1
|
Zhang Y, Wu Y, Liu Z, Yang K, Lin H, Xiong K. Non-coding RNAs as potential targets in metformin therapy for cancer. Cancer Cell Int 2024; 24:333. [PMID: 39354464 PMCID: PMC11445969 DOI: 10.1186/s12935-024-03516-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
Metformin, a widely used oral hypoglycemic drug, has emerged as a potential therapeutic agent for cancer treatment. While initially known for its role in managing diabetes, accumulating evidence suggests that metformin exhibits anticancer properties through various mechanisms. Several cellular or animal experiments have attempted to elucidate the role of non-coding RNA molecules, including microRNAs and long non-coding RNAs, in mediating the anticancer effects of metformin. The present review summarized the current understanding of the mechanisms by which non-coding RNAs modulate the response to metformin in cancer cells. The regulatory roles of non-coding RNAs, particularly miRNAs, in key cellular processes such as cell proliferation, cell death, angiogenesis, metabolism and epigenetics, and how metformin affects these processes are discussed. This review also highlights the role of lncRNAs in cancer types such as lung adenocarcinoma, breast cancer, and renal cancer, and points out the need for further exploration of the mechanisms by which metformin regulates lncRNAs. In addition, the present review explores the potential advantages of metformin-based therapies over direct delivery of ncRNAs, and this review highlights the mechanisms of non-coding RNA regulation when metformin is combined with other therapies. Overall, the present review provides insights into the molecular mechanisms underlying the anticancer effects of metformin mediated by non-coding RNAs, offering novel opportunities for the development of personalized treatment strategies in cancer patients.
Collapse
Affiliation(s)
- Yihan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, Jiangxi, China
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang, China
| | - Yunhao Wu
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang, China
| | - Zixu Liu
- The First School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Kangping Yang
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang, China
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Department of Pathophysiology, School of Basic Medical Sciences, Nanchang, China
| | - Kai Xiong
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, Jiangxi, China.
| |
Collapse
|
2
|
Yu S, Chen C, Chen M, Liang J, Jiang K, Lou B, Lu J, Zhu X, Zhou D. MAGOH promotes gastric cancer progression via hnRNPA1 expression inhibition-mediated RONΔ160/PI3K/AKT signaling pathway activation. J Exp Clin Cancer Res 2024; 43:32. [PMID: 38268030 PMCID: PMC10809607 DOI: 10.1186/s13046-024-02946-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/05/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is associated with high mortality and heterogeneity and poses a great threat to humans. Gene therapies for the receptor tyrosine kinase RON and its spliceosomes are attracting increasing amounts of attention due to their unique characteristics. However, little is known about the mechanism involved in the formation of the RON mRNA alternative spliceosome RONΔ160. METHODS Fourteen human GC tissue samples and six normal gastric tissue samples were subjected to label-free relative quantitative proteomics analysis, and MAGOH was identified as a candidate protein for subsequent studies. The expression of MAGOH in clinical specimens was verified by quantitative real-time PCR and western blotting. We then determined the biological function of MAGOH in GC through in vitro and in vivo experiments. RNA pulldown, RNA sequencing and RNA immunoprecipitation (RIP) were subsequently conducted to uncover the underlying mechanism by which MAGOH regulated the formation of RONΔ160. RESULTS Proteomic analysis revealed that MAGOH, which is located at key nodes and participates in RNA processing and mRNA splicing, was upregulated in GC tissue and GC cell lines and was associated with poor prognosis. Functional analysis showed that MAGOH promoted the proliferation, migration and invasion of GC cells in vitro and in vivo. Mechanistically, MAGOH inhibited the expression of hnRNPA1 and reduced the binding of hnRNPA1 to RON mRNA, thereby promoting the formation of RONΔ160 to activate the PI3K/AKT signaling pathway and consequently facilitating GC progression. CONCLUSIONS Our study revealed that MAGOH could promote the formation of RONΔ160 and activate the PI3K/AKT signaling pathway through the inhibition of hnRNPA1 expression. We elucidate a novel mechanism and potential therapeutic targets for the growth and metastasis of GC based on the MAGOH-RONΔ160 axis, and these findings have important guiding significance and clinical value for the future development of effective therapeutic strategies for GC.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinxiao Liang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kecheng Jiang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Lou
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohua Zhu
- Department of Oncology, Shaoxing People's Hospital, Shaoxing, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Orchard SG, Lockery JE, Broder JC, Ernst ME, Espinoza S, Gibbs P, Wolfe R, Polekhina G, Zoungas S, Loomans-Kropp HA, Woods RL. Association of metformin, aspirin, and cancer incidence with mortality risk in adults with diabetes. JNCI Cancer Spectr 2023; 7:pkad017. [PMID: 36857596 PMCID: PMC10042437 DOI: 10.1093/jncics/pkad017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Metformin and aspirin are commonly co-prescribed to people with diabetes. Metformin may prevent cancer, but in older people (over 70 years), aspirin has been found to increase cancer mortality. This study examined whether metformin reduces cancer mortality and incidence in older people with diabetes; it used randomization to 100 mg aspirin or placebo in the ASPirin in Reducing Events in the Elderly (ASPREE) trial to quantify aspirin's impact on metformin users. METHODS Analysis included community-dwelling ASPREE participants (aged ≥70 years, or ≥65 years for members of US minority populations) with diabetes. Diabetes was defined as a fasting blood glucose level greater than 125 mg/dL, self-report of diabetes, or antidiabetic medication use. Cox proportional hazards regression models were used to analyze the association of metformin and a metformin-aspirin interaction with cancer incidence and mortality, with adjustment for confounders. RESULTS Of 2045 participants with diabetes at enrollment, 965 were concurrently using metformin. Metformin was associated with a reduced cancer incidence risk (adjusted hazard ratio [HR] = 0.68, 95% confidence interval [CI] = 0.51 to 0.90), but no conclusive benefit for cancer mortality (adjusted HR = 0.72, 95% CI = 0.43 to 1.19). Metformin users randomized to aspirin had greater risk of cancer mortality compared with placebo (HR = 2.53, 95% CI = 1.18 to 5.43), but no effect was seen for cancer incidence (HR = 1.11, 95% CI = 0.75 to 1.64). The possible effect modification of aspirin on cancer mortality, however, was not statistically significant (interaction P = .11). CONCLUSIONS In community-dwelling older adults with diabetes, metformin use was associated with reduced cancer incidence. Increased cancer mortality risk in metformin users randomized to aspirin warrants further investigation. ASPREE TRIAL REGISTRATION ClinicalTrials.gov ID NCT01038583.
Collapse
Affiliation(s)
- Suzanne G Orchard
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
| | - Jessica E Lockery
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
- Translational Immunology and Nanotechnology Research Theme, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Department of Internal Medicine, Division of Cancer Prevention and Control, Ohio State University, Columbus, OH, USA
| | - Jonathan C Broder
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy and Department of Family Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Sara Espinoza
- Division of Geriatrics, Gerontology and Palliative Medicine, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, and Geriatrics Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Peter Gibbs
- The Walter & Eliza Hall Institute of Medical Research, Royal Parade, Parkville, Melbourne, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
| | - Galina Polekhina
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
| | - Sophia Zoungas
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
| | - Holli A Loomans-Kropp
- Department of Internal Medicine, Division of Cancer Prevention and Control, Ohio State University, Columbus, OH, USA
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Robyn L Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne,VIC, Australia
| |
Collapse
|
4
|
Xie D, Chen F, Zhang Y, Shi B, Song J, Chaudhari K, Yang SH, Zhang GJ, Sun X, Taylor HS, Li D, Huang Y. Let-7 underlies metformin-induced inhibition of hepatic glucose production. Proc Natl Acad Sci U S A 2022; 119:e2122217119. [PMID: 35344434 PMCID: PMC9169108 DOI: 10.1073/pnas.2122217119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/26/2022] [Indexed: 12/16/2022] Open
Abstract
SignificanceA clear mechanistic understanding of metformin's antidiabetic effects is lacking. This is because suprapharmacological concentrations of metformin have been used in most studies. Using mouse models and human primary hepatocytes, we show that metformin, at clinically relevant doses, suppresses hepatic glucose production by activating a conserved regulatory pathway encompassing let-7, TET3, and a fetal isoform of hepatocyte nuclear factor 4 alpha (HNF4α). We demonstrate that metformin no longer has potent antidiabetic actions in a liver-specific let-7 loss-of-function mouse model and that hepatic delivery of let-7 ameliorates hyperglycemia and improves glucose homeostasis. Our results thus reveal an important role of the hepatic let-7/TET3/HNF4α axis in mediating the therapeutic effects of metformin and suggest that targeting this axis may be a potential therapeutic for diabetes.
Collapse
Affiliation(s)
- Di Xie
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520
| | - Fan Chen
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Yuanyuan Zhang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Bei Shi
- Medical Basic Experimental Teaching Center, China Medical University, Shenyang 110004, China
| | - Jiahui Song
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Kiran Chaudhari
- Department of Pharmacology and Neuroscience, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Shao-Hua Yang
- Department of Pharmacology and Neuroscience, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Gary J. Zhang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Xiaoli Sun
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingqun Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
5
|
Tőkés AM, Vári-Kakas S, Kulka J, Törőcsik B. Tumor Glucose and Fatty Acid Metabolism in the Context of Anthracycline and Taxane-Based (Neo)Adjuvant Chemotherapy in Breast Carcinomas. Front Oncol 2022; 12:850401. [PMID: 35433453 PMCID: PMC9008716 DOI: 10.3389/fonc.2022.850401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is characterized by considerable metabolic diversity. A relatively high percentage of patients diagnosed with breast carcinoma do not respond to standard-of-care treatment, and alteration in metabolic pathways nowadays is considered one of the major mechanisms responsible for therapeutic resistance. Consequently, there is an emerging need to understand how metabolism shapes therapy response, therapy resistance and not ultimately to analyze the metabolic changes occurring after different treatment regimens. The most commonly applied neoadjuvant chemotherapy regimens in breast cancer contain an anthracycline (doxorubicin or epirubicin) in combination or sequentially administered with taxanes (paclitaxel or docetaxel). Despite several efforts, drug resistance is still frequent in many types of breast cancer, decreasing patients’ survival. Understanding how tumor cells rapidly rewire their signaling pathways to persist after neoadjuvant cancer treatment have to be analyzed in detail and in a more complex system to enable scientists to design novel treatment strategies that target different aspects of tumor cells and tumor resistance. Tumor heterogeneity, the rapidly changing environmental context, differences in nutrient use among different cell types, the cooperative or competitive relationships between cells pose additional challenges in profound analyzes of metabolic changes in different breast carcinoma subtypes and treatment protocols. Delineating the contribution of metabolic pathways to tumor differentiation, progression, and resistance to different drugs is also the focus of research. The present review discusses the changes in glucose and fatty acid pathways associated with the most frequently applied chemotherapeutic drugs in breast cancer, as well the underlying molecular mechanisms and corresponding novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Mária Tőkés
- 2nd Department of Pathology, Semmelweis University Budapest, Budapest, Hungary
- *Correspondence: Anna Mária Tőkés,
| | - Stefan Vári-Kakas
- Department of Computers and Information Technology, Faculty of Electrical Engineering and Information Technology, University of Oradea, Oradea, Romania
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University Budapest, Budapest, Hungary
| | - Beáta Törőcsik
- Department of Biochemistry, Semmelweis University Budapest, Budapest, Hungary
| |
Collapse
|
6
|
Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components. Cancers (Basel) 2022; 14:cancers14061462. [PMID: 35326612 PMCID: PMC8945922 DOI: 10.3390/cancers14061462] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chemotherapy resistance is a common occurrence during cancer treatment that cancer researchers are attempting to understand and overcome. Mitochondria are a crucial intracellular signaling core that are becoming important determinants of numerous aspects of cancer genesis and progression, such as metabolic reprogramming, metastatic capability, and chemotherapeutic resistance. Mitophagy, or selective autophagy of mitochondria, can influence both the efficacy of tumor chemotherapy and the degree of drug resistance. Regardless of the fact that mitochondria are well-known for coordinating ATP synthesis from cellular respiration in cellular bioenergetics, little is known its mitophagy regulation in chemoresistance. Recent advancements in mitochondrial research, mitophagy regulatory mechanisms, and their implications for our understanding of chemotherapy resistance are discussed in this review. Abstract Cancer chemotherapy resistance is one of the most critical obstacles in cancer therapy. One of the well-known mechanisms of chemotherapy resistance is the change in the mitochondrial death pathways which occur when cells are under stressful situations, such as chemotherapy. Mitophagy, or mitochondrial selective autophagy, is critical for cell quality control because it can efficiently break down, remove, and recycle defective or damaged mitochondria. As cancer cells use mitophagy to rapidly sweep away damaged mitochondria in order to mediate their own drug resistance, it influences the efficacy of tumor chemotherapy as well as the degree of drug resistance. Yet despite the importance of mitochondria and mitophagy in chemotherapy resistance, little is known about the precise mechanisms involved. As a consequence, identifying potential therapeutic targets by analyzing the signal pathways that govern mitophagy has become a vital research goal. In this paper, we review recent advances in mitochondrial research, mitophagy control mechanisms, and their implications for our understanding of chemotherapy resistance.
