon Küsters
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - Ulrich T Hopt
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - Marc P Stemmler
- Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany German Cancer Consortium (DKTK), Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
224
|
Abstract
Chemoresistant metastatic relapse of minimal residual disease plays a significant role for poor prognosis of cancer. Growing evidence supports a critical role of cancer stem cell (CSC) behind the mechanisms for this deadly disease. This review briefly introduces the basics of the conventional chemotherapies, updates the CSC theories, highlights the molecular and cellular mechanisms by which CSC smartly designs and utilizes multiple lines of self-defense to avoid being killed by chemotherapy, and concisely summarizes recent progress in studies on CSC-targeted therapies in the end, with the hope to help guide future research toward developing more effective therapeutic strategies to eradicate tumor cells in the patients.
Collapse
Affiliation(s)
- Jihe Zhao
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 6900 Lake Nona Boulevard, Orlando, FL 32827, USA.
| |
Collapse
|
225
|
Noncoding RNAs in Tumor Epithelial-to-Mesenchymal Transition. Stem Cells Int 2016; 2016:2732705. [PMID: 26989421 PMCID: PMC4773551 DOI: 10.1155/2016/2732705] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/20/2016] [Indexed: 12/21/2022] Open
Abstract
Epithelial-derived tumor cells acquire the capacity for epithelial-to-mesenchymal transition (EMT), which enables them to invade adjacent tissues and/or metastasize to distant organs. Cancer metastasis is the main cause of cancer-related death. Molecular mechanisms involved in the switch from an epithelial phenotype to mesenchymal status are complicated and are controlled by a variety of signaling pathways. Recently, a set of noncoding RNAs (ncRNAs), including miRNAs and long noncoding RNAs (lncRNAs), were found to modulate gene expressions at either transcriptional or posttranscriptional levels. These ncRNAs are involved in EMT through their interplay with EMT-related transcription factors (EMT-TFs) and EMT-associated signaling. Reciprocal regulatory interactions between lncRNAs and miRNAs further increase the complexity of the regulation of gene expression and protein translation. In this review, we discuss recent findings regarding EMT-regulating ncRNAs and their associated signaling pathways involved in cancer progression.
Collapse
|
226
|
Ahmad A, Li Y, Sarkar FH. The bounty of nature for changing the cancer landscape. Mol Nutr Food Res 2016; 60:1251-63. [PMID: 26799714 DOI: 10.1002/mnfr.201500867] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/11/2016] [Accepted: 01/11/2016] [Indexed: 12/18/2022]
Abstract
The landscape of cancer has changed considerably in past several years, due mainly to aggressive screening, accumulation of data from basic and epidemiological studies, and the advances in translational research. Natural anticancer agents have always been a part and parcel of cancer research. The initial focus on natural anticancer agents was in context of their cancer chemopreventive properties but their ability to selectively target oncogenic signaling pathways has also been recognized. In light of the rapid advancements in our understanding of the role of microRNAs, cancer stem cells, and epigenetic events in cancer initiation and progression, a number of natural anticancer agents are showing promise in vitro, in vivo as well as in preclinical studies. Moreover, parent structures of natural agents are being extensively modified with the hope of improving efficacy, specificity, and bioavailability. In this article, we focus on two natural agents, 3,3'-diindolylmethane and garcinol, along with 3,4-difluorobenzo curcumin, a synthetic analog of natural agent curcumin. We showcase how these anticancer agents are changing cancer landscape by modulating novel microRNAs, epigenetic factors, and cancer stem cell markers. These activities are relevant and being appreciated for overcoming drug resistance and inhibition of metastases, the two overarching clinical challenges in modern medicine.
Collapse
Affiliation(s)
- Aamir Ahmad
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Yiwei Li
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Fazlul H Sarkar
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA.,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
227
|
Liang S, Gong X, Zhang G, Huang G, Lu Y, Li Y. MicroRNA-140 regulates cell growth and invasion in pancreatic duct adenocarcinoma by targeting iASPP. Acta Biochim Biophys Sin (Shanghai) 2016; 48:174-81. [PMID: 26787707 DOI: 10.1093/abbs/gmv127] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/21/2015] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs are ∼22 nucleotide RNAs processed from RNA hairpin structures that play important roles in regulating protein expression level via binding to mRNA, either suppressing its translation or speeding up its degradation. In humans, they regulate most protein-coding genes, including genes important in cancer and other diseases. In this study, the expression of microRNA-140 (miR-140) was demonstrated to be significantly suppressed in pancreatic duct adenocarcinoma specimens and cell lines, compared with their adjacent normal tissues. With the help of bioinformatics analysis, inhibitor of apoptosis-stimulating protein of p53 (iASPP) was identified to be a direct target of miR-140, and luciferase reporter experiment confirmed this discovery. Overexpression of miR-140 decreases the protein expressions of iASPP, ΔNp63, MMP2, and MMP9. Growth and invasion of PANC-1 cells were attenuated by overexpression of miR-140 in vitro. The suppressive effect of miR-140 on PANC-1 cell line could be partly balanced out by manual overexpression of iASPP. Above all, these findings provided insights into the functional mechanism of miR-140, suggested that the miR-140/iASPP axis may interfere with the proliferative and invasive property of pancreatic duct adenocarcinoma cells, and indicated that miR-140 could be a potential therapeutic target for pancreatic duct adenocarcinoma.
