1
|
Zhang R, Yin H, Yang M, Liu J, Zhen D, Zhang Z. Advanced progress of the relationship between renin-angiotensin-aldosterone system inhibitors and cancers. J Hypertens 2024; 42:1862-1873. [PMID: 39248142 DOI: 10.1097/hjh.0000000000003836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024]
Abstract
Hypertension and cancers are the most common causes of death in humans, as well as common co-diseases among elderly population. Studies have shown that hypertension is associated with carcinogenesis. The renin-angiotensin-aldosterone system (RAAS) is a crucial regulatory system of blood pressure, fluid, and electrolyte homeostasis, which plays an essential role in the pathogenesis of hypertension, whose mechanism is relatively clear. Studies have indicated that RAAS also widely exists in cancer tissues of different systems, which can affect the risk of cancers by stimulating cancer angiogenesis, participating in cancer-related oxidative stress, and regulating cancer-related immunity. Therefore, inhibiting RAAS activity seems beneficial to decreasing the risk of cancers. As one of the most commonly used antihypertensive drugs, RAAS inhibitors have been widely used in clinical practice. However, the conclusions of clinical studies on the relationship between RAAS inhibitors and cancers are not entirely consistent, which has been widely concerned by clinicians. The latest findings suggest that while RAAS inhibitors may reduce the risk of digestive cancers, respiratory cancers, urological cancers, gynecological cancers, and skin cancers, ACEIs may increase the risk of lung cancer, endometrial cancer, basal cell carcinoma, and squamous cell carcinoma. This article comprehensively reviews animal experiments, clinical studies, and meta-analyses on the relationship between RAAS inhibitors and cancers, to provide references for related studies in the future.
Collapse
Affiliation(s)
- Ruixing Zhang
- The First Clinical Medical College, Lanzhou University
- Department of Heart Center
| | - Hongtao Yin
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | - Mengdi Yang
- The First Clinical Medical College, Lanzhou University
| | - Jinjin Liu
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | - Donghu Zhen
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | | |
Collapse
|
2
|
Muambo KE, Im H, Macha FJ, Oh JE. Reproductive toxicity and molecular responses induced by telmisartan in Daphnia magna at environmentally relevant concentrations. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124525. [PMID: 39004206 DOI: 10.1016/j.envpol.2024.124525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
With aging population increasing globally, the use of pharmaceutically active compounds is rising. The cardiovascular drug telmisartan has been widely detected in various environmental compartments, including biota, surface waters, and sewage treatment plant effluents at concentrations ranging from ng/L to μg/L levels. This study evaluated the effects of telmisartan on the microcrustacean Daphnia magna at a wide range of concentrations (0.35, 0.70, 1.40, 500, and 1000 μg/L) and revealed significant ecotoxicological implications of this drug, even at environmentally relevant concentration. Acute exposure to telmisartan (1.40, 500, and 1000 μg/L) resulted in a notable decrease in heart rate, while chronic exposure accelerated the time to the first brood by 3 days and reduced neonate body size. Molecular investigations revealed marked downregulation of vitellogenin genes (Vtg1 and Vtg2). Non-monotonic dose responses were observed for gene expression, early-stage body length, and the total number of offspring produced, while the heart rate and time to the first brood showed clear concentration-dependent responses. These findings highlight the potential risks, notably to reproductive capacity, associated with exposure to telmisartan in environmentally relevant concentration, suggesting the need for further studies on the potential long-term ecological consequences.