Collapse
|
7
|
Metformin and Breast Cancer: Where Are We Now? Int J Mol Sci 2022; 23:ijms23052705. [PMID: 35269852 PMCID: PMC8910543 DOI: 10.3390/ijms23052705] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most prevalent cancer and the leading cause of cancer-related death among women worldwide. Type 2 diabetes–associated metabolic traits such as hyperglycemia, hyperinsulinemia, inflammation, oxidative stress, and obesity are well-known risk factors for breast cancer. The insulin sensitizer metformin, one of the most prescribed oral antidiabetic drugs, has been suggested to function as an antitumoral agent, based on epidemiological and retrospective clinical data as well as preclinical studies showing an antiproliferative effect in cultured breast cancer cells and animal models. These benefits provided a strong rationale to study the effects of metformin in routine clinical care of breast cancer patients. However, the initial enthusiasm was tempered after disappointing results in randomized controlled trials, particularly in the metastatic setting. Here, we revisit the current state of the art of metformin mechanisms of action, critically review past and current metformin-based clinical trials, and briefly discuss future perspectives on how to incorporate metformin into the oncologist’s armamentarium for the prevention and treatment of breast cancer.
Collapse
|
8
|
Uprety B, Abrahamse H. Targeting Breast Cancer and Their Stem Cell Population through AMPK Activation: Novel Insights. Cells 2022; 11:576. [PMID: 35159385 PMCID: PMC8834477 DOI: 10.3390/cells11030576] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Despite some significant advancements, breast cancer has become the most prevalent cancer in the world. One of the main reasons for failure in treatment and metastasis has been attributed to the presence of cancer initiating cells-cancer stem cells. Consequently, research is now being focussed on targeting cancer cells along with their stem cell population. Non-oncology drugs are gaining increasing attention for their potent anticancer activities. Metformin, a drug commonly used to treat type 2 diabetes, is the best example in this regard. It exerts its therapeutic action by activating 5' adenosine monophosphate-activated protein kinase (AMPK). Activated AMPK subsequently phosphorylates and targets several cellular pathways involved in cell growth and proliferation and the maintenance of stem-like properties of cancer stem cells. Therefore, AMPK is emerging as a target of choice for developing effective anticancer drugs. Vanadium compounds are well-known PTP inhibitors and AMPK activators. They find extensive applications in treatment of diabetes and obesity via PTP1B inhibition and AMPK-mediated inhibition of adipogenesis. However, their role in targeting cancer stem cells has not been explored yet. This review is an attempt to establish the applications of insulin mimetic vanadium compounds for the treatment of breast cancer by AMPK activation and PTP1B inhibition pathways.
Collapse
Affiliation(s)
- Bhawna Uprety
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | | |
Collapse
|
9
|
Meerson A, Khatib S, Mahajna J. Natural Products Targeting Cancer Stem Cells for Augmenting Cancer Therapeutics. Int J Mol Sci 2021; 22:ijms222313044. [PMID: 34884848 PMCID: PMC8657727 DOI: 10.3390/ijms222313044] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSC) have been identified in several types of solid tumors. In some cases, CSC may be the source of all the tumor cells, the cause of the tumor's resistance to chemotherapeutic agents, and the source of metastatic cells. Thus, a combination therapy targeting non-CSC tumor cells as well as specifically targeting CSCs holds the potential to be highly effective. Natural products (NPs) have been a historically rich source of biologically active compounds and are known for their ability to influence multiple signaling pathways simultaneously with negligible side effects. In this review, we discuss the potential of NPs in targeting multiple signaling pathways in CSC and their potential to augment the efficacy of standard cancer therapy. Specifically, we focus on the anti-CSC activities of flavonoids, FDA-approved drugs originating from natural sources. Additionally, we emphasize the potential of NPs in targeting microRNA-mediated signaling, given the roles of microRNA in the maintenance of the CSC phenotype.
Collapse
Affiliation(s)
- Ari Meerson
- Department of Natural Products and Nutrition, MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel; (A.M.); (S.K.)
- Faculty of Sciences, Tel Hai Academic College, Qiryat Shemona 12208, Israel
| | - Soliman Khatib
- Department of Natural Products and Nutrition, MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel; (A.M.); (S.K.)
- Faculty of Sciences, Tel Hai Academic College, Qiryat Shemona 12208, Israel
| | - Jamal Mahajna
- Department of Natural Products and Nutrition, MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel; (A.M.); (S.K.)
- Faculty of Sciences, Tel Hai Academic College, Qiryat Shemona 12208, Israel
- Correspondence:
| |
Collapse
|
10
|
Bahmad HF, Daher D, Aljamal AA, Elajami MK, Oh KS, Alvarez Moreno JC, Delgado R, Suarez R, Zaldivar A, Azimi R, Castellano A, Sackstein R, Poppiti RJ. Repurposing of Anticancer Stem Cell Drugs in Brain Tumors. J Histochem Cytochem 2021; 69:749-773. [PMID: 34165342 PMCID: PMC8647630 DOI: 10.1369/00221554211025482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/03/2021] [Indexed: 11/22/2022] Open
Abstract
Brain tumors in adults may be infrequent when compared with other cancer etiologies, but they remain one of the deadliest with bleak survival rates. Current treatment modalities encompass surgical resection, chemotherapy, and radiotherapy. However, increasing resistance rates are being witnessed, and this has been attributed, in part, to cancer stem cells (CSCs). CSCs are a subpopulation of cancer cells that reside within the tumor bulk and have the capacity for self-renewal and can differentiate and proliferate into multiple cell lineages. Studying those CSCs enables an increasing understanding of carcinogenesis, and targeting CSCs may overcome existing treatment resistance. One approach to weaponize new drugs is to target these CSCs through drug repurposing which entails using drugs, which are Food and Drug Administration-approved and safe for one defined disease, for a new indication. This approach serves to save both time and money that would otherwise be spent in designing a totally new therapy. In this review, we will illustrate drug repurposing strategies that have been used in brain tumors and then further elaborate on how these approaches, specifically those that target the resident CSCs, can help take the field of drug repurposing to a new level.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Darine Daher
- Faculty of Medicine, American University of
Beirut, Beirut, Lebanon
| | - Abed A. Aljamal
- Department of Internal Medicine, Mount Sinai
Medical Center, Miami Beach, Florida
| | - Mohamad K. Elajami
- Department of Internal Medicine, Mount Sinai
Medical Center, Miami Beach, Florida
| | - Kei Shing Oh
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Juan Carlos Alvarez Moreno
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Ruben Delgado
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Richard Suarez
- Department of Pathology, Herbert Wertheim
College of Medicine, Florida International University, Miami, Florida
| | - Ana Zaldivar
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Roshanak Azimi
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
| | - Amilcar Castellano
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
- Department of Pathology, Herbert Wertheim
College of Medicine, Florida International University, Miami, Florida
| | - Robert Sackstein
- Department of Translational Medicine,
Translational Glycobiology Institute, Herbert Wertheim College of Medicine,
Florida International University, Miami, Florida
| | - Robert J. Poppiti
- Arkadi M. Rywlin M.D. Department of Pathology
and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach,
Florida
- Department of Pathology, Herbert Wertheim
College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
11
|
Rizvi F, Shaukat L, Azhar A, Jafri A, Aslam U, Imran-ul-Haq H. Preclinical meritorious anticancer effects of Metformin against breast cancer: An In vivo trial. J Taibah Univ Med Sci 2021; 16:504-512. [PMID: 34408607 PMCID: PMC8348326 DOI: 10.1016/j.jtumed.2021.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE This research aims to evaluate the preclinical meritorious and anticancer effects of Metformin in a Xenograft model of breast cancer. METHODS This interventional trial was conducted during a defined period of 5 months (August 2016 January 2017). We used a Xenograft model of nude BALB/c mice. A sample size of 50 mice, allocated into two groups and designated as Group A and Group B for Metformin and negative control groups, respectively. The anticancer activity of Metformin has been evaluated by comparing the tumour volume, tumour weight, tumour regression ratio, percentage regression, and survival rate. RESULTS Compared with the control group, Metformin can significantly reduce the progression of tumour in the Xenograft model of breast cancer induced by MCF-7. This is reflected by significant differences in tumour volume at the final follow-up (p = <0.001). Our findings are further supported by a significant reduction of the tumour growth rate (p = <0.001) and tumour weight (p = <0.001) in the Metformin group than in the control group. Similarly, the total survival rate and tumour regression are more significantly correlated in the Metformin group. CONCLUSION This study demonstrates that Metformin can significantly reduce the tumour growth and can increase the survival rate in a Xenograft model of breast cancer.