Collapse
Affiliation(s)
- Shuai Liang
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuejun Gong
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Gewen Zhang
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Gengwen Huang
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yebin Lu
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yixiong Li
- Department of Pancreatic Biliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
228
|
Missing link between microRNA and prostate cancer. Tumour Biol 2016; 37:5683-704. [DOI: 10.1007/s13277-016-4900-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/20/2016] [Indexed: 12/12/2022] Open
|
229
|
Abstract
Pancreatic cancer remains the fourth leading cause of cancer-related death in the US and is expected to be the second leading cause of cancer-related death by 2030. Therefore, it is important to better understand the molecular pathogenesis, phenotypes and features of pancreatic cancer in order to design novel molecularly targeted therapies for achieving better therapeutic outcome of patients with pancreatic cancer. Recently, the roles of microRNAs (miRNAs) in the development and progression of pancreatic cancer became a hot topic in the scientific community of pancreatic cancer research. By conducting miRNA expression profiling, the aberrant expression of miRNAs was revealed in the serum and in cancer tissues from patients with pancreatic cancer. These aberrantly expressed miRNAs are critically correlated with the disease stage, drug resistance, and survival of pancreatic cancer patients. Hence, targeting these tiny molecules, the specific miRNAs, could provide an efficient and optimal approach in the therapy of pancreatic cancer. Indeed, the pre-clinical and in vivo experiments showed that nanoparticle delivery of synthetic oligonucleotides or treatment with natural agents could be useful to modulate the expression of miRNAs and thereby inhibit pancreatic cancer growth and progression, suggesting that targeting miRNAs combined with conventional anti-cancer therapeutics could be a novel therapeutic strategy for increasing drug sensitivity and achieving better therapeutic outcome of patients diagnosed with pancreatic cancer.
Collapse
Affiliation(s)
- Yiwei Li
- 1. Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Fazlul H Sarkar
- 1. Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; 2. Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
230
|
Venkatesan N, Kanwar J, Deepa PR, Khetan V, Crowley TM, Raguraman R, Sugneswari G, Rishi P, Natarajan V, Biswas J, Krishnakumar S. Clinico-Pathological Association of Delineated miRNAs in Uveal Melanoma with Monosomy 3/Disomy 3 Chromosomal Aberrations. PLoS One 2016; 11:e0146128. [PMID: 26812476 PMCID: PMC4728065 DOI: 10.1371/journal.pone.0146128] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/14/2015] [Indexed: 12/15/2022] Open
Abstract
Purpose To correlate the differentially expressed miRNAs with clinico-pathological features in uveal melanoma (UM) tumors harbouring chromosomal 3 aberrations among South Asian Indian cohort. Methods Based on chromosomal 3 aberration, UM (n = 86) were grouped into monosomy 3 (M3; n = 51) and disomy 3 (D3; n = 35) by chromogenic in-situ hybridisation (CISH). The clinico-pathological features were recorded. miRNA profiling was performed in formalin fixed paraffin embedded (FFPE) UM samples (n = 6) using Agilent, Human miRNA microarray, 8x15KV3 arrays. The association between miRNAs and clinico-pathological features were studied using univariate and multivariate analysis. miRNA-gene targets were predicted using Target-scan and MiRanda database. Significantly dys-regulated miRNAs were validated in FFPE UM (n = 86) and mRNAs were validated in frozen UM (n = 10) by qRT-PCR. Metastasis free-survival and miRNA expressions were analysed by Kaplen-Meier analysis in UM tissues (n = 52). Results Unsupervised analysis revealed 585 differentially expressed miRNAs while supervised analysis demonstrated 82 miRNAs (FDR; Q = 0.0). Differential expression of 8 miRNAs: miR-214, miR-149*, miR-143, miR-146b, miR-199a, let7b, miR-1238 and miR-134 were studied. Gene target prediction revealed SMAD4, WISP1, HIPK1, HDAC8 and C-KIT as the post-transcriptional regulators of miR-146b, miR-199a, miR-1238 and miR-134. Five miRNAs (miR-214, miR146b, miR-143, miR-199a and miR-134) were found to be differentially expressed in M3/ D3 UM tumors. In UM patients with liver metastasis, miR-149* and miR-134 expressions were strongly correlated. Conclusion UM can be stratified using miRNAs from FFPE sections. miRNAs predicting liver metastasis and survival have been identified. Mechanistic linkage of de-regulated miRNA/mRNA expressions provide new insights on their role in UM progression and aggressiveness.