Collapse
Affiliation(s)
- Kimberly Etombi Muambo
- Department of Civil and Environmental Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyungjoon Im
- Institute for Environment and Energy, Pusan National University, Busan, 46241, Republic of Korea
| | - Fulgence Jacob Macha
- Biocolloids and Surfaces Laboratory, Department of Chemical Engineering, McGill University, Montreal, Canada
| | - Jeong-Eun Oh
- Department of Civil and Environmental Engineering, Pusan National University, Busan, 46241, Republic of Korea; Institute for Environment and Energy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
3
|
Quan W, Xu CS, Ma C, Chen X, Yu DH, Li ZY, Wang DW, Tang F, Wan GP, Wan J, Wang ZF, Li ZQ. Anti-tumor effects of telmisartan in glioma-astrocyte non-contact co-cultures: A critical role of astrocytic IL-6-mediated paracrine growth promotion. Int Immunopharmacol 2024; 139:112707. [PMID: 39032472 DOI: 10.1016/j.intimp.2024.112707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/22/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Telmisartan, an angiotensin II type 1 receptor (AT1R) blocker, exhibits broad anti-tumor activity. However, in vitro, anti-proliferative effects are shown at doses far beyond the therapeutic plasma concentration. Considering the role of tumor microenvironment in glioma progression, glioma-astrocyte co-cultures were employed to test the anti-tumor potential of low-dose telmisartan. When a high dose was required for a direct anti-proliferative effect on glioma cell lines, a low dose significantly inhibited glioma cell proliferation and migration in the co-culture system. Under co-culture conditions, upregulated IL-6 expression in astrocytes played a critical role in glioma progression. Silencing IL-6 in astrocytes or IL-6R in glioma cells reduced proliferation and migration. Telmisartan (5 μM) inhibited astrocytic IL-6 expression, and its anti-tumor effects were reversed by silencing IL-6 or IL-6R and inhibiting signal transducer and activator of transcription 3 (STAT3) activity in glioma cells. Moreover, the telmisartan-driven IL-6 downregulation was not imitated by losartan, an AT1R blocker with little capacity of peroxisome proliferator-activated receptor-gamma (PPARγ) activation, but was eliminated by a PPARγ antagonist, indicating that the anti-glioma effects of telmisartan rely on its PPARγ agonistic activity rather than AT1R blockade. This study highlights the importance of astrocytic IL-6-mediated paracrine signaling in glioma growth and the potential of telmisartan as an adjuvant therapy for patients with glioma, especially those with hypertension.
Collapse
Affiliation(s)
- Wei Quan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng-Shi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xi Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dong-Hu Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi-Yu Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dan-Wen Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Feng Tang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Gui-Ping Wan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Hubei, China.
| |
Collapse
|
4
|
Pan Y, Li Y, Fan H, Cui H, Chen Z, Wang Y, Jiang M, Wang G. Roles of the peroxisome proliferator-activated receptors (PPARs) in the pathogenesis of hepatocellular carcinoma (HCC). Biomed Pharmacother 2024; 177:117089. [PMID: 38972148 DOI: 10.1016/j.biopha.2024.117089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024] Open
Abstract
Hepatocellular carcinoma (HCC) holds a prominent position among global cancer types. Classically, HCC manifests in individuals with a genetic predisposition when they encounter risk elements, particularly in the context of liver cirrhosis. Peroxisome proliferator-activated receptors (PPARs), which are transcription factors activated by fatty acids, belong to the nuclear hormone receptor superfamily and play a pivotal role in the regulation of energy homeostasis. At present, three distinct subtypes of PPARs have been recognized: PPARα, PPARγ, and PPARβ/δ. They regulate the transcription of genes responsible for cellular development, energy metabolism, inflammation, and differentiation. In recent years, with the rising incidence of HCC, there has been an increasing focus on the mechanisms and roles of PPARs in HCC. PPARα primarily mediates the occurrence and development of HCC by regulating glucose and lipid metabolism, inflammatory responses, and oxidative stress. PPARβ/δ is closely related to the self-renewal ability of liver cancer stem cells (LCSCs) and the formation of the tumor microenvironment. PPARγ not only influences tumor growth by regulating the glucose and lipid metabolism of HCC, but its agonists also have significant clinical significance for the treatment of HCC. Therefore, this review offers an exhaustive examination of the role of the three PPAR subtypes in HCC progression, focusing on their mediation of critical cellular processes such as glucose and lipid metabolism, inflammation, oxidative stress, and other pivotal signaling pathways. At the end of the review, we discuss the merits and drawbacks of existing PPAR-targeted therapeutic strategies and suggest a few alternative combinatorial therapeutic approaches that diverge from conventional methods.