Collapse
Affiliation(s)
- Fatima Rizvi
- Dow International Medical College (DUHS), Department of Pharmacology, Karachi, Pakistan
| | - Lubna Shaukat
- DUHS, Department of Oral and Maxillofacial Surgery, Karachi, Pakistan
| | - Arfa Azhar
- AKUH, Department of Biological and Biomedical Sciences, Karachi, Pakistan
| | - Alia Jafri
- Department of Biochemistry Institute, JSMU, Karachi, Pakistan
| | | | | |
Collapse
|
12
|
Precilla DS, Kuduvalli SS, Purushothaman M, Marimuthu P, Ramachandran MA, Anitha TS. Wnt/β-catenin Antagonists: Exploring New Avenues to Trigger Old Drugs in Alleviating Glioblastoma Multiforme. Curr Mol Pharmacol 2021; 15:338-360. [PMID: 33881978 DOI: 10.2174/1874467214666210420115431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/24/2020] [Accepted: 01/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glioblastoma multiforme is one of the most heterogenous primary brain tumor with high mortality. Nevertheless, of the current therapeutic approaches, survival rate remains poor with 12 to 15 months following preliminary diagnosis, this warrants the need for effective treatment modality. Wnt/β-catenin pathway is presumably the most noteworthy pathway up-regulated in almost 80% GBM cases contributing to tumor-initiation, progression and survival. Therefore, therapeutic strategies targeting key components of Wnt/β-catenin cascade using established genotoxic agents like temozolomide and pharmacological inhibitors would be an effective approach to modulate Wnt/β-catenin pathway. Recently, drug repurposing by means of effective combination therapy has gained importance in various solid tumors including GBM, by targeting two or more proteins in a single pathway, thereby possessing the ability to overcome the hurdle implicated by chemo-resistance in GBM. OBJECTIVE In this context, by employing computational tools, an attempt has been carried out to speculate the novel combinations against Wnt/β-catenin signaling pathway. METHODS We have explored the binding interactions of three conventional drugs namely temozolomide, metformin, chloroquine along with three natural compounds viz., epigallocatechin gallate, naringenin and phloroglucinol on the major receptors of Wnt/β-catenin signaling. RESULTS It was noted that all the experimental compounds possessed profound interaction with the two major receptors of Wnt/β-catenin pathway. CONCLUSION To the best of our knowledge, this study is the first of its kind to characterize the combined interactions of the afore-mentioned drugs on Wnt/β-catenin signaling in silico and this will putatively open up new avenues for combination therapies in GBM treatment.
Collapse
Affiliation(s)
- Daisy S Precilla
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Shreyas S Kuduvalli
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | | | - Parthiban Marimuthu
- Structural Bioinformatics Laboratory - Pharmacy, Faculty of Science and Engineering, Åbo Akademi University, Turku. Finland
| | | | | |
Collapse
|
13
|
Chowdhury S, Ghosh S. Cancer Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
14
|
Xu J, Zhou L, Liu Y. Cellular Senescence in Kidney Fibrosis: Pathologic Significance and Therapeutic Strategies. Front Pharmacol 2020; 11:601325. [PMID: 33362554 PMCID: PMC7759549 DOI: 10.3389/fphar.2020.601325] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/21/2020] [Indexed: 01/10/2023] Open
Abstract
Age-related disorders such as chronic kidney disease (CKD) are increasingly prevalent globally and pose unprecedented challenges. In many aspects, CKD can be viewed as a state of accelerated and premature aging. Aging kidney and CKD share many common characteristic features with increased cellular senescence, a conserved program characterized by an irreversible cell cycle arrest with altered transcriptome and secretome. While developmental senescence and acute senescence may positively contribute to the fine-tuning of embryogenesis and injury repair, chronic senescence, when unresolved promptly, plays a crucial role in kidney fibrogenesis and CKD progression. Senescent cells elicit their fibrogenic actions primarily by secreting an assortment of inflammatory and profibrotic factors known as the senescence-associated secretory phenotype (SASP). Increasing evidence indicates that senescent cells could be a promising new target for therapeutic intervention known as senotherapy, which includes depleting senescent cells, modulating SASP and restoration of senescence inhibitors. In this review, we discuss current understanding of the role and mechanism of cellular senescence in kidney fibrosis. We also highlight potential options of targeting senescent cells for the treatment of CKD.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Nunes M, Henriques Abreu M, Bartosch C, Ricardo S. Recycling the Purpose of Old Drugs to Treat Ovarian Cancer. Int J Mol Sci 2020; 21:ijms21207768. [PMID: 33092251 PMCID: PMC7656306 DOI: 10.3390/ijms21207768] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023] Open
Abstract
The main challenge in ovarian cancer treatment is the management of recurrences. Facing this scenario, therapy selection is based on multiple factors to define the best treatment sequence. Target therapies, such as bevacizumab and polymerase (PARP) inhibitors, improved patient survival. However, despite their achievements, ovarian cancer survival remains poor; these therapeutic options are highly costly and can be associated with potential side effects. Recently, it has been shown that the combination of repurposed, conventional, chemotherapeutic drugs could be an alternative, presenting good patient outcomes with few side effects and low costs for healthcare institutions. The main aim of this review is to strengthen the importance of repurposed drugs as therapeutic alternatives, and to propose an in vitro model to assess the therapeutic value. Herein, we compiled the current knowledge on the most promising non-oncological drugs for ovarian cancer treatment, focusing on statins, metformin, bisphosphonates, ivermectin, itraconazole, and ritonavir. We discuss the primary drug use, anticancer mechanisms, and applicability in ovarian cancer. Finally, we propose the use of these therapies to perform drug efficacy tests in ovarian cancer ex vivo cultures. This personalized testing approach could be crucial to validate the existing evidences supporting the use of repurposed drugs for ovarian cancer treatment.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal;
- Porto Comprehensive Cancer Center (PCCC), 4200-162 Porto, Portugal; (M.H.A.); (C.B.)
| | - Miguel Henriques Abreu
- Porto Comprehensive Cancer Center (PCCC), 4200-162 Porto, Portugal; (M.H.A.); (C.B.)
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPOP), 4200-162 Porto, Portugal
| | - Carla Bartosch
- Porto Comprehensive Cancer Center (PCCC), 4200-162 Porto, Portugal; (M.H.A.); (C.B.)
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), 4200-162 Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center—Portuguese Oncology Institute of Porto (CI-IPOP), 4200-162 Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal;
- Porto Comprehensive Cancer Center (PCCC), 4200-162 Porto, Portugal; (M.H.A.); (C.B.)
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal
- Correspondence: ; Tel.: +351-225-570-700
| |
Collapse
|
16
|
Samuel SM, Varghese E, Koklesová L, Líšková A, Kubatka P, Büsselberg D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers (Basel) 2020; 12:E2482. [PMID: 32883003 PMCID: PMC7565921 DOI: 10.3390/cancers12092482] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the leaps and bounds in achieving success in the management and treatment of breast cancers through surgery, chemotherapy, and radiotherapy, breast cancer remains the most frequently occurring cancer in women and the most common cause of cancer-related deaths among women. Systemic therapeutic approaches, such as chemotherapy, although beneficial in treating and curing breast cancer subjects with localized breast tumors, tend to fail in metastatic cases of the disease due to (a) an acquired resistance to the chemotherapeutic drug and (b) the development of intrinsic resistance to therapy. The existence of cancer stem cells (CSCs) plays a crucial role in both acquired and intrinsic chemoresistance. CSCs are less abundant than terminally differentiated cancer cells and confer chemoresistance through a unique altered metabolism and capability to evade the immune response system. Furthermore, CSCs possess active DNA repair systems, transporters that support multidrug resistance (MDR), advanced detoxification processes, and the ability to self-renew and differentiate into tumor progenitor cells, thereby supporting cancer invasion, metastasis, and recurrence/relapse. Hence, current research is focusing on targeting CSCs to overcome resistance and improve the efficacy of the treatment and management of breast cancer. Studies revealed that metformin (1, 1-dimethylbiguanide), a widely used anti-hyperglycemic agent, sensitizes tumor response to various chemotherapeutic drugs. Metformin selectively targets CSCs and improves the hypoxic microenvironment, suppresses the tumor metastasis and inflammation, as well as regulates the metabolic programming, induces apoptosis, and reverses epithelial-mesenchymal transition and MDR. Here, we discuss cancer (breast cancer) and chemoresistance, the molecular mechanisms of chemoresistance in breast cancers, and metformin as a chemo-sensitizing/re-sensitizing agent, with a particular focus on breast CSCs as a critical contributing factor to acquired and intrinsic chemoresistance. The review outlines the prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer/chemo-sensitizing drug in the treatment of breast cancer. It intends to provide a rationale for the use of metformin as a combinatory therapy in a clinical setting.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Lenka Koklesová
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
17
|
MicroRNA-21-Enriched Exosomes as Epigenetic Regulators in Melanomagenesis and Melanoma Progression: The Impact of Western Lifestyle Factors. Cancers (Basel) 2020; 12:cancers12082111. [PMID: 32751207 PMCID: PMC7464294 DOI: 10.3390/cancers12082111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
DNA mutation-induced activation of RAS-BRAF-MEK-ERK signaling associated with intermittent or chronic ultraviolet (UV) irradiation cannot exclusively explain the excessive increase of malignant melanoma (MM) incidence since the 1950s. Malignant conversion of a melanocyte to an MM cell and metastatic MM is associated with a steady increase in microRNA-21 (miR-21). At the epigenetic level, miR-21 inhibits key tumor suppressors of the RAS-BRAF signaling pathway enhancing proliferation and MM progression. Increased MM cell levels of miR-21 either result from endogenous upregulation of melanocytic miR-21 expression or by uptake of miR-21-enriched exogenous exosomes. Based on epidemiological data and translational evidence, this review provides deeper insights into environmentally and metabolically induced exosomal miR-21 trafficking beyond UV-irradiation in melanomagenesis and MM progression. Sources of miR-21-enriched exosomes include UV-irradiated keratinocytes, adipocyte-derived exosomes in obesity, airway epithelium-derived exosomes generated by smoking and pollution, diet-related exosomes and inflammation-induced exosomes, which may synergistically increase the exosomal miR-21 burden of the melanocyte, the transformed MM cell and its tumor environment. Several therapeutic agents that suppress MM cell growth and proliferation attenuate miR-21 expression. These include miR-21 antagonists, metformin, kinase inhibitors, beta-blockers, vitamin D, and plant-derived bioactive compounds, which may represent new options for the prevention and treatment of MM.
Collapse
|
18
|
Malinowski B, Musiała N, Wiciński M. Metformin's Modulatory Effects on miRNAs Function in Cancer Stem Cells-A Systematic Review. Cells 2020; 9:cells9061401. [PMID: 32512882 PMCID: PMC7348732 DOI: 10.3390/cells9061401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) have been reported in various hematopoietic and solid tumors, therefore, are considered to promote cancer progression, metastasis, recurrence and drug resistance. However, regulation of CSCs at the molecular level is not fully understood. microRNAs (miRNAs) have been identified as key regulators of CSCs by modulating their major functions: self-renewal capacity, invasion, migration and proliferation. Various studies suggest that metformin, an anti-diabetic drug, has an anti-tumor activity but its precise mechanism of action has not been understood. The present article was written in accordance to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. We systematically reviewed evidence for metformin’s ability to eradicate CSCs through modulating the expression of miRNAs in various solid tumors. PubMed and MEDLINE were searched from January 1990 to January 2020 for in vitro studies. Two authors independently selected and reviewed articles according to predefined eligibility criteria and assessed risk of bias of included studies. Four papers met the inclusion criteria and presented low risk bias. All of the included studies reported a suppression of CSCs’ major function after metformin dosage. Moreover, it was showed that metformin anti-tumor mechanism of action is based on regulation of miRNAs expression. Metformin inhibited cell survival, clonogenicity, wound-healing capacity, sphere formation and promotes chemosensitivity of tumor cells. Due to the small number of publications, aforementioned evidences are limited but may be consider as background for clinical studies.