Collapse
Affiliation(s)
- Nalini Venkatesan
- Larsen & Toubro Department of Ocular Pathology, Vision Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani-333031, Rajasthan, India
| | - Jagat Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Molecular and Medical Research (MMR) Strategic Research Centre, Faculty of Health, Deakin University, Pigdons Road, Waurn Ponds, Geelong, Victoria 3217, Australia
| | - Perinkulam Ravi Deepa
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani-333031, Rajasthan, India
| | - Vikas Khetan
- Department of Vitreoretinal and Ocular Oncology, Medical Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
| | - Tamsyn M. Crowley
- School of Medicine, Deakin University, and Australian Animal Health Laboratories, CSIRO, Australia
| | - Rajeswari Raguraman
- Larsen & Toubro Department of Ocular Pathology, Vision Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
| | - Ganesan Sugneswari
- Department of Vitreoretinal and Ocular Oncology, Medical Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
| | - Pukhraj Rishi
- Department of Vitreoretinal and Ocular Oncology, Medical Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
| | - Viswanathan Natarajan
- Department of Bio-statistics, Medical Research Foundation, Sankara Nethralaya, 41, College road, Chennai—600006, India
| | - Jyotirmay Biswas
- Larsen & Toubro Department of Ocular Pathology, Vision Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
| | - Subramanian Krishnakumar
- Larsen & Toubro Department of Ocular Pathology, Vision Research Foundation, Sankara Nethralaya, 18/41, College road, Chennai—600006, India
- * E-mail:
| |
Collapse
|
231
|
Wang F, Ma L, Zhang Z, Liu X, Gao H, Zhuang Y, Yang P, Kornmann M, Tian X, Yang Y. Hedgehog Signaling Regulates Epithelial-Mesenchymal Transition in Pancreatic Cancer Stem-Like Cells. J Cancer 2016; 7:408-17. [PMID: 26918054 PMCID: PMC4749361 DOI: 10.7150/jca.13305] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/29/2015] [Indexed: 01/05/2023] Open
Abstract
Hedgehog (Hh) signaling is crucially involved in tumorigenesis. This study aimed to assess the role of Hh signaling in the regulation of epithelial-mesenchymal transition (EMT), stemness properties and chemoresistance of human pancreatic Panc-1 cancer stem cells (CSCs). Panc-1 cells were transfected with recombinant lentiviral vectors to silence SMO and serum-free floating-culture system was used to isolate Panc-1 tumorspheres. The expression of CSC and EMT markers was detected by flow cytometry, real-time RT-PCR and Western blot analysis. Malignant behaviors of Panc-1 CSC were evaluated by tumorigenicity assays and nude mouse lung metastasis model. We found that tumorspheres derived from pancreatic cancer cell line Panc-1 possessed self-renewal, differentiation and stemness properties. Hh pathway and EMT were active in Panc-1 tumorspheres. Inhibition of Hh signaling by SMO knockdown inhibited self-renewal, EMT, invasion, chemoresistance, pulmonary metastasis, tumorigenesis of pancreatic CSCs. In conclusion, Hh signaling contributes to the maintenance of stem-like properties and chemoresistance of pancreatic CSC and promotes the tumorigenesis and metastasis of pancreatic cancer. Hh pathway is a potential molecular target for the development of therapeutic strategies for pancreatic CSCs.
Collapse
Affiliation(s)
- Feng Wang
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Ling Ma
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Zhengkui Zhang
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Xiaoran Liu
- 4. Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hongqiao Gao
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Yan Zhuang
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Pei Yang
- 2. Department of Agricultural & Biological Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Marko Kornmann
- 3. Clinic of General, Visceral and Transplantation Surgery, University of Ulm, Ulm 89081, Germany
| | - Xiaodong Tian
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Yinmo Yang
- 1. Department of General Surgery, Peking University First Hospital, Beijing 100034, People's Republic of China
| |
Collapse
|
232
|
The miR-491-3p/mTORC2/FOXO1 regulatory loop modulates chemo-sensitivity in human tongue cancer. Oncotarget 2016; 6:6931-43. [PMID: 25749387 PMCID: PMC4466660 DOI: 10.18632/oncotarget.3165] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/18/2015] [Indexed: 12/14/2022] Open
Abstract
We found that levels of miR-491-3p were decreased in multidrug-resistant tongue cancer (TC) cells. Induction of miR-491-3p expression sensitized TC cells to chemotherapy. In agreement, functional inhibition of miR-491-3p enhanced resistance of TC cells to chemotherapy. We found that miR-491-3p directly targeted mTORC2 component Rictor and inhibited mTORC2 activity, which was increased in resistant TC cells with high p-Akt(Ser473), p-SGK1(Ser422) and p-FOXO1(Thr24) levels. Inhibition of mTORC2 activity via either Rictor knockdown or mTOR inhibitor in turn sensitized TC cells to chemotherapy. In agreement, overexpression of Rictor increased the mTORC2 activity and induced resistance of TC cells to chemotherapy. As a feedback loop, mTORC2 downregulated miR-491-3p expression by inactivating FOXO1, which otherwise would transcriptionally induce miR-491-3p expression. Levels of miR-491-3 and Rictor or mTORC2 activity negatively correlated in TC tissues. Finally, low levels of miR-491-3p and highly expressed Rictor were associated with poor prognosis in tongue cancer patients. These data provide a rationale for targeted intervention on miR-491-3p/mTORC2 axis to enhance the efficacy of chemotherapy against tongue cancer.
Collapse
|
233
|
Wang H, Zhao Q, Deng K, Guo X, Xia J. Lin28: an emerging important oncogene connecting several aspects of cancer. Tumour Biol 2016; 37:2841-8. [PMID: 26762415 DOI: 10.1007/s13277-015-4759-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/29/2015] [Indexed: 12/29/2022] Open
Abstract
RNA-binding protein Lin28 was originally found as a heterochronic gene which played a significant role in the development of Caenorhabditis elegans. The tumor suppressor let-7 is a downstream target of Lin28, which has a wide variety of target genes which are involved in many aspects of cellular activities. By inhibition of let-7 and directly binding the target RNAs, Lin28 plays an important role in different biological and pathological processes including differentiation, metabolism, proliferation, pluripotency, and tumorigenesis. Overexpression of Lin28 has been reported in several kinds of cancers and is correlated with poor outcomes. It has been shown that Lin28 could affect the progression of cancers in several ways, such as promoting proliferation, increasing glucose metabolism, and inducing epithelial-mesenchymal transition (EMT) and cancer stem cells. Decrease of Lin28 expression or reactivation of let-7 in cancer cells could induce a reverse effect, indicating their therapeutic values in developing novel strategies for cancer treatment. Here, we will overview the regulatory mechanisms and functions of Lin28 in cancers.