Collapse
Affiliation(s)
- Yujie Pan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Hongyu Fan
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, No. 246 Baojian Road, Harbin 150086, China
| | - Huijuan Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhiyue Chen
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yunzhu Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Mengyu Jiang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
5
|
Hijazi MA, Gessner A, El-Najjar N. Repurposing of Chronically Used Drugs in Cancer Therapy: A Chance to Grasp. Cancers (Basel) 2023; 15:3199. [PMID: 37370809 DOI: 10.3390/cancers15123199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Despite the advancement in drug discovery for cancer therapy, drug repurposing remains an exceptional opportunistic strategy. This approach offers many advantages (faster, safer, and cheaper drugs) typically needed to overcome increased challenges, i.e., side effects, resistance, and costs associated with cancer therapy. However, not all drug classes suit a patient's condition or long-time use. For that, repurposing chronically used medications is more appealing. This review highlights the importance of repurposing anti-diabetic and anti-hypertensive drugs in the global fight against human malignancies. Extensive searches of all available evidence (up to 30 March 2023) on the anti-cancer activities of anti-diabetic and anti-hypertensive agents are obtained from multiple resources (PubMed, Google Scholar, ClinicalTrials.gov, Drug Bank database, ReDo database, and the National Institutes of Health). Interestingly, more than 92 clinical trials are evaluating the anti-cancer activity of 14 anti-diabetic and anti-hypertensive drugs against more than 15 cancer types. Moreover, some of these agents have reached Phase IV evaluations, suggesting promising official release as anti-cancer medications. This comprehensive review provides current updates on different anti-diabetic and anti-hypertensive classes possessing anti-cancer activities with the available evidence about their mechanism(s) and stage of development and evaluation. Hence, it serves researchers and clinicians interested in anti-cancer drug discovery and cancer management.
Collapse
Affiliation(s)
- Mohamad Ali Hijazi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Beirut Arab University, Beirut P.O. Box 11-5020, Lebanon
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Nahed El-Najjar
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
6
|
Chang YL, Chou CH, Li YF, Huang LC, Kao Y, Hueng DY, Tsai CK. Antiproliferative and apoptotic effects of telmisartan in human glioma cells. Cancer Cell Int 2023; 23:111. [PMID: 37291545 DOI: 10.1186/s12935-023-02963-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
Glioblastoma is the most common primary central nervous system tumor in adults. Angiotensin II receptor blockers (ARBs) are broadly applied to treat hypertension. Moreover, research has revealed that ARBs have the capacity to suppress the growth of several cancer types. In this study, we assessed the effects of three ARBs with the ability to cross the blood brain barrier (telmisartan, valsartan and fimasartan) on cell proliferation in three glioblastoma multiforme (GBM) cell lines. Telmisartan markedly suppressed the proliferation, migration, and invasion of these three GBM cell lines. Microarray data analysis revealed that telmisartan regulates DNA replication, mismatch repair, and the cell cycle pathway in GBM cells. Furthermore, telmisartan induced G0/G1 phase arrest and apoptosis. The bioinformatic analysis and western blotting results provide evidence that SOX9 is a downstream target of telmisartan. Telmisartan also suppressed tumor growth in vivo in an orthotopic transplant mouse model. Therefore, telmisartan is a potential treatment for human GBM.
Collapse
Affiliation(s)
- Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chung-Hsing Chou
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Cheng-Gong Road, Taipei, 11490, Taiwan
| | - Yao-Feng Li
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Ying Kao
- Division of Neurosurgery, Department of Surgery, Taipei City Hospital Zhongxing Branch, Taipei, Taiwan
| | - Dueng-Yuan Hueng
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Cheng-Gong Road, Taipei, 11490, Taiwan.
| |
Collapse
|
7
|
Psilopatis I, Vrettou K, Troungos C, Theocharis S. The Role of Peroxisome Proliferator-Activated Receptors in Endometrial Cancer. Int J Mol Sci 2023; 24:ijms24119190. [PMID: 37298140 DOI: 10.3390/ijms24119190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Endometrial carcinoma is the most common malignant tumor of the female genital tract in the United States. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor proteins which regulate gene expression. In order to investigate the role of PPARs in endometrial cancer, we conducted a literature review using the MEDLINE and LIVIVO databases and were able to identify 27 relevant studies published between 2000 and 2023. The PPARα and PPARβ/δ isoforms seemed to be upregulated, whereas PPARγ levels were reported to be significantly lower in endometrial cancer cells. Interestingly, PPAR agonists were found to represent potent anti-cancer therapeutic alternatives. In conclusion, PPARs seem to play a significant role in endometrial cancer.