Collapse
|
19
|
Brown JR, Chan DK, Shank JJ, Griffith KA, Fan H, Szulawski R, Yang K, Reynolds RK, Johnston C, McLean K, Uppal S, Liu JR, Cabrera L, Taylor SE, Orr BC, Modugno F, Mehta P, Bregenzer M, Mehta G, Shen H, Coffman LG, Buckanovich RJ. Phase II clinical trial of metformin as a cancer stem cell-targeting agent in ovarian cancer. JCI Insight 2020; 5:133247. [PMID: 32369446 PMCID: PMC7308054 DOI: 10.1172/jci.insight.133247] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUNDEpidemiologic studies suggest that metformin has antitumor effects. Laboratory studies indicate metformin impacts cancer stem-like cells (CSCs). As part of a phase II trial, we evaluated the impact of metformin on CSC number and on carcinoma-associated mesenchymal stem cells (CA-MSCs) and clinical outcomes in nondiabetic patients with advanced-stage epithelial ovarian cancer (EOC).METHODSThirty-eight patients with stage IIC (n = 1)/III (n = 25)/IV (n = 12) EOC were treated with either (a) neoadjuvant metformin, debulking surgery, and adjuvant chemotherapy plus metformin or (b) neoadjuvant chemotherapy and metformin, interval debulking surgery, and adjuvant chemotherapy plus metformin. Metformin-treated tumors, compared with historical controls, were evaluated for CSC number and chemotherapy response. Primary endpoints were (a) a 2-fold or greater reduction in aldehyde dehydrogenase-positive (ALDH+) CD133+ CSCs and (b) a relapse-free survival at 18 months of more than 50%.RESULTSMetformin was well tolerated. Median progression-free survival was 18.0 months (95% CI 14.0-21.6) with relapse-free survival at 18 months of 59.3% (95% CI 38.6-70.5). Median overall survival was 57.9 months (95% CI 28.0-not estimable). Tumors treated with metformin had a 2.4-fold decrease in ALDH+CD133+ CSCs and increased sensitivity to cisplatin ex vivo. Furthermore, metformin altered the methylation signature in CA-MSCs, which prevented CA-MSC-driven chemoresistance in vitro.CONCLUSIONTranslational studies confirm an impact of metformin on EOC CSCs and suggest epigenetic change in the tumor stroma may drive the platinum sensitivity ex vivo. Consistent with this, metformin therapy was associated with better-than-expected overall survival, supporting the use of metformin in phase III studies.TRIAL REGISTRATIONClinicalTrials.gov NCT01579812.
Collapse
Affiliation(s)
- Jason R. Brown
- Division of Hematology and Oncology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Daniel K. Chan
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jessica J. Shank
- Department of Obstetrics and Gynecology, Naval Medical Center, San Diego, California, USA
| | - Kent A. Griffith
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Huihui Fan
- Van Andel Institute, Grand Rapids, Michigan, USA
| | - Robert Szulawski
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Kun Yang
- Division of Hematology and Oncology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - R. Kevin Reynolds
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Carolyn Johnston
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Karen McLean
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Shitanshu Uppal
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - J. Rebecca Liu
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Lourdes Cabrera
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Sarah E. Taylor
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Brian C. Orr
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Francesmary Modugno
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Pooja Mehta
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Michael Bregenzer
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Geeta Mehta
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Hui Shen
- Van Andel Institute, Grand Rapids, Michigan, USA
| | - Lan G. Coffman
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Ronald J. Buckanovich
- Magee-Womens Research Institute, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Metformin: Sentinel of the Epigenetic Landscapes That Underlie Cell Fate and Identity. Biomolecules 2020; 10:biom10050780. [PMID: 32443566 PMCID: PMC7277648 DOI: 10.3390/biom10050780] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/08/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
The biguanide metformin is the first drug to be tested as a gerotherapeutic in the clinical trial TAME (Targeting Aging with Metformin). The current consensus is that metformin exerts indirect pleiotropy on core metabolic hallmarks of aging, such as the insulin/insulin-like growth factor 1 and AMP-activated protein kinase/mammalian Target Of Rapamycin signaling pathways, downstream of its primary inhibitory effect on mitochondrial respiratory complex I. Alternatively, but not mutually exclusive, metformin can exert regulatory effects on components of the biologic machinery of aging itself such as chromatin-modifying enzymes. An integrative metabolo-epigenetic outlook supports a new model whereby metformin operates as a guardian of cell identity, capable of retarding cellular aging by preventing the loss of the information-theoretic nature of the epigenome. The ultimate anti-aging mechanism of metformin might involve the global preservation of the epigenome architecture, thereby ensuring cell fate commitment and phenotypic outcomes despite the challenging effects of aging noise. Metformin might therefore inspire the development of new gerotherapeutics capable of preserving the epigenome architecture for cell identity. Such gerotherapeutics should replicate the ability of metformin to halt the erosion of the epigenetic landscape, mitigate the loss of cell fate commitment, delay stochastic/environmental DNA methylation drifts, and alleviate cellular senescence. Yet, it remains a challenge to confirm if regulatory changes in higher-order genomic organizers can connect the capacity of metformin to dynamically regulate the three-dimensional nature of epigenetic landscapes with the 4th dimension, the aging time.
Collapse
|
21
|
Guo J, Li Y, Duan H, Yuan L. Metformin Suppresses the Proliferation and Promotes the Apoptosis of Colon Cancer Cells Through Inhibiting the Expression of Long Noncoding RNA-UCA1. Onco Targets Ther 2020; 13:4169-4181. [PMID: 32523353 PMCID: PMC7234977 DOI: 10.2147/ott.s245091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE LncRNA-UCA1 has been proven to facilitate the proliferation and metastasis of colon cancer. Whether metformin inhibits the progression of colon cancer by suppressing lncRNA-UCA1 remains unknown. In this research, we aimed to explore the role of Metformin playing in pathogenesis of colon cancer. MATERIALS AND METHODS Using qRT-PCR, we measured the expression of five tumor-promoting lncRNAs in SW480 and SW620 colon cancer cells. Then, we conducted Western blotting and immunohistochemistry to evaluate the effects of MET or UCA1 knockdown or the combined MET+ UCA1 knockdown on the activities of the PI3K/AKT and ERK pathways in vitro and in tumor tissues obtained from tumor-bearing nude mice. RESULTS The results from CCK-8 assays showed that MET dose-dependently and time-dependently inhibited the viability of the colon cancer cells in vitro. Flow cytometry revealed that MET promoted the apoptosis of the SW480 and SW620 cells. qRT-PCR showed that lncRNA-UCA1 had the highest expression among the five lncRNAs. Suppressing UCA1 expression by siRNA or shRNA could further enhance the metformin-mediated anticancer effects against colon cancer in vitro and in vivo. Metformin decreased the UCA1 expression and further inhibited the proliferation and promoted the apoptosis of the colon cancer cells, which were associated with inactivation of the PI3K/AKT and ERK signaling pathways in vitro and in the tumor tissues obtained from the mice. CONCLUSION These results indicated that metformin has potential anticancer properties and revealed the anticancer mechanisms of metformin against colon cancer via regulating lncRNA-UCA1.
Collapse
Affiliation(s)
- Jianbo Guo
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - He Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Lu Yuan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
22
|
Das PK, Pillai S, Rakib MA, Khanam JA, Gopalan V, Lam AKY, Islam F. Plasticity of Cancer Stem Cell: Origin and Role in Disease Progression and Therapy Resistance. Stem Cell Rev Rep 2020; 16:397-412. [PMID: 31965409 DOI: 10.1007/s12015-019-09942-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In embryonic development and throughout life, there are some cells can exhibit phenotypic plasticity. Phenotypic plasticity is the ability of cells to differentiate into multiple lineages. In normal development, plasticity is highly regulated whereas cancer cells re-activate this dynamic ability for their own progression. The re-activation of these mechanisms enables cancer cells to acquire a cancer stem cell (CSC) phenotype- a subpopulation of cells with increased ability to survive in a hostile environment and resist therapeutic insults. There are several contributors fuel CSC plasticity in different stages of disease progression such as a complex network of tumour stroma, epidermal microenvironment and different sub-compartments within tumour. These factors play a key role in the transformation of tumour cells from a stable condition to a progressive state. In addition, flexibility in the metabolic state of CSCs helps in disease progression. Moreover, epigenetic changes such as chromatin, DNA methylation could stimulate the phenotypic change of CSCs. Development of resistance to therapy due to highly plastic behaviour of CSCs is a major cause of treatment failure in cancers. However, recent studies explored that plasticity can also expose the weaknesses in CSCs, thereby could be utilized for future therapeutic development. Therefore, in this review, we discuss how cancer cells acquire the plasticity, especially the role of the normal developmental process, tumour microenvironment, and epigenetic changes in the development of plasticity. We further highlight the therapeutic resistance property of CSCs attributed by plasticity. Also, outline some potential therapeutic options against plasticity of CSCs. Graphical Abstract .
Collapse
Affiliation(s)
- Plabon Kumar Das
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Suja Pillai
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, 4029, Australia.
| | - Md Abdur Rakib
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Jahan Ara Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Vinod Gopalan
- School of Medicine, Griffith University Gold Coast Campus, Gold Coast, QLD, 4222, Australia
| | - Alfred K Y Lam
- School of Medicine, Griffith University Gold Coast Campus, Gold Coast, QLD, 4222, Australia
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, 4029, Australia.
| |
Collapse
|
23
|
Sun Y, Lei B, Huang Q. SOX18 Affects Cell Viability, Migration, Invasiveness, and Apoptosis in Hepatocellular Carcinoma (HCC) Cells by Participating in Epithelial-to-Mesenchymal Transition (EMT) Progression and Adenosine Monophosphate Activated Protein Kinase (AMPK)/Mammalian Target of Rapamycin (mTOR). Med Sci Monit 2019; 25:6244-6254. [PMID: 31427562 PMCID: PMC6713035 DOI: 10.12659/msm.915729] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common malignancies around the world. It has been verified that the expression of SOX18 is correlated to poor clinical prognosis in patients with ovarian cancer, non-small cell lung cancer, or breast invasive ductal carcinoma. However, the expression pattern and biological function of SOX18 in HCC tissues remains unclear. In this study, we set out to investigate the associated biological function and potential molecular mechanism of the SOX18 gene in HCC cells. Material/Methods The mRNA and protein expression levels of experimental related genes were detected by real-time polymerase chain reaction and western blotting assay, respectively. The MTT method was used to assess cell viability, and cell apoptosis analysis was performed by means of FACScan flow cytometry. Wound-healing assay and Transwell analysis were performed to evaluate the ability of cell migration and invasiveness, respectively. Results SOX18 was highly expressed in various HCC cell lines. In addition, SOX18 promoted cell viability, migration, and invasion and simultaneously induce cell apoptosis. SOX18 promoted epithelial-to-mesenchymal transition (EMT) progression, and SOX18 downregulation activated the autophagy signaling pathway AMPK/mTOR in HCC cells. Conclusions SOX18 downregulation in HCC cells suppressed cell viability and metastasis, induced cell apoptosis and hindered the occurrence and progression of tumor cells by participating in the EMT process and regulating the autophagy signaling pathway AMPK/mTOR.