Collapse
Affiliation(s)
- Hao Wang
- Department of General Surgery and Translational Medicine Center, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214002, Jiangsu, China
| | - Qin Zhao
- Department of General Surgery and Translational Medicine Center, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214002, Jiangsu, China
| | - Kaiyuan Deng
- Department of General Surgery and Translational Medicine Center, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiaoqiang Guo
- Department of General Surgery and Translational Medicine Center, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214002, Jiangsu, China
| | - Jiazeng Xia
- Department of General Surgery and Translational Medicine Center, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
234
|
Peng JF, Zhuang YY, Huang FT, Zhang SN. Noncoding RNAs and pancreatic cancer. World J Gastroenterol 2016; 22:801-814. [PMID: 26811626 PMCID: PMC4716078 DOI: 10.3748/wjg.v22.i2.801] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023] Open
Abstract
Noncoding RNAs (ncRNAs) represent a class of RNA molecules that typically do not code for proteins. Emerging data suggest that ncRNAs play an important role in several physiological and pathological conditions such as cancer. The best-characterized ncRNAs are the microRNAs (miRNAs), which are short, approximately 22-nucleotide sequences of RNA of approximately 22-nucleotide in length that regulate gene expression at the posttranscriptional level, through transcript degradation or translational repression. MiRNAs can function as master gene regulators, impacting a variety of cellular pathways important to normal cellular functions as well as cancer development and progression. In addition to miRNAs, long ncRNAs, which are transcripts longer than 200 nucleotides, have recently emerged as novel drivers of tumorigenesis. However, the molecular mechanisms of their regulation and function, and the significance of other ncRNAs such as piwi-interacting RNAs in pancreas carcinogenesis are largely unknown. This review summarizes the growing body of evidence supporting the vital roles of ncRNAs in pancreatic cancer, focusing on their dysregulation through both genetic and epigenetic mechanisms, and highlighting the promise of ncRNAs in diagnostic and therapeutic applications of pancreatic cancer.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Predictive Value of Tests
- Prognosis
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- RNA, Untranslated/therapeutic use
- Transcription, Genetic
Collapse
|
235
|
Singh R, Lillard JW, Singh S. Epigenetic Changes and Potential Targets in Pancreatic Cancer. EPIGENETIC ADVANCEMENTS IN CANCER 2016:27-63. [DOI: 10.1007/978-3-319-24951-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
236
|
Jia L, Yang A. Noncoding RNAs in Therapeutic Resistance of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016:265-295. [DOI: 10.1007/978-981-10-1498-7_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
237
|
Garajová I, Le Large TYS, Giovannetti E, Kazemier G, Biasco G, Peters GJ. The Role of MicroRNAs in Resistance to Current Pancreatic Cancer Treatment: Translational Studies and Basic Protocols for Extraction and PCR Analysis. Methods Mol Biol 2016; 1395:163-187. [PMID: 26910074 DOI: 10.1007/978-1-4939-3347-1_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a common cause of cancer death and has the worst prognosis of any major malignancy, with less than 5 % of patients alive 5-years after diagnosis. The therapeutic options for metastatic PDAC have changed in the past few years from single agent gemcitabine treatment to combination regimens. Nowadays, FOLFIRINOX or gemcitabine with nab-paclitaxel are new standard combinations in frontline metastatic setting in PDAC patients with good performance status. MicroRNAs (miRNA) are small, noncoding RNA molecules affecting important cellular processes such as inhibition of apoptosis, cell proliferation, epithelial-to-mesenchymal transition (EMT), metastases, and resistance to common cytotoxic and anti-signaling therapy in PDAC. A functional association between miRNAs and chemoresistance has been described for several common therapies. Therefore, in this review, we summarize the current knowledge on the role of miRNAs in the resistance to current anticancer treatment used for patients affected by metastatic PDAC.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, CCA 1.42, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Tessa Y S Le Large
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, CCA 1.42, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Department of Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, CCA 1.42, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Geert Kazemier
- Department of Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Guido Biasco
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, CCA 1.42, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands.
| |
Collapse
|
238
|
Ren ZG, Dong SX, Han P, Qi J. miR-203 promotes proliferation, migration and invasion by degrading SIK1 in pancreatic cancer. Oncol Rep 2015; 35:1365-74. [PMID: 26719072 DOI: 10.3892/or.2015.4534] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/09/2015] [Indexed: 11/05/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the most lethal human cancers and it is insensitive to many chemotherapeutic drugs. The molecular basis of pancreatic cancer remains to be elucidated. Investigations into the molecular mechanism involved in the development and progression as well as drug resistance of the disease may be useful to understand the pathogenesis and progression of the disease and offer new targets for effective therapies. In the present study, we showed that salt-inducible kinase 1 (SIK1) was downregulated and loss of SIK1 was associated with gemcitabine resistance in pancreatic cancer. In pancreatic cancer cells, SIK1 inhibited proliferation, migration and invasion. An analysis of potential microRNA target sites was performed using the prediction algorithms, miRanda, TargetScan and PicTar. The three algorithms predicted that miR-203 is capable of targeting 3'UTR of SIK1. Subsequent experiments confirmed the prediction. In addition, the results showed that miR-203 promotes proliferation, migration and invasion in pancreatic cancer cells, whereas the restoration of SIK1 abrogated the regulation of pre-miR‑203-mediated proliferation, migration and invasion.