Collapse
Affiliation(s)
- Iason Psilopatis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
| | - Kleio Vrettou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
| | - Constantinos Troungos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 16, Goudi, 11527 Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
| |
Collapse
|
8
|
Ribeiro E, Costa B, Vasques-Nóvoa F, Vale N. In Vitro Drug Repurposing: Focus on Vasodilators. Cells 2023; 12:671. [PMID: 36831338 PMCID: PMC9954697 DOI: 10.3390/cells12040671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Drug repurposing aims to identify new therapeutic uses for drugs that have already been approved for other conditions. This approach can save time and resources compared to traditional drug development, as the safety and efficacy of the repurposed drug have already been established. In the context of cancer, drug repurposing can lead to the discovery of new treatments that can target specific cancer cell lines and improve patient outcomes. Vasodilators are a class of drugs that have been shown to have the potential to influence various types of cancer. These medications work by relaxing the smooth muscle of blood vessels, increasing blood flow to tumors, and improving the delivery of chemotherapy drugs. Additionally, vasodilators have been found to have antiproliferative and proapoptotic effects on cancer cells, making them a promising target for drug repurposing. Research on vasodilators for cancer treatment has already shown promising results in preclinical and clinical studies. However, additionally research is needed to fully understand the mechanisms of action of vasodilators in cancer and determine the optimal dosing and combination therapy for patients. In this review, we aim to explore the molecular mechanisms of action of vasodilators in cancer cell lines and the current state of research on their repurposing as a treatment option. With the goal of minimizing the effort and resources required for traditional drug development, we hope to shed light on the potential of vasodilators as a viable therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Francisco Vasques-Nóvoa
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
9
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
10
|
Kast RE, Alfieri A, Assi HI, Burns TC, Elyamany AM, Gonzalez-Cao M, Karpel-Massler G, Marosi C, Salacz ME, Sardi I, Van Vlierberghe P, Zaghloul MS, Halatsch ME. MDACT: A New Principle of Adjunctive Cancer Treatment Using Combinations of Multiple Repurposed Drugs, with an Example Regimen. Cancers (Basel) 2022; 14:2563. [PMID: 35626167 PMCID: PMC9140192 DOI: 10.3390/cancers14102563] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
In part one of this two-part paper, we present eight principles that we believe must be considered for more effective treatment of the currently incurable cancers. These are addressed by multidrug adjunctive cancer treatment (MDACT), which uses multiple repurposed non-oncology drugs, not primarily to kill malignant cells, but rather to reduce the malignant cells' growth drives. Previous multidrug regimens have used MDACT principles, e.g., the CUSP9v3 glioblastoma treatment. MDACT is an amalgam of (1) the principle that to be effective in stopping a chain of events leading to an undesired outcome, one must break more than one link; (2) the principle of Palmer et al. of achieving fractional cancer cell killing via multiple drugs with independent mechanisms of action; (3) the principle of shaping versus decisive operations, both being required for successful cancer treatment; (4) an idea adapted from Chow et al., of using multiple cytotoxic medicines at low doses; (5) the idea behind CUSP9v3, using many non-oncology CNS-penetrant drugs from general medical practice, repurposed to block tumor survival paths; (6) the concept from chess that every move creates weaknesses and strengths; (7) the principle of mass-by adding force to a given effort, the chances of achieving the goal increase; and (8) the principle of blocking parallel signaling pathways. Part two gives an example MDACT regimen, gMDACT, which uses six repurposed drugs-celecoxib, dapsone, disulfiram, itraconazole, pyrimethamine, and telmisartan-to interfere with growth-driving elements common to cholangiocarcinoma, colon adenocarcinoma, glioblastoma, and non-small-cell lung cancer. gMDACT is another example of-not a replacement for-previous multidrug regimens already in clinical use, such as CUSP9v3. MDACT regimens are designed as adjuvants to be used with cytotoxic drugs.
Collapse
Affiliation(s)
| | - Alex Alfieri
- Department of Neurosurgery, Cantonal Hospital of Winterthur, 8400 Winterthur, Switzerland; (A.A.); (M.-E.H.)
| | - Hazem I. Assi
- Naef K. Basile Cancer Center, American University of Beirut, Beirut 1100, Lebanon;
| | - Terry C. Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA;
| | - Ashraf M. Elyamany
- Oncology Unit, Hemato-Oncology Department, SECI Assiut University Egypt/King Saud Medical City, Riyadh 7790, Saudi Arabia;
| | - Maria Gonzalez-Cao
- Translational Cancer Research Unit, Dexeus University Hospital, 08028 Barcelona, Spain;
| | | | - Christine Marosi
- Clinical Division of Medical Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Michael E. Salacz
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA;
| | - Iacopo Sardi
- Department of Pediatric Oncology, Meyer Children’s Hospital, Viale Pieraccini 24, 50139 Florence, Italy;
| | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium;
| | - Mohamed S. Zaghloul
- Children’s Cancer Hospital & National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Marc-Eric Halatsch
- Department of Neurosurgery, Cantonal Hospital of Winterthur, 8400 Winterthur, Switzerland; (A.A.); (M.-E.H.)