Collapse
Affiliation(s)
- Yanni Sun
- Department of Hepatology, Yantai City Hospital for Infectious Diseases, Yantai, Shandong, China (mainland)
| | - Bo Lei
- Department of Hepatology, Yantai City Hospital for Infectious Diseases, Yantai, Shandong, China (mainland)
| | - Qingxian Huang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China (mainland)
| |
Collapse
|
24
|
Metformin causes cancer cell death through downregulation of p53-dependent differentiated embryo chondrocyte 1. J Biomed Sci 2018; 25:81. [PMID: 30442142 PMCID: PMC6238313 DOI: 10.1186/s12929-018-0478-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/18/2018] [Indexed: 12/27/2022] Open
Abstract
Background Metformin is the most commonly used first-line medicine for type II diabetes mellitus. Acting via AMP-activated protein kinase, it has been used for more than 60 years and has an outstanding safety record. Metformin also offers protection against cancer, but its precise mechanisms remain unclear. Methods We first examined the cytotoxic effects of metformin in the HeLa human cervical carcinoma and ZR-75-1 breast cancer cell lines using assays of cell viability, cleaved poly-ADP-ribose polymerase, and Annexin V-fluorescein isothiocyanate apoptosis, as well as flow cytometric analyses of the cell cycle profile and reactive oxygen species (ROS). We later clarified the effect of metformin on p53 protein stability using transient transfection and cycloheximide chase analyses. Results We observed that metformin represses cell cycle progression, thereby inducing subG1 populations, and had induced apoptosis through downregulation of p53 protein and a target gene, differentiated embryo chondrocyte 1 (DEC1). In addition, metformin increased intracellular ROS levels, but N-acetyl cysteine, a ROS scavenger, failed to suppress metformin-induced apoptosis. Further results showed that metformin disrupted the electron transport chain and collapsed the mitochondrial membrane potential, which may be the cause of the elevated ROS levels. Examination of the mechanisms underlying metformin-induced HeLa cell death revealed that reduced stability of p53 in metformin-treated cells leads to decreases in DEC1 and induction of apoptosis. Conclusion The involvement of DEC1 provides new insight into the positive or negative functional roles of p53 in the metformin-induced cytotoxicity in tumor cells.
Collapse
|
25
|
Wu C, Qin N, Ren H, Yang M, Liu S, Wang Q. Metformin Regulating miR-34a Pathway to Inhibit Egr1 in Rat Mesangial Cells Cultured with High Glucose. Int J Endocrinol 2018; 2018:6462793. [PMID: 29681936 PMCID: PMC5841108 DOI: 10.1155/2018/6462793] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 12/10/2017] [Accepted: 01/03/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Activating AMPKα negatively regulates Egr1 to inhibit inflammatory cytokines in high glucose. miR-34a inhibition increases phosphorylated AMPKα through mediating SIRT1 to suppress the development of fatty liver. AIM OF THE STUDY To clarify the function of Egr1 on the inflammation and fibrosis in high glucose-cultured MCs, as well as to explore the effects of metformin on miR-34a pathway and Egr1 expression. METHODS We transfected MCs with miR-34a inhibitor. And MCs were transfected with small interfering RNA for silencing Egr1 and SIRT1. Quantitative real-time PCR was used to assay the transcription levels of Egr1 mRNA and miR-34a. Western blot was used to test the protein. And ELISA was used to measure inflammatory factors. RESULTS High glucose upregulates Egr1 to aggravate the inflammation and fibrosis in MCs. miR-34a suppresses the activation of SIRT1/AMPKα and results in promoting Egr1 in high glucose-cultured MCs. Metformin attenuates high glucose-stimulated inflammation and fibrosis in MCs by regulating miR-34a-mediated SIRT1/AMPKα activity and the downstream Egr1 protein. CONCLUSION We enriched the effects of miR-34a pathway regulating Egr1 in high glucose-cultured MCs. It provides a foundation for future researches considering Egr1 as a therapeutic target and a new direction for the clinical application of metformin in early DKD.
Collapse
Affiliation(s)
- Can Wu
- Department of Endoscope, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Ningning Qin
- Department of Endocrinology, The Second People's Hospital of Fuxin City, Fuxin, Liaoning, China
| | - Huiwen Ren
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Min Yang
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Shuang Liu
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| | - Qiuyue Wang
- Department of Endocrinology, The First Hospital Affiliated to China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Lam TG, Jeong YS, Kim SA, Ahn SG. New metformin derivative HL156A prevents oral cancer progression by inhibiting the insulin-like growth factor/AKT/mammalian target of rapamycin pathways. Cancer Sci 2018; 109:699-709. [PMID: 29285837 PMCID: PMC5834796 DOI: 10.1111/cas.13482] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/14/2017] [Accepted: 12/24/2017] [Indexed: 12/14/2022] Open
Abstract
Metformin is a biguanide widely prescribed as an antidiabetic drug for type 2 diabetes mellitus patients. The purpose of the present study was to observe the effects of the new metformin derivative, HL156A, on human oral cancer cell and to investigate its possible mechanisms. It was observed that HL156A significantly decreased FaDu and YD‐10B cell viability and colony formation in a dose‐dependent way. HL156A also markedly reduced wound closure and migration of FaDu and YD‐10B cells. We observed that HL156A decreased mitochondrial membrane potential and induced reactive oxygen species (ROS) levels and apoptotic cells with caspase‐3 and ‐9 activation. HL156A inhibited the expression and activation of insulin‐like growth factor (IGF)‐1 and its downstream proteins, AKT, mammalian target of rapamycin (mTOR), and ERK1/2. In addition, HL156A activated AMP‐activated protein kinase/nuclear factor kappa B (AMPK‐NF‐κB) signaling of FaDu and YD‐10B cells. A xenograft mouse model further showed that HL156A suppressed AT84 mouse oral tumor growth, accompanied by down‐regulated p‐IGF‐1, p‐mTOR, proliferating cell nuclear antigen (PCNA) and promoted p‐AMPK and TUNEL expression. These results suggest the potential value of the new metformin derivative HL156A as a candidate for a therapeutic modality for the treatment of oral cancer.
Collapse
Affiliation(s)
- Thuy Giang Lam
- Department of Pathology, College of Dentistry, Chosun University, Gwangju, South Korea
| | - Yun Soo Jeong
- Department of Pathology, College of Dentistry, Chosun University, Gwangju, South Korea
| | - Soo-A Kim
- Department of Biochemistry, College of Oriental Medicine, Dongguk University, Gyeongju, South Korea
| | - Sang-Gun Ahn
- Department of Pathology, College of Dentistry, Chosun University, Gwangju, South Korea
| |
Collapse
|
27
|
Smigiel JM, Parameswaran N, Jackson MW. Targeting Pancreatic Cancer Cell Plasticity: The Latest in Therapeutics. Cancers (Basel) 2018; 10:cancers10010014. [PMID: 29320425 PMCID: PMC5789364 DOI: 10.3390/cancers10010014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/27/2017] [Accepted: 01/04/2018] [Indexed: 02/07/2023] Open
Abstract
Mortality remains alarmingly high for patients diagnosed with pancreatic ductal adenocarcinoma (PDAC), with 93% succumbing to the disease within five years. The vast majority of PDAC cases are driven by activating mutations in the proto-oncogene KRAS, which results in constitutive proliferation and survival signaling. As efforts to target RAS and its downstream effectors continue, parallel research aimed at identifying novel targets is also needed in order to improve therapeutic options and efficacy. Recent studies demonstrate that self-renewing cancer stem cells (CSCs) contribute to metastatic dissemination and therapy failure, the causes of mortality from PDAC. Here, we discuss current challenges in PDAC therapeutics, highlight the contribution of mesenchymal/CSC plasticity to PDAC pathogenesis, and propose that targeting the drivers of plasticity will prove beneficial. Increasingly, intrinsic oncogenic and extrinsic pro-growth/survival signaling emanating from the tumor microenvironment (TME) are being implicated in the de novo generation of CSC and regulation of tumor cell plasticity. An improved understanding of key regulators of PDAC plasticity is providing new potential avenues for targeting the properties associated with CSC (including enhanced invasion and migration, metastatic outgrowth, and resistance to therapy). Finally, we describe the growing field of therapeutics directed at cancer stem cells and cancer cell plasticity in order to improve the lives of patients with PDAC.
Collapse
Affiliation(s)
- Jacob M Smigiel
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Neetha Parameswaran
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Mark W Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
28
|
García-Calzón S, Perfilyev A, Männistö V, de Mello VD, Nilsson E, Pihlajamäki J, Ling C. Diabetes medication associates with DNA methylation of metformin transporter genes in the human liver. Clin Epigenetics 2017; 9:102. [PMID: 28947922 PMCID: PMC5609005 DOI: 10.1186/s13148-017-0400-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/04/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Given that metformin is the most common pharmacological therapy for type 2 diabetes, understanding the function of this drug is of great importance. Hepatic metformin transporters are responsible for the pharmacologic action of metformin. However, epigenetics in genes encoding metformin transporters has not been fully elucidated. We examined the DNA methylation of these genes in the liver of subjects with type 2 diabetes and tested whether epigenetic alterations associate with diabetes medication, i.e., metformin or insulin plus metformin treatment. RESULTS DNA methylation in OCT1 encoded by SLC22A1, OCT3 encoded by SLC22A3, and MATE1 encoded by SLC47A1 was assessed in the human liver. Lower average and promoter DNA methylation of SLC22A1, SLC22A3, and SLC47A1 was found in diabetic subjects receiving just metformin, compared to those who took insulin plus metformin or no diabetes medication. Moreover, diabetic subjects receiving just metformin had a similar DNA methylation pattern in these genes compared to non-diabetic subjects. Notably, DNA methylation was also associated with gene expression, glucose levels, and body mass index, i.e., higher SLC22A3 methylation was related to lower SLC22A3 expression and to insulin plus metformin treatment, higher fasting glucose levels and higher body mass index. Importantly, metformin treatment did also directly decrease DNA methylation of SLC22A1 in hepatocytes cultured in vitro. CONCLUSIONS Our study supports that metformin decreases DNA methylation of metformin transporter genes in the human liver. Moreover, higher methylation levels in these genes associate with hyperglycaemia and obesity.
Collapse
Affiliation(s)
- Sonia García-Calzón
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Jan Waldenströms gata 35, CRC 91:12, 205 02 Malmö, Sweden
| | - Alexander Perfilyev
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Jan Waldenströms gata 35, CRC 91:12, 205 02 Malmö, Sweden
| | - Ville Männistö
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Vanessa D de Mello
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 80100 Joensuu, Finland
| | - Emma Nilsson
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Jan Waldenströms gata 35, CRC 91:12, 205 02 Malmö, Sweden
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 80100 Joensuu, Finland.,Clinical Nutrition and Obesity Center, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Charlotte Ling
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Jan Waldenströms gata 35, CRC 91:12, 205 02 Malmö, Sweden
| |
Collapse
|
29
|
Blumrich EM, Dringen R. Metformin Accelerates Glycolytic Lactate Production in Cultured Primary Cerebellar Granule Neurons. Neurochem Res 2017; 44:188-199. [PMID: 28688035 DOI: 10.1007/s11064-017-2346-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 12/13/2022]
Abstract
Metformin is the most frequently used drug for the treatment of type-II diabetes. As metformin has been reported to cross the blood-brain barrier, brain cells will encounter this drug. To test whether metformin may affect the metabolism of neurons, we exposed cultured rat cerebellar granule neurons to metformin. Treatment with metformin caused a time- and concentration-dependent increase in glycolytic lactate release from viable neurons as demonstrated by the three-to fivefold increase in extracellular lactate concentration determined after exposure to metformin. Half-maximal stimulation of lactate production was found after incubation of neurons for 4 h with around 2 mM or for 24 h with around 0.5 mM metformin. Neuronal cell viability was not affected by millimolar concentrations of metformin during acute incubations in the hour range nor during prolonged incubations, although alterations in cell morphology were observed during treatment with 10 mM metformin for days. The acute stimulation of neuronal lactate release by metformin was persistent upon removal of metformin from the medium and was not affected by the presence of modulators of adenosine monophosphate activated kinase activity. In contrast, rabeprazole, an inhibitor of the organic cation transporter 3, completely prevented metformin-mediated stimulation of neuronal lactate production. In summary, the data presented identify metformin as a potent stimulator of glycolytic lactate production in viable cultured neurons and suggest that organic cation transporter 3 mediates the uptake of metformin into neurons.