Collapse
Affiliation(s)
- Zhi-Guo Ren
- Department of General Surgery, Affiliated Hospital of Shandong Medical College, Linyi, Shandong 276004, P.R. China
| | - Shu-Xiao Dong
- The Second Department of General Surgery, Linyi People's Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| | - Ping Han
- The Second Department of General Surgery, Linyi People's Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| | - Jian Qi
- The Second Department of General Surgery, Linyi People's Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
239
|
Alemar B, Gregório C, Ashton-Prolla P. miRNAs As Diagnostic and Prognostic Biomarkers in Pancreatic Ductal Adenocarcinoma and Its Precursor Lesions: A Review. Biomark Insights 2015; 10:113-24. [PMID: 26688661 PMCID: PMC4677802 DOI: 10.4137/bmi.s27679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/30/2015] [Accepted: 09/06/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a rare but lethal tumor, is difficult to diagnose without performing an invasive procedure. miRNAs are known to be deregulated in PDAC patients, and recent studies have shown that they can be used as diagnostic and prognostic of the disease. The detection of miRNAs in samples acquired through minimally or noninvasive procedures, such as serum, plasma, and saliva, can have a positive impact on the clinical management of these patients. This article is a comprehensive review of the major studies that have evaluated the expression of miRNAs as biomarkers in pancreatic cancer and its premalignant lesions.
Collapse
Affiliation(s)
- Bárbara Alemar
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cleandra Gregório
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Ashton-Prolla
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
240
|
Samadi AK, Bilsland A, Georgakilas AG, Amedei A, Amin A, Bishayee A, Azmi AS, Lokeshwar BL, Grue B, Panis C, Boosani CS, Poudyal D, Stafforini DM, Bhakta D, Niccolai E, Guha G, Vasantha Rupasinghe HP, Fujii H, Honoki K, Mehta K, Aquilano K, Lowe L, Hofseth LJ, Ricciardiello L, Ciriolo MR, Singh N, Whelan RL, Chaturvedi R, Ashraf SS, Shantha Kumara HMC, Nowsheen S, Mohammed SI, Keith WN, Helferich WG, Yang X. A multi-targeted approach to suppress tumor-promoting inflammation. Semin Cancer Biol 2015; 35 Suppl:S151-S184. [PMID: 25951989 PMCID: PMC4635070 DOI: 10.1016/j.semcancer.2015.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.
Collapse
Affiliation(s)
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Asfar S Azmi
- Department of Pathology, Wayne State Univeristy, Karmanos Cancer Center, Detroit, MI, USA
| | - Bal L Lokeshwar
- Department of Urology, University of Miami, Miller School of Medicine, Miami, FL, United States; Miami Veterans Administration Medical Center, Miami, FL, United States
| | - Brendan Grue
- Department of Environmental Science, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada.
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
241
|
Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna J, Marino M, Martinez-Chantar ML, Nawroth R, Sanchez-Garcia I, Sharma D, Saxena NK, Singh N, Vlachostergios PJ, Guo S, Honoki K, Fujii H, Georgakilas AG, Bilsland A, Amedei A, Niccolai E, Amin A, Ashraf SS, Boosani CS, Guha G, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Keith WN, Nowsheen S. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin Cancer Biol 2015; 35 Suppl:S25-S54. [PMID: 25892662 PMCID: PMC4898971 DOI: 10.1016/j.semcancer.2015.02.006] [Citation(s) in RCA: 483] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 02/08/2023]
Abstract
Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.
Collapse
Affiliation(s)
- Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States.
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Jamal Mahajna
- MIGAL-Galilee Technology Center, Cancer Drug Discovery Program, Kiryat Shmona, Israel
| | - Maria Marino
- Department of Science, University Roma Tre, V.le G. Marconi, 446, 00146 Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Dipali Sharma
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Neeraj K Saxena
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Neetu Singh
- Tissue and Cell Culture Unit, CSIR-Central Drug Research Institute, Council of Scientific & Industrial Research, Lucknow, India
| | | | - Shanchun Guo
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - Chandra S Boosani
- Department of BioMedical Sciences, Creighton University, Omaha, NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Sophie Chen
- Department of Research and Development, Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey GU2 7YG, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Asfar S Azmi
- Department of Pathology, Karmonas Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dorota Halicka
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| |
Collapse
|
242
|
Down-regulation of miR-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Oncotarget 2015; 6:1740-9. [PMID: 25638153 PMCID: PMC4359328 DOI: 10.18632/oncotarget.2714] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022] Open
Abstract
Recent studies have demonstrated that acquisition of epithelial-to-mesenchymal transition (EMT) is associated with drug resistance in pancreatic cancer cells; however, the underlying mechanisms are not fully elucidated. Emerging evidence suggests that microRNAs play a crucial role in controlling EMT. The aims of this study were to explore the potential role of miR-223 in governing EMT in gemcitabine-resistant (GR) pancreatic cancer cells. To achieve this goal, real-time reverse transcription-PCR and western blot analysis were used to validate whether GR cells acquired EMT in AsPC-1 and PANC-1 cells. Invasion, migration, and detachment assays were performed to further identify the EMT characteristics in GR cells. The miR-223 inhibitor was used to determine its role in GR-induced EMT. We found that GR cells acquired EMT features, which obtained elongated fibroblastoid morphology, decreased expression of epithelial marker E-cadherin, and up-regulation of mesenchymal markers. Furthermore, we observed that GR cells are associated with high expression of miR-223. Notably, inhibition of miR-223 led to the reversal of EMT phenotype. More importantly, miR-223 governs GR-induced EMT in part due to down-regulation of its target Fbw7 and subsequent upregulation of Notch-1 in pancreatic cancer. Our study implied that down-regulation of miR-223 could be a novel therapy for pancreatic cancer.