| |
Collapse
|
11
|
Associations between AGT M235T Polymorphism and Cancer: An Updated Meta-Analysis. J Renin Angiotensin Aldosterone Syst 2022; 2022:7862709. [PMID: 35317386 PMCID: PMC8916873 DOI: 10.1155/2022/7862709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/17/2022] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
Abstract
We assessed the relationship between AGT gene M235T polymorphism and the susceptibility to cancer by performing an updated meta-analysis. This study retrospectively searched related articles in the electronic databases. Afterwards, we determined combined odds ratios (ORs) and related 95% confidence intervals (CIs) by the fixed- or random-effects model. The present meta-analysis enrolled altogether 9 articles. On the whole, the relationship between AGT M235T polymorphism and the cancer risk was not significant among the entire population (TT vs. MM: OR = 1.28, 95%CI = 0.80 − 2.04; TM vs. MM: OR = 0.90, 95%CI = 0.53 − 1.52; recessive model: OR = 1.13, 95%CI = 0.83 − 1.52; dominant model: OR = 0.93, 95%CI = 0.55 − 1.57). Subgroup analysis by ethnicity, cancer type, and study quality for the relationship between the AGT M235T polymorphism and cancer risk showed no significant association. According to findings in the present meta-analysis, AGT M235T polymorphism may not be related to cancer susceptibility.
Collapse
|
12
|
Khorsand M, Mostafavi-Pour Z, Razban V, Khajeh S, Zare R. Combinatorial effects of telmisartan and docetaxel on cell viability and metastatic gene expression in human prostate and breast cancer cells. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2022; 11:11-20. [PMID: 35463822 PMCID: PMC9012430 DOI: 10.22099/mbrc.2022.42638.1700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is a unique process resulting in enhanced cell motility, invasiveness, and metastasis in cancer. The EMT is regulated by several transcription factors, including Snail and Slug, which exert crucial roles during cancer progression. We have studied the effects of Docetaxel as the first-line chemotherapy agent for prostate cancer, and Telmisartan as an anti-hypertensive drug on the expression level of Snail and Slug. In addition, the effects of Docetaxel, Telmisartan and their combination on cancer cell proliferation were investigated. The PC3, DU145, MDA-MB468, and HEK cell lines were used for this study. Quantitative RT-PCR analysis and MTT assay were used to study the expression of Snail and Slug level and cell proliferative assay, respectively. We found that a combination of Docetaxel + Telmisartan effectively inhibits the cell proliferation in cancerous cells in comparison with each drug alone (P<0.05). Furthermore, in these cell lines, Docetaxel, Telmisartan and their combination significantly diminished the expression level of Snail and Slug genes compared to control cells (P<0.001), however, in the HEK cell line, this effect was seen only in the combination group. Our data imply that Telmisartan and its combination with Docetaxel exert strong inhibitory effects on the expression level of Snail and Slug genes. Also, these drugs and their combination could inhibit cancer cell proliferation. In conclusion, the combination of Telmisartan and Docetaxel has the potential to suppress the metastasis of prostate and breast cancer cells.