Collapse
Affiliation(s)
- Eva-Maria Blumrich
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.,Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany. .,Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany.
| |
Collapse
|
30
|
Ye YQ, Zhang L, Xie J, Zhu HY, Xie Y, Zeng B. Role of VEGF in metformin induced apoptosis of HepG2 cells. Shijie Huaren Xiaohua Zazhi 2017; 25:966-973. [DOI: 10.11569/wcjd.v25.i11.966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the molecular mechanism of metformin induced apoptosis of HepG2 cells.
METHODS HepG2 cells were treated with different concentrations (0-20 mmol/L) of metformin (MET) for 24 h or 10 mmol/L MET for different times (0-48 h), and the effect of MET on cell proliferation was measured by MTT assay. Annexin V-FITC/PI flow cytometry was used to determine the apoptosis rate. RT-PCR was used to analyze the expression of vascular endothelial growth factor (VEGF) in HepG2 cells treated with MET.
RESULTS MET had an obvious inhibitory effect on HepG2 cell proliferation. After treatment with 0, 5, 10, 15, and 20 mmol/L MET for 24 h, the relative cell viability rates of HepG2 cells were 100%, 80.56% ± 0.72%, 71.06% ± 0.70%, 64.73% ± 0.35%, and 54.73% ± 0.40%, respectively, showing a dose-dependent manner. After treatment with 10 mmol/L MET for 0, 12, 24, 36, and 48 h, the relative cell viability rates of HepG2 cells were 100%, 83.40% ± 0.70%, 69.86% ± 0.45%, 60.40% ± 0.88%, and 50.70% ± 0.45%, respectively, showing a time-dependent manner. Annexin V-FITC/PI flow cytometry revealed that the apoptosis rates of HepG2 cell were increased after treatment with 0, 5, 10, 15, and 20 mmol/L MET for 24 h, and the apoptosis rates were 2.78% ± 0.68%, 9.33% ± 0.22%, 17.13% ± 0.10%, 21.61% ± 0.20%, and 25.26% ± 1.09%, respectively, showing a dose-dependent manner. The apoptosis rates of HepG2 cells were increased after treatment with 10 mmol/L for 0, 12, 24, 36, 48 h, and the apoptosis rates were 2.05% ± 0.04%, 8.10% ± 0.08%, 16.53% ± 0.93%, 20.95% ± 0.16%, and 25.65% ± 0.44%, showing a time-dependent manner. The expression of VEGF decreased after treatment with different concentrations of MET for 24 h or 10 mmol/L MET for different times, showing a dose- and time-dependent manner.
CONCLUSION MET can inhibit HepG2 cell proliferation via inducing apoptosis, which may involve the expression of VEGF.
Collapse
|
31
|
Sabnis HS, Bradley HL, Tripathi S, Yu WM, Tse W, Qu CK, Bunting KD. Synergistic cell death in FLT3-ITD positive acute myeloid leukemia by combined treatment with metformin and 6-benzylthioinosine. Leuk Res 2016; 50:132-140. [PMID: 27760406 DOI: 10.1016/j.leukres.2016.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/23/2016] [Accepted: 10/04/2016] [Indexed: 02/08/2023]
Abstract
Current therapy for acute myeloid leukemia (AML) primarily includes high-dose cytotoxic chemotherapy with or without allogeneic stem cell transplantation. Targeting unique cellular metabolism of cancer cells is a potentially less toxic approach. Monotherapy with mitochondrial inhibitors like metformin have met with limited success since escape mechanisms such as increased glycolytic ATP production, especially in hyperglycemia, can overcome the metabolic blockade. As an alternative strategy for metformin therapy, we hypothesized that the combination of 6-benzylthioinosine (6-BT), a broad-spectrum metabolic inhibitor, and metformin could block this drug resistance mechanism. Metformin treatment alone resulted in significant suppression of ROS and mitochondrial respiration with increased glycolysis accompanied by modest cytotoxicity (10-25%). In contrast, 6-BT monotherapy resulted in inhibition of glucose uptake, decreased glycolysis, and decreased ATP with minimal changes in ROS and mitochondrial respiration. The combination of 6-BT with metformin resulted in significant cytotoxicity (60-70%) in monocytic AML cell lines and was associated with inhibition of FLT3-ITD activated STAT5 and reduced c-Myc and GLUT-1 expression. Therefore, although the anti-tumor and metabolic effects of metformin have been limited by the metabolic reprogramming within cells, the novel combination of 6-BT and metformin targets this bypass mechanism resulting in reduced glycolysis, STAT5 inhibition, and increased cell death.
Collapse
Affiliation(s)
- Himalee S Sabnis
- Department of Pediatrics, Division of Hem/Onc/BMT, Emory University, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Heath L Bradley
- Department of Pediatrics, Division of Hem/Onc/BMT, Emory University, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Shweta Tripathi
- Department of Pediatrics, Division of Hem/Onc/BMT, Emory University, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Wen-Mei Yu
- Department of Pediatrics, Division of Hem/Onc/BMT, Emory University, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - William Tse
- Department of Medicine, Division of Bone Marrow Transplantation, University of Louisville, Louisville, KY, USA, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Division of Hem/Onc/BMT, Emory University, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kevin D Bunting
- Department of Pediatrics, Division of Hem/Onc/BMT, Emory University, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
32
|
de Barros Machado A, Dos Reis V, Weber S, Jauckus J, Brum IS, von Eye Corleta H, Strowitzki T, Capp E, Germeyer A. Proliferation and metastatic potential of endometrial cancer cells in response to metformin treatment in a high versus normal glucose environment. Oncol Lett 2016; 12:3626-3632. [PMID: 27900046 DOI: 10.3892/ol.2016.5041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/22/2016] [Indexed: 11/06/2022] Open
Abstract
In order to improve our understanding of the potential preventive and therapeutic role of metformin, the present study aimed to investigate the capability of low-dose metformin in the efficient inhibition of cancer development and the reduction of the metastasis of endometrial adenocarcinoma type I and primary endometrial epithelial cells (eEPs), with the drug acting as a treatment in a hyperinsulinemic environment exposed to high and normal glucose conditions. The Ishikawa endometrial adenocarcinoma cell line and primary eEPs were exposed to an environment with high (17 mM) or normal glucose (5 mM) and treated with insulin, low-dose metformin (0.1 mM) or a combined treatment. Metastatic potential was assessed by migration and invasion assays, and relative cell proliferation was determined. Metformin at a low dose potently inhibited the insulin action, decreasing the ability of the endometrial cancer (EC) cell line to migrate and invade in a high and normal glucose environment, and decreasing the migration ability of the primary eEPs. In the EC cell line, the insulin treatment increased the proliferation, without any subsequent reduction of proliferation by the addition of 0.1 mM metformin; however, relative cell proliferation sensitivity to metformin was observed in the range between 1 and 5 mM regardless of the glucose concentration present. Overall, metformin at 0.1 mM is not efficient enough to decrease the proliferation in an EC cell line. However, at this concentration, metformin can inhibit the insulin action in endometrial epithelial cancer cells, demonstrating an anti-metastatic effect in high and normal glucose environments.
Collapse
Affiliation(s)
- Amanda de Barros Machado
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Vania Dos Reis
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Sebastian Weber
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Julia Jauckus
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Ilma Simoni Brum
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Helena von Eye Corleta
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Edison Capp
- Laboratory of Molecular Endocrine Biology and Tumor Biology, Department of Physiology, Institute of Basic Sciences of Health, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Ariane Germeyer
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
33
|
Wang J, Liu X, Jiang Z, Li L, Cui Z, Gao Y, Kong D, Liu X. A novel method to limit breast cancer stem cells in states of quiescence, proliferation or differentiation: Use of gel stress in combination with stem cell growth factors. Oncol Lett 2016; 12:1355-1360. [PMID: 27446437 PMCID: PMC4950051 DOI: 10.3892/ol.2016.4757] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 05/26/2016] [Indexed: 01/02/2023] Open
Abstract
The majority of cancer stem cells exist in the G0, or quiescent phase of the cell cycle. However, the cells can escape quiescence following routine radiotherapy and chemotherapy, resulting in tumor recurrence. Presently, achieving the accurate regulation of cancer stem cell growth in order to study a specific state, including the quiescent (mostly G0 or G1 phase), proliferative (mostly S phase) or differential (mostly G2/M phase) states, can be challenging. This makes the determination of cell cycle state-specific characteristics and analysis of potential intervention treatments difficult, particularly for quiescent cells. Breast cancer stem cells were cultured on a soft or hard agar matrix surface in the presence or absence of stem cell growth factors. Cells could be successfully limited in either the quiescent, proliferative or differentiated states. These findings provide a foundation for further study of the cell cycle in breast cancer stem cells.
Collapse
Affiliation(s)
- Jing Wang
- Department of Tumor Surgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Xiangming Liu
- Department of Esophageal Neoplasms, Cancer Hospital of Tianjin Medical University, Tianjin 300060, P.R. China
| | - Zhongmin Jiang
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin 300450, P.R. China
| | - Lili Li
- Department of Orthopedics, Cancer Hospital of Tianjin Medical University, Tianjin 300060, P.R. China
| | - Zhigang Cui
- Department of Tumor Surgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Yuan Gao
- Department of Tumor Surgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Di Kong
- Department of Tumor Surgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Xiaozhi Liu
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin 300450, P.R. China
| |
Collapse
|
34
|
Sun Y, Tao C, Huang X, He H, Shi H, Zhang Q, Wu H. Metformin induces apoptosis of human hepatocellular carcinoma HepG2 cells by activating an AMPK/p53/miR-23a/FOXA1 pathway. Onco Targets Ther 2016; 9:2845-53. [PMID: 27274280 PMCID: PMC4869652 DOI: 10.2147/ott.s99770] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The antidiabetic drug metformin has been shown to possess antitumor functions in many types of cancers. Although studies have revealed its beneficial effects on the prognosis of hepatocellular carcinoma (HCC), the detailed molecular mechanism underlying this event remains largely unknown. In this work, we showed that miR-23a was significantly induced upon metformin treatment; inhibition of miR-23a abrogated the proapoptotic effect of metformin in HepG2 cells. We next established forkhead box protein A1 (FOXA1) as the functional target of miR-23a, and silencing FOXA1 mimicked the effect of metformin. Moreover, the phosphorylation of AMP-activated protein kinase (AMPK) and the expression of p53 were increased upon metformin treatment, and the inhibition of p53 abrogated the induction of miR-23a by metformin, suggesting that AMPK/p53 signaling axis is responsible for the induction of miR-23a by metformin. In summary, we unraveled a novel AMPK/p53/miR-23a/FOXA1 axis in the regulation of apoptosis in HCC, and the application of metformin could, therefore, be effective in the treatment of HCC.