Collapse
|
243
|
Sun C, Sang M, Li S, Sun X, Yang C, Xi Y, Wang L, Zhang F, Bi Y, Fu Y, Li D. Hsa-miR-139-5p inhibits proliferation and causes apoptosis associated with down-regulation of c-Met. Oncotarget 2015; 6:39756-92. [PMID: 26497851 PMCID: PMC4741860 DOI: 10.18632/oncotarget.5476] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 10/07/2015] [Indexed: 01/01/2023] Open
Abstract
Hsa-miRNA-139-5p (miR-139-5p) has recently been discovered having anticancer efficacy in different organs. However, the role of miR-139-5p on lung cancer is still ambiguous. In this study, we investigated the role of miR-139-5p on development of lung cancer. Results indicated miR-139-5p was significantly down-regulated in primary tumor tissues and very low levels were found in a non-small cell lung cancer (NSCLC) cell lines. Ectopic expression of miR-139-5p in NSCLC cell lines significantly suppressed cell growth through inhibition of cyclin D1 and up-regulation of p57(Kip2). In addition, miR-139-5p induced apoptosis, as indicated by up-regulation of key apoptosis gene cleaved caspase-3, and down-regulation of anti-apoptosis gene Bcl2. Moreover, miR-139-5p inhibited cellular metastasis through inhibition of matrix metalloproteinases (MMP)-7 and MMP-9. Further, oncogene c-Met was revealed to be a putative target of miR-139-5p, which was inversely correlated with miR-139-5p expression. Taken together, our results demonstrated that miR-139-5p plays a pivotal role in lung cancer through inhibiting cell proliferation, metastasis, and promoting apoptosis by targeting oncogenic c-Met.
Collapse
Affiliation(s)
- Chengcao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
- Institute of Global Health, Wuhan University, 430071 Wuhan, P. R. China
| | - Ming Sang
- Central Laboratory of the Fourth Affiliated Hospital in Xiangyang, College of Basic Medical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 442000 Shiyan, P. R. China
| | - Shujun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
- Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, 430071 Wuhan, P. R. China
| | - Xiaodong Sun
- Central Laboratory of the Fourth Affiliated Hospital in Xiangyang, College of Basic Medical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 442000 Shiyan, P. R. China
| | - Cuili Yang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Yongyong Xi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Liang Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Feng Zhang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Yongyi Bi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| | - Yunfeng Fu
- The Third Xiang-ya Hospital of Central South University, 410013 Changsha, P. R. China
| | - Dejia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P.R.China
| |
Collapse
|
244
|
Gemcitabine resistance in pancreatic ductal adenocarcinoma. Drug Resist Updat 2015; 23:55-68. [PMID: 26690340 DOI: 10.1016/j.drup.2015.10.002] [Citation(s) in RCA: 296] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 09/15/2015] [Accepted: 10/23/2015] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) ranks fourth among cancer related deaths. The disappointing 5-year survival rate of below 5% stems from drug resistance to all known therapies, as well as from disease presentation at a late stage when PDA is already metastatic. Gemcitabine has been the cornerstone of PDA treatment in all stages of the disease for the last two decades, but gemcitabine resistance develops within weeks of chemotherapy initiation. From a mechanistic perspective, gemcitabine resistance may result from alterations in drug metabolism until the point that the cytidine analog is incorporated into the DNA, or from mitigation of gemcitabine-induced apoptosis. Both of these drug resistance modalities can be either intrinsic to the cancer cell, or influenced by the cancer microenvironment. Mechanisms of intrinsic gemcitabine resistance are difficult to tackle, as many of the genes that drive the carcinogenic process itself also interfere with gemcitabine-induced apoptosis. In this regard, recent understanding of the involvement of microRNAs in gemcitabine resistance may offer new opportunities to overcome intrinsic gemcitabine resistance. The characteristically fibrotic and immune infiltrated stroma of PDA that accompanies tumor inception and expansion is a lush ground for treatments aimed at targeting tumor microenvironment-mediated drug resistance. In the last couple of years, drugs interfering with tumor microenvironment have matured to clinical trials. Although drugs inducing 'stromal depletion' have yet failed to improve survival, they have greatly increased our understanding of tumor microenvironment-mediated drug resistance. In this review we summarize the current knowledge on intrinsic and environment-mediated gemcitabine resistance, and discuss the impact of these pathways on patient screening, and on future treatments aimed to potentiate gemcitabine activity.
Collapse
|
245
|
Li Y, Sarkar FH. Role of BioResponse 3,3'-Diindolylmethane in the Treatment of Human Prostate Cancer: Clinical Experience. Med Princ Pract 2015; 25 Suppl 2:11-7. [PMID: 26501150 PMCID: PMC4848191 DOI: 10.1159/000439307] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/11/2015] [Indexed: 01/09/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) progression after androgen deprivation therapy shows upregulated expression of androgen receptor (AR) splice variants, induced epithelial-to-mesenchymal transition phenotypes and enhanced stem cell characteristics, all of which are associated with resistance to enzalutamide. Since there is no curative treatment for CRPC, innovative treatments are urgently needed. In our recent study, we found that resistance to enzalutamide was partly due to deregulated expression of microRNAs such as miR-34a, miR-124, miR-27b, miR-320 and let-7, which play important roles in regulating AR and stem cell marker gene expression that appears to be linked with resistance to enzalutamide. Importantly, we found that BioResponse 3,3'-diindolylmethane (BR-DIM) treatment in vitro and in vivo caused downregulation in the expression of wild-type AR. The AR splice variants, Lin28B and EZH2, appear to be deregulated through the re-expression of let-7, miR-27b, miR-320 and miR-34a in human prostate cancer (PCa). BR-DIM administered in clinical trials was well tolerated, and 93% of patients had detectable prostatic DIM levels. The inhibitory effects of BR-DIM on AR and AR target gene such as prostate-specific antigen were also observed in the clinical trial. Our preclinical and clinical studies provide the scientific basis for a 'proof-of-concept' clinical trial in CRPC patients treated with enzalutamide in combination with BR-DIM. This strategy could be expanded in future clinical trials in patients with PCa to determine whether or not they could achieve a better treatment outcome which could be partly mediated by delaying or preventing the development of CRPC.