Collapse
Affiliation(s)
- Marjan Khorsand
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zohreh Mostafavi-Pour
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran,Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,#Zohreh Mostafavi-Pour and Vahid Razban are both corresponding authors and have got the same contribution in this work,Corresponding Author: Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.Tel:+98 71-32 303029 ; Fax: +98 71-32 303029 , E. mail:
| | - Vahid Razban
- Molecular Medicine Department, School of Advanced Medical Sciences and Technology, Shiraz University of Medical Sciences, Shiraz, Iran,Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,#Zohreh Mostafavi-Pour and Vahid Razban are both corresponding authors and have got the same contribution in this work
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Zare
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Khorsand M, Khajeh S, Eslami M, Nezafat N, Ghasemi Y, Razban V, Mostafavi‐Pour Z. Telmisartan anti‐cancer activities mechanism through targeting N‐cadherin by mimicking ADH‐1 function. J Cell Mol Med 2022; 26:2392-2403. [PMID: 35224849 PMCID: PMC8995460 DOI: 10.1111/jcmm.17259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/02/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
This study aimed to investigate if Telmisartan as a novel N‐cadherin antagonist, can overcome cell migration of cancer cells. We investigated the mechanism and influence of Docetaxel and Telmisartan (as an analogous to ADH‐1, which is a well‐known N‐cadherin antagonist) on cancer cells. The effect of ADH‐1 and Telmisartan on cell attachment in PC3, DU145, MDA‐MB‐468 cell lines using recombinant human N‐cadherin was studied. Cell viability assay was performed to examine the anti‐proliferative effects of Telmisartan, ADH‐1 and Docetaxel. Migration was examined via wound healing assay, and apoptosis was determined by flow cytometry. The expression of AKT‐1 as a downstream gene of N‐cadherin signalling pathway was assayed by real‐time PCR. Treatment of PC3, MDA‐MB‐468 and DU145 cells with Telmisartan (0.1 µM) and ADH‐1 (40 µM) resulted in 50%, 58% and approximately 20% reduction in cell attachment to N‐cadherin coated plate respectively. It shows reduction of cell attachment in PC3 and MDA‐MB‐468 cell lines appeared to be more sensitive than that of DU145 cells to the Telmisartan and ADH‐1 treatments. Telmisartan (0.1 µM) and Docetaxel (0.01 nM) significantly reduced cell migration in PC3 and MDA‐MB‐468 cell lines compared with the control group. Using Real‐time PCR, we found that Telmisartan, Docetaxel and ADH‐1 had significant influence on the AKT‐1 mRNA level. The results of the current study for the first time suggest that, Telmisartan, exerts anti‐proliferation and anti‐migration effects by targeting antagonistically N‐cadherin. Also, these data suggest that Telmisartan as a less expensive alternative to ADH‐1 could potentiate Docetaxel anticancer effects.
Collapse
Affiliation(s)
- Marjan Khorsand
- Department of Biochemistry School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Mahboobeh Eslami
- Pharmaceutical Sciences Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center Shiraz University of Medical Sciences Shiraz Iran
- Department of Pharmaceutical Biotechnology School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center Shiraz University of Medical Sciences Shiraz Iran
- Department of Pharmaceutical Biotechnology School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Vahid Razban
- Molecular Medicine Department School of Advanced Medical Sciences and Technology Shiraz University of Medical Sciences Shiraz Iran
- Stem Cell Technology Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Zohreh Mostafavi‐Pour
- Department of Biochemistry School of Medicine Shiraz University of Medical Sciences Shiraz Iran
- Autophagy Research Center Shiraz University of Medical Sciences Shiraz Iran
| |
Collapse
|
14
|
Tsujiya Y, Hasegawa A, Yamamori M, Okamura N. Telmisartan-Induced Cytotoxicity via G 2/M Phase Arrest in Renal Cell Carcinoma Cell Lines. Biol Pharm Bull 2021; 44:1878-1885. [PMID: 34853271 DOI: 10.1248/bpb.b21-00654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney cancer. Given that stage IV RCC is intractable, there is a need for a novel treatment strategy. We investigated the antitumor effects of telmisartan (TEL) and their underlying mechanisms in RCC, including their impact on apoptosis, Akt/mammalian target of rapamycin (mTOR) pathways, and the cell cycle using two human RCC cell lines: 786-O and Caki-2. Cell viability was detected via fluorescence-based assays. Cells were stained with Hoechst 33342 to observe chromatin condensation, and Western blotting was performed to analyze protein expression. The cell cycle was assessed using flow cytometry. Invasion and migration assays were performed using 24-well chambers. TEL induced cell death in a dose-dependent manner and increased the percentage of cells with high chromatin condensation and Bax/Bcl-2 ratio in both cell lines. TEL-induced cell death was attenuated by neither peroxisome proliferator-activated receptor-γ nor -δ inhibitors. Although TEL elevated c-Jun N-terminal kinase levels and p38 phosphorylation rates in Caki-2 cells, as well as extracellular signal-regulated kinase phosphorylation rates in 786-O cells, their inhibitors did not suppress TEL-induced cell death. TEL decreased Akt phosphorylation in 786-O cells and mTOR phosphorylation in both cell lines, increased the population of cells in the G2/M phase, and altered G2/M-related proteins in both cell lines. TEL moderately suppressed cell invasion and migration in 786-O and Caki-2 cells, respectively, and increased cell invasion in Caki-2 cells, suggesting a potential therapeutic role of TEL in RCC.
Collapse
Affiliation(