Collapse
Affiliation(s)
- Yunpeng Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chonglin Tao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiaming Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Han He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hongqi Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qiyu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Huanhuan Wu
- Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
35
|
Shen Y, Miao N, Xu J, Gan X, Xu D, Zhou L, Xue H, Zhang W, Lu L. Metformin Prevents Renal Fibrosis in Mice with Unilateral Ureteral Obstruction and Inhibits Ang II-Induced ECM Production in Renal Fibroblasts. Int J Mol Sci 2016; 17:ijms17020146. [PMID: 26805826 PMCID: PMC4783880 DOI: 10.3390/ijms17020146] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 01/13/2023] Open
Abstract
Renal fibrosis is the final common pathway of chronic kidney disease (CKD), and no effective medication is available clinically for managing its progression. Metformin was initially developed as an anti-diabetic drug and recently gained attention for its potential in the treatment of other diseases. In this study, we investigated its effects on renal fibrosis in a mouse model of unilateral ureteral obstruction (UUO) in vivo and in angiotensin II (Ang II)–treated renal fibroblast NRK-49F cells in vitro. Our data showed that UUO induced renal fibrosis and combined with the activation of ERK signaling, the upregulation of fibronectin, collagen I, and transforming growth factor-β (TGF-β). The administration of metformin inhibited the activation of ERK signaling and attenuated the production of extracellular matrix (ECM) proteins and collagen deposition in the obstructed kidneys. In cultured renal fibroblasts, Ang II increased the expression of fibronectin and collagen I and also activated ERK signaling and TGF-β in a time-dependent manner. Pretreatment of the cells with metformin blocked Ang II–induced ERK signaling activation and ECM overproduction. Our results show that metformin prevents renal fibrosis, possibly through the inhibition of ERK signaling, and may be a novel strategy for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Yang Shen
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Naijun Miao
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Jinlan Xu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Xinxin Gan
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Dan Xu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Li Zhou
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Hong Xue
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Wei Zhang
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Limin Lu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
36
|
Sarkar FH. Novel Holistic Approaches for Overcoming Therapy Resistance in Pancreatic and Colon Cancers. Med Princ Pract 2016; 25 Suppl 2:3-10. [PMID: 26228733 PMCID: PMC5588517 DOI: 10.1159/000435814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/08/2015] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal (GI) cancers, such as of the colon and pancreas, are highly resistant to both standard and targeted therapeutics. Therapy-resistant and heterogeneous GI cancers harbor highly complex signaling networks (the resistome) that resist apoptotic programming. Commonly used gemcitabine or platinum-based regimens fail to induce meaningful (i.e. disease-reversing) perturbations in the resistome, resulting in high rates of treatment failure. The GI cancer resistance networks are, in part, due to interactions between parallel signaling and aberrantly expressed microRNAs (miRNAs) that collectively promote the development and survival of drug-resistant cancer stem cells with epithelial-to-mesenchymal transition (EMT) characteristics. The lack of understanding of the resistance networks associated with this subpopulation of cells as well as reductionist, single protein-/pathway-targeted approaches have made 'effective drug design' a difficult task. We propose that the successful design of novel therapeutic regimens to target drug-resistant GI tumors is only possible if network-based drug avenues and agents, in particular 'natural agents' with no known toxicity, are correctly identified. Natural agents (dietary agents or their synthetic derivatives) can individually alter miRNA profiles, suppress EMT pathways and eliminate cancer stem-like cells that derive from pancreatic cancer and colon cancer, by partially targeting multiple yet meaningful networks within the GI cancer resistome. However, the efficacy of these agents as combinations (e.g. consumed in the diet) against this resistome has never been studied. This short review article provides an overview of the different challenges involved in the understanding of the GI resistome, and how novel computational biology can help in the design of effective therapies to overcome resistance.
Collapse
Affiliation(s)
- Fazlul H. Sarkar
- *Fazlul H. Sarkar, PhD, Departments of Pathology and Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, 740 HWCRC, Detroit, MI 48201 (USA), E-Mail
| |
Collapse
|
37
|
Abstract
Metformin is the most commonly prescribed drug for type 2 diabetes mellitus as it is inexpensive, safe, and efficient in ameliorating hyperglycemia and hyperinsulinemia. Epidemiological and clinical observation studies have shown that metformin use reduces risk of cancer in patients with type 2 diabetes mellitus and improves prognosis and survival rate of the cancer patients. Furthermore, ongoing clinical trials of metformin in cancer therapy are extending to nondiabetic population. Thus, metformin is emerging as an attractive candidate for cancer prevention and treatment. This review summarizes recent progress in studies of metformin use in cancer and discusses the possibility to enhance its efficacy and to prevent cancer metastasis.
Collapse
|
38
|
Denise C, Paoli P, Calvani M, Taddei ML, Giannoni E, Kopetz S, Kazmi SMA, Pia MM, Pettazzoni P, Sacco E, Caselli A, Vanoni M, Landriscina M, Cirri P, Chiarugi P. 5-fluorouracil resistant colon cancer cells are addicted to OXPHOS to survive and enhance stem-like traits. Oncotarget 2015; 6:41706-21. [PMID: 26527315 PMCID: PMC4747183 DOI: 10.18632/oncotarget.5991] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/09/2015] [Indexed: 12/24/2022] Open
Abstract
Despite marked tumor shrinkage after 5-FU treatment, the frequency of colon cancer relapse indicates that a fraction of tumor cells survives treatment causing tumor recurrence. The majority of cancer cells divert metabolites into anabolic pathways through Warburg behavior giving an advantage in terms of tumor growth. Here, we report that treatment of colon cancer cell with 5-FU selects for cells with mesenchymal stem-like properties that undergo a metabolic reprogramming resulting in addiction to OXPHOS to meet energy demands. 5-FU treatment-resistant cells show a de novo expression of pyruvate kinase M1 (PKM1) and repression of PKM2, correlating with repression of the pentose phosphate pathway, decrease in NADPH level and in antioxidant defenses, promoting PKM2 oxidation and acquisition of stem-like phenotype. Response to 5-FU in a xenotransplantation model of human colon cancer confirms activation of mitochondrial function. Combined treatment with 5-FU and a pharmacological inhibitor of OXPHOS abolished the spherogenic potential of colon cancer cells and diminished the expression of stem-like markers. These findings suggest that inhibition of OXPHOS in combination with 5-FU is a rational combination strategy to achieve durable treatment response in colon cancer.
Collapse
Affiliation(s)
- Corti Denise
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Maura Calvani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Scott Kopetz
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Syed Mohammad Ali Kazmi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Morelli Maria Pia
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Piergiorgio Pettazzoni
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elena Sacco
- SYSBIO Centre for Systems Biology, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Anna Caselli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Marco Vanoni
- SYSBIO Centre for Systems Biology, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Matteo Landriscina
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
39
|
Geng M, Xu H, Ren R, Qu Q, Shangguan C, Wu J, Jiang J, Li H, Cao W. Prognostic value of clinicopathological characteristics in patients with pancreatic cancer. Chin J Cancer Res 2015; 27:509-15. [PMID: 26543338 DOI: 10.3978/j.issn.1000-9604.2015.06.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The purpose of this study was to analyze the effects of all clinical characteristics on the overall survival time, in order to provide a basis for determining the prognostic factor of patients with pancreatic cancer. METHODS A total of 103 pancreatic cancer patients were admitted to the Department of Radiotherapy and Chemotherapy of the Ruijin Hospital, Shanghai Jiaotong University School of Medicine, between January 2002 and December 2012. There were 68 men and 35 women; the median age was 62 years. Diagnoses of pancreatic cancer in all patients were confirmed by histopathology, cytology, or clinical diagnosis. The Kaplan-Meier method was performed to calculate the overall survival rate. The log-rank method was used to examine the univariate analysis. The Cox regression model was performed for multivariate analysis. RESULTS The median survival time was 293 days, the 1-, 2-, and 3-year survival rates were 27.18%, 5.83%, and 1.94%, respectively. Cox regression analysis revealed that age (P=0.015), Karnofsky performance status (PS) (P=0.002), surgical types (P<0.001), and platelet counts (P<0.001) were independent prognostic factors affecting the overall survival of patients with pancreatic cancer. CONCLUSIONS Pancreatic cancer had a poor prognosis, the general physical condition, age, the availability of radical surgery, and platelet counts were factors influencing the overall survival of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Mei Geng
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Haoping Xu
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Ruobing Ren
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Qing Qu
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Chengfang Shangguan
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Junwei Wu
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Jinsong Jiang
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Hao Li
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Weiguo Cao
- 1 Department of Oncology, 2 Department of Radiation Oncology, Ruijin Hosptial, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
40
|
Tian RH, Zhang YG, Wu Z, Liu X, Yang JW, Ji HL. Effects of metformin on survival outcomes of lung cancer patients with type 2 diabetes mellitus: a meta-analysis. Clin Transl Oncol 2015; 18:641-9. [PMID: 26459254 DOI: 10.1007/s12094-015-1412-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/18/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Anti-cancer effect of metformin on different kinds of lung cancer has been studied frequently. However, the association between metformin and the prognosis of lung cancer in type 2 diabetes patients is still controversial. METHODS An electronic search was conducted using PubMed/Medicine, EMBASE and Cochrane library databases. Statistical analyses were carried out using either random-effects or fixed-effects models according to the heterogeneity examined by I (2) statistics. RESULTS Six studies involving 2350 patients were included in the current meta-analysis. In all, the pooled HR of overall survival (OS) was 0.90 (95 % CI 0.84-0.96; P = 0.003). Sub-group analysis showed that when stratified by region the HR of OS was 0.52 (95 % CI 0.31-0.87; P = 0.012) and 0.86 (95 % CI 0.67-1.11, P = 0.361) for Asian and Western countries. When stratified by study design, the HR of OS was 0.78 (95 % CI 0.52-1.15, P = 0.206) and 0.82 (95 % CI 0.59-1.16, P = 0.264) for cohort and medical data-based studies. When stratified by lung cancer subtype, HR of OS was 0.52 (95 % CI 0.31-0.87; P = 0.012), 1.06 (95 % CI 0.51-2.19; P = 0.878) and 0.82 (95 % CI 0.59-1.16; P = 0.264) for SCLS, NSCLC and non-divided subtypes, respectively. CONCLUSION Metformin use may associate with a good prognosis in lung cancer patients with type 2 diabetes but the effect was modest. However, it could achieve benefits in a selective sub-group of lung cancer patients especially in SCLC patients from Asian. Further studies are warranted to confirm this efficacy.