Collapse
Affiliation(s)
- Yiwei Li
- Department of University School of Medicine, Detroit, Mich., USA
| | - Fazlul H. Sarkar
- Department of University School of Medicine, Detroit, Mich., USA
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Mich., USA
| |
Collapse
|
246
|
Shen W, Pang H, Liu J, Zhou J, Zhang F, Liu L, Ma N, Zhang N, Zhang H, Liu L. Epithelial-mesenchymal transition contributes to docetaxel resistance in human non-small cell lung cancer. Oncol Res 2015; 22:47-55. [PMID: 25700358 PMCID: PMC7592784 DOI: 10.3727/096504014x14098532393473] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is an aggressive malignancy with high morbidity and mortality. Chemotherapy has always been the principal treatment measure, but its acquired resistance becomes a critical problem. In the current study, we established a new docetaxel-resistant human non-small lung cancer (NSCLC) cell line A549/Docetaxel. The resistance index (RI) of A549/Docetaxel cells and A549 induced by TGF-β to docetaxel were 8.91 and 11.5, respectively. Compared to the parental A549 cells, the multiplication time of A549/Docetaxel was prolonged, the proportion of the cell cycle in the S phase decreased while that in the G1 phase increased, and apoptotic rate was much lower. The morphology of the resistant cells eventuated epithelial-mesenchymal transition (EMT), which was confirmed by the higher expression of fibronectin, vimentin (mesenchymal markers), and lower expression of E-cadherin (epithelial marker) at mRNA and proteins levels. Furthermore, the representative markers for docetaxel resistance were examined, including ABCB1 (MDR1), Bcl-2, Bax, and tubulin, to figure out the mechanisms of the resistance of A549/Docetaxel. In summary, we have established a typical docetaxel-resistant human NSCLC cell line A549/Docetaxel, and it was suggested that the multidrug resistance of A549/Docetaxel was related to EMT.
Collapse
Affiliation(s)
- Weiwei Shen
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Hailin Pang
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Jiayu Liu
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Jing Zhou
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Feng Zhang
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Lele Liu
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Ningqiang Ma
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Ning Zhang
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Helong Zhang
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| | - Lili Liu
- Department of Oncology, Tangdu Hospital, the Fourth Military Medical UniversityXian, ShaanxiChina
| |
Collapse
|
247
|
Song Z, Liu Z, Sun J, Sun FL, Li CZ, Sun JZ, Xu LY. The MRTF-A/B function as oncogenes in pancreatic cancer. Oncol Rep 2015; 35:127-38. [PMID: 26498848 DOI: 10.3892/or.2015.4329] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/06/2015] [Indexed: 11/05/2022] Open
Abstract
Despite evidence that MRTF-A/B, co-activators of serum response factor (SRF), promotes tumor cell invasion and metastasis in cancer, there are no studies describing MRTF-A/B in pancreatic cancer. To clarify involvement of MRTF-A/B expression in pancreatic cancer, we used quantitative reverse transcription-polymerase chain reaction and western blot analysis to detect MRTF-A/B in pancreatic cancer, intraductal papillary mucinous neoplasm (IPMN) and non-neoplastic pancreata. MRTF-A/B expression differs significantly between cancer and non-neoplastic tissues as well as between non-neoplastic tissues and IPMN bulk tissues. Next, we studied the roles of MRTF-A/B in vitro. Overexpression of MRTF-A/B promoted epithelial-mesenchymal transition (EMT) and generated stem cell-like cells in normal pancreatic cells. We performed quantitative reverse transcription-polymerase chain reaction to detect the level of MRTF-A/B in 19 pancreatic cancer cell lines. We found that their expression was associated with gemcitabine resistance. Like in normal pancreatic cells, MRTF-A/B also promoted EMT and promoted formation of stem cell-like cells in pancreatic cancer and they could regulate microRNA expression associated with EMT and CICs. Finally, to further demonstrate the roles of MRTF-A/B in vivo, we performed nude mouse model of s.c. xenograft and found that overexpression of MRTF-A and MRTF-B promoted pancreatic cancer growth. Elucidating the roles of MRTF-A/B will help us to further understand molecular basis of the disease and offer new gene targets for effective therapies.
Collapse
Affiliation(s)
- Zhao Song
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Zhao Liu
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Jing Sun
- Department of Radiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Feng-Lei Sun
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Chuan-Zhi Li
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Jiu-Zheng Sun
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Li-You Xu
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
248
|
Ramasamy TS, Ayob AZ, Myint HHL, Thiagarajah S, Amini F. Targeting colorectal cancer stem cells using curcumin and curcumin analogues: insights into the mechanism of the therapeutic efficacy. Cancer Cell Int 2015; 15:96. [PMID: 26457069 PMCID: PMC4599442 DOI: 10.1186/s12935-015-0241-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 09/07/2015] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer is one of the commonest cancers in the world and it is also a common cause of cancer-related death worldwide. Despite advanced treatment strategies, the disease is rarely cured completely due to recurrence. Evidence shows that this is due to a small population of cells, called cancer stem cells (CSCs), in the tumour mass that have the self-renewal and differentiation potential to give rise to a new tumour population. Many pre-clinical and clinical studies have used curcumin and its analogues as anti-cancer agents in various types of cancer, including colorectal cancer. Intriguingly, curcumin and its analogues have also recently been shown to be effective in lowering tumour recurrence by targeting the CSC population, hence inhibiting tumour growth. In this review, we highlight the efficacy of curcumin and its analogues in targeting colorectal CSC and also the underlying molecular mechanism involved. Curcumin, in the presence or absence of other anti-cancer agents, has been shown to reduce the size of tumour mass and growth in both in vivo and in vitro studies by affecting many intracellular events that are associated with cancer progression and CSC formation. An insight into the molecular mechanism has unraveled the mode of action via which curcumin could affect the key regulators in CSC, importantly; (1) the signaling pathways, including Wnt/β-catenin, Sonic Hedgehog, Notch and PI3K/Akt/mTOR, (2) microRNA and (3) the epithelial-mesenchymal transition at multiple levels. Therefore, curcumin could play a role as chemosensitiser whereby the colorectal CSCs are now sensitised towards the anti-cancer therapy, therefore, combination therapy using anti-cancer agent with curcumin could be much more effective than treatment using a single cancer agent. This potential treatment modality can be further developed by employing an effective delivery system using a nanotechnology based approach to treat colorectal cancer.
Collapse
Affiliation(s)
- Thamil Selvee Ramasamy
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia ; Cell and Molecular Biology Laboratory, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ain Zubaidah Ayob
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia ; Cell and Molecular Biology Laboratory, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hsu Hsu Lynn Myint
- Faculty of Medicine and Health Science, School of Healthy Aging, Medical Aesthetics and Regenerative Medicine, UCSI University, Kuala Lumpur, Malaysia
| | - Sharmanee Thiagarajah
- Faculty of Medicine and Health Science, School of Healthy Aging, Medical Aesthetics and Regenerative Medicine, UCSI University, Kuala Lumpur, Malaysia
| | - Farahnaz Amini
- Faculty of Medicine and Health Science, School of Healthy Aging, Medical Aesthetics and Regenerative Medicine, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
249
|
Hagiwara K, Gailhouste L, Yasukawa K, Kosaka N, Ochiya T. A robust screening method for dietary agents that activate tumour-suppressor microRNAs. Sci Rep 2015; 5:14697. [PMID: 26423775 PMCID: PMC4589759 DOI: 10.1038/srep14697] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 09/07/2015] [Indexed: 12/17/2022] Open
Abstract
Certain dietary agents, such as natural products, have been reported to show anti-cancer effects. However, the underlying mechanisms of these substances in human cancer remain unclear. We recently found that resveratrol exerts an anti-cancer effect by upregulating tumour-suppressor microRNAs (miRNAs). In the current study, we aimed to identify new dietary products that have the ability to activate tumour-suppressor miRNAs and that therefore may serve as novel tools for the prevention and treatment of human cancers. We describe the generation and use of an original screening system based on a luciferase-based reporter vector for monitoring miR-200c tumour-suppressor activity. By screening a library containing 139 natural substances, three natural compounds — enoxolone, magnolol and palmatine chloride — were identified as being capable of inducing miR-200c expression in breast cancer cells at 10 μM. Moreover, these molecules suppressed the invasiveness of breast cancer cells in vitro. Next, we identified a molecular pathway by which the increased expression of miR-200c induced by natural substances led to ZEB1 inhibition and E-cadherin induction. These results indicate that our method is a valuable tool for a fast identification of natural molecules that exhibit tumour-suppressor activity in human cancer through miRNA activation.
Collapse
Affiliation(s)
- Keitaro Hagiwara
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Department of Biological Sciences, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Luc Gailhouste
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Ken Yasukawa
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Integrative Bioscience and Biomedical Engineering, Graduate School of Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Nobuyoshi Kosaka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
250
|
Cong J, Liu R, Wang X, Jiang H, Zhang Y. MiR-634 decreases cell proliferation and induces apoptosis by targeting mTOR signaling pathway in cervical cancer cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1694-701. [PMID: 26367112 DOI: 10.3109/21691401.2015.1080171] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Taking the emergence of continuous resistance to chemotherapy and the evidence that miRNAs are associated with chemoresistance in cancers into consideration, it is of significant importance to reveal the miRNAs functions for the treatment of cancer. As a novel tumor suppressor, MiR-634 is known to induce apoptosis in tumor cell which is essential for tumorigenesis. Herein, we elucidated the regulation effects of miR-634 in gene expression and discovery of its target gene in cell proliferation and invasion that would aid therapeutic apoptosis. As a result, by targeting mTOR signal pathway, miR-634 inhibited cell proliferation, migration and invasiveness in cervical cancer cells and the block of miR-634 enhances the mTOR expression at both the mRNA and protein levels which regulated the expression of mTOR negatively. Taken together, these results further indicated that miR-634 is an effective target for cancer treatment, and the findings provided in this work might lead to the better understanding of the malignant behavior of cervical carcinoma.
Collapse
Affiliation(s)
- Jianglin Cong
- a Department of Gynaecology , Qilu Hospital, Shandong Univeristy , China .,b Department of Gynaecology , Qingdao University affiliated Yantai Yuhuangding Hospital , Shandong , China , and
| | - Riming Liu
- c Department of Laboratory , Qingdao University affiliated Yantai Yuhuangding Hospital , Shandong , China
| | - Xuan Wang
- b Department of Gynaecology , Qingdao University affiliated Yantai Yuhuangding Hospital , Shandong , China , and
| | - Haiyang Jiang
- b Department of Gynaecology , Qingdao University affiliated Yantai Yuhuangding Hospital , Shandong , China , and
| | - Youzhong Zhang
- a Department of Gynaecology , Qilu Hospital, Shandong Univeristy , China
| |
Collapse
|