Collapse
Affiliation(s)
- R-H Tian
- Department of Respiratory, Affiliated Haian People's Hospital of Nantong University, 17 Cheng Zhong Ba Middle Road, Haian, 226600, Jiangsu, People's Republic of China
| | - Y-G Zhang
- Department of Respiratory, Shouguang People's Hospital, Shouguang, 262700, Shandong, People's Republic of China
| | - Z Wu
- Department of Respiratory, Affiliated Haian People's Hospital of Nantong University, 17 Cheng Zhong Ba Middle Road, Haian, 226600, Jiangsu, People's Republic of China.
| | - X Liu
- Department of Respiratory, Affiliated Haian People's Hospital of Nantong University, 17 Cheng Zhong Ba Middle Road, Haian, 226600, Jiangsu, People's Republic of China.
| | - J-W Yang
- Department of Respiratory, Affiliated Haian People's Hospital of Nantong University, 17 Cheng Zhong Ba Middle Road, Haian, 226600, Jiangsu, People's Republic of China
| | - H-L Ji
- Department of Medical Affairs, Affiliated Haian People's Hospital of Nantong University, Haian, 226600, Jiangsu, People's Republic of China
| |
Collapse
|
41
|
Metformin inhibits the proliferation, metastasis, and cancer stem-like sphere formation in osteosarcoma MG63 cells in vitro. Tumour Biol 2015; 36:9873-83. [PMID: 26164004 DOI: 10.1007/s13277-015-3751-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 07/02/2015] [Indexed: 01/08/2023] Open
Abstract
Metformin is an oral drug that has been widely used to treat type 2 diabetes mellitus. Interestingly, accumulated evidence indicate that metformin may reduce the risk of cancer in patients with type 2 diabetes and inhibit tumor cell growth and survival in numerous malignancies, including osteosarcoma (OS) cells. In the present study, we aimed to investigate the effects of metformin on the proliferation, migration, invasion, and sphere formation in OS MG63 cells in vitro. Metformin suppressed OS MG63 cell proliferation in a dose- and time-dependent manner and markedly blocked anti-metastatic potentials, migration, and invasion, by downregulating matrix metalloproteinase 2 (MMP2) and MMP9. Besides, we established OS cancer stem-like cell (CSC) model with sarcosphere formation assay and demonstrated that metformin posed damage on CSCs in OS by inhibiting sphere formation and by inducing their stemness loss. The stemness of CSCs in OS such as self-renewal and differentiation potentials was both impaired with a significant decrease of Oct-4 and Nanog activation. Consistent with this, the positive rates of CD90, CD133, and stage-specific embryonic antigen-4 (SSEA-4) were all observed with reductions in response to metformin exposure. In addition, Western blot showed that metformin activated AMPKα at Tyr172, followed by a downregulated phosphorylation of mammalian target of rapamycin (mTOR)/S6 and feedback activation of p-AKT Ser(473) in both OS MG63 cells and CSCs. This indicates that AMPK/mTOR/S6 signaling pathway might be involved in the growth inhibition of both OS MG63 cells and CSCs. These results suggest that metformin, a potential anti-neoplastic agent, might make it a novel therapeutic choice for the treatment of OS in the future.
Collapse
|
42
|
Zhang L, Tong X, Li J, Huang Y, Hu X, Chen Y, Huang J, Wang J, Liu B. Apoptotic and autophagic pathways with relevant small-molecule compounds, in cancer stem cells. Cell Prolif 2015; 48:385-97. [PMID: 26013704 DOI: 10.1111/cpr.12191] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/24/2015] [Indexed: 02/05/2023] Open
Abstract
Accumulating evidence demonstrates existence of cancer stem cells (CSCs), which are suspected of contributing to cancer cell self-renewal capacity and resistance to radiation and/or chemotherapy. Including evasion of apoptosis and autophagic cell death, CSCs have revealed abilities to resist cell death, making them appealing targets for cancer therapy. Recently, molecular mechanisms of apoptosis and of autophagy in CSCs have been gradually explored, comparing them in stem cells and in cancer cells; distinct expression of these systems in CSCs may elucidate how these cells exert their capacity of unlimited self-renewal and hierarchical differentiation. Due to their proposed ability to drive tumour initiation and progression, CSCs may be considered to be potentially useful pharmacological targets. Further, multiple compounds have been verified as triggering apoptosis and/or autophagy, suppressing tumour growth, thus providing new strategies for cancer therapy. In this review, we summarized regulation of apoptosis and autophagy in CSCs to elucidate how key proteins participate in control of survival and death; in addition, currently well-studied compounds that target CSC apoptosis and autophagy are selectively presented. With increasing attention to CSCs in cancer therapy, researchers are now trying to find responses to unsolved questions as unambiguous as possible, which may provide novel insight into future anti-cancer regimes.
Collapse
Affiliation(s)
- Lan Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xupeng Tong
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jingjing Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyue Hu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jinhui Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bo Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
43
|
Li SC, Khan M, Caplin M, Meyer T, Öberg K, Giandomenico V. Somatostatin Analogs Treated Small Intestinal Neuroendocrine Tumor Patients Circulating MicroRNAs. PLoS One 2015; 10:e0125553. [PMID: 25942502 PMCID: PMC4420277 DOI: 10.1371/journal.pone.0125553] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/24/2015] [Indexed: 12/17/2022] Open
Abstract
We previously detected and investigated nine altered microRNAs in small intestinal neuroendocrine tumor (SI-NET) tissues at different stages of disease. The aims of this study are to: 1) analyze whether SI-NET tissue microRNAs can be also detected in patient serum samples, 2) investigate a potential somatostatin analogs (SSAs) role on microRNA levels regulation in SSA-treated patient samples and 3) elucidate whether the serum microRNA levels in samples collected in different hospitals are predictable and steady. Our results show that tissue microRNAs are detectable in patient serum samples, and miR-96, -182, -183, -196a and -200a levels are lower in SI-NET untreated patients than in SSA-treated patients at all different stages. Conversely, miR-31, -129-5p, -133a and -215 levels do not show any difference in untreated SI-NET patients and SSA-treated patients at all different stages. Our findings also show that miR-200a exhibits an atypical behavior with high levels in both untreated and SSA-treated patients at liver metastasis stage, and unequivocally never at the earlier stages. Serum samples collected in two hospitals keep alike microRNA level pattern, elucidating that the results are not dependent on samples handling. In conclusion, SI-NET tissue microRNAs are always detectable in untreated and SSA-treated patient serum samples, SSAs play an unknown role in eliciting SSA-treated patients’ microRNA levels higher than in untreated patients, and this study enlightens that miR-200a might be involved in the liver metastasis during SI-NET progression.
Collapse
Affiliation(s)
- Su-Chen Li
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mohid Khan
- Neuroendocrine Tumor Unit, Centre for Gastroenterology, Royal Free Hospital, London, United Kingdom; The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Martyn Caplin
- Neuroendocrine Tumor Unit, Centre for Gastroenterology, Royal Free Hospital, London, United Kingdom
| | - Tim Meyer
- Department of Oncology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Kjell Öberg
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Endocrine Oncology Clinic, Uppsala University Hospital, Science for Life Laboratory, Uppsala, Sweden
| | - Valeria Giandomenico
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Endocrine Oncology Clinic, Uppsala University Hospital, Science for Life Laboratory, Uppsala, Sweden
| |
Collapse
|
44
|
Zhang G, An H, Fang X. MicroRNA-144 regulates proliferation, invasion, and apoptosis of cells in malignant solitary pulmonary nodule via zinc finger E-box-binding homeobox 1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5960-7. [PMID: 26191328 PMCID: PMC4503199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
AIMS This study was to investigate the expression of microRNA (miR)-144 in malignant solitary pulmonary nodule (SPN) tissues and peripheral blood, as well as the biological function of miR-144 in the occurrence and development of lung cancer. METHODS In this study, 39 malignant and 30 benign SPN patients were included. The expression of miR-144 was examined using quantitative real-time polymerase chain reaction. Receiver operating characteristic (ROC) curve was used to identify the clinical value of miR-144 in the early diagnosis of malignant SPN. MTT assay was performed to determine A549 cell proliferation and Transwell assay was used to detect changes in A549 cell invasion and migration ability. Flow cytometry was performed to monitor cell apoptosis, while Western blotting assay was used to measure protein expression levels. At last, dual-luciferase reporter assay was employed to test whether miR-144 regulates zinc finger E-box-binding homeobox 1 (ZEB1) gene expression. RESULTS Expression of miR-144 was reduced in patients with malignant SPN. miR-144 had diagnostic value for malignant SPN. Proliferation of A549 cells was inhibited by miR-144. Invasion ability of A549 cells was reduced by miR-144. Apoptosis of A549 cells was promoted by miR-144. miR-144 induced A549 cell apoptosis by targeting ZEB1 protein. miR-144 regulated the expression of ZEB1 by interacting with its 3'-UTR region. CONCLUSIONS Expression of miR-144 is reduced in malignant SPN tissues and peripheral blood, being of clinical value in the diagnosis of malignant SPN. miR-144 promotes the apoptosis of lung cancer cells, and inhibits the proliferation, invasion and migration of lung cancer by regulating ZEB1 gene.
Collapse
Affiliation(s)
- Guizhi Zhang
- Department of Respiratory Medicine, General Hospital of Chinese People’s Liberation ArmyBeijing 100853, P. R. China
| | - Huaijie An
- Center of Basic Medical Sciences, Navy General Hospital of People’s Liberation ArmyBeijing 100048, P. R. China
| | - Xiangqun Fang
- Department of Respiratory Medicine, General Hospital of Chinese People’s Liberation ArmyBeijing 100853, P. R. China
| |
Collapse
|
45
|
Ali S, Suresh R, Banerjee S, Bao B, Xu Z, Wilson J, Philip PA, Apte M, Sarkar FH. Contribution of microRNAs in understanding the pancreatic tumor microenvironment involving cancer associated stellate and fibroblast cells. Am J Cancer Res 2015; 5:1251-1264. [PMID: 26046003 PMCID: PMC4449452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/15/2015] [Indexed: 06/04/2023] Open
Abstract
Understanding of molecular events associated with tumor microenvironment in pancreatic cancer (PC) is an active area of research especially because of the rich desmoplasia seen in human PC. Desmoplasia is contributed by several cell types including cancer-associated fibroblast (CAF) and stellate cells (PSCs), which are believed to play critical roles in conferring aggressiveness to PC. The aberrant expression of microRNAs (miRNAs) in PSCs and CAF cells appears to play a pivotal role in the development and progression of PC. In this study, expression analysis of miR-21/miR-221 in conditioned media derived from PSCs/CAF cells, and from PSCs/CAF cells showed up-regulation of both miRNAs compared to MIAPaCa-2 PC cells. In addition, miR-21 expression in stellate cells derived from normal pancreas was substantially lower when compared to PSCs or CAF cells. COLO-357 PC cells cultured in the presence of conditioned media derived from PSC/CAF cells led to a significant increase in clonogenicity and pancreatosphere formation. Furthermore, inhibition of miR-21 with antisense oligonucleotide (ASO) transfection resulted in decreased migration/invasive capacity of PSCs. Similarly, the effect of ASO-miR-221 transfection in CAF cells reduced the expression of NF-κB and K-Ras (target of miR-221) along with inhibition of migration/invasion. Moreover, miRNA expression profiling of PSCs, MIAPaCa-2, and COLO-357 cells, and further validation by real-time PCR, showed several differentially expressed miRNAs, among which four was significantly up-regulated. Collectively, these results suggest a crosstalk between PSCs/CAF cells and PC cells, resulting in the up-regulation of miR-21/miR-221 expression which in part may confer aggressiveness to PC. We conclude that targeting these miRNAs could be useful for developing precision medicine for the prevention of tumor progression and/or for the treatment of PC.
Collapse
Affiliation(s)
- Shadan Ali
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
| | - Raagini Suresh
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
| | - Sanjeev Banerjee
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
| | - Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
| | - Zhihong Xu
- Pancreatic Research Group, University of New South Wales, and Ingham Institute for Applied Medical ResearchSydney, Australia
| | - Jeremy Wilson
- Pancreatic Research Group, University of New South Wales, and Ingham Institute for Applied Medical ResearchSydney, Australia
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
| | - Minoti Apte
- Pancreatic Research Group, University of New South Wales, and Ingham Institute for Applied Medical ResearchSydney, Australia
| | - Fazlul H Sarkar
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan
| |
Collapse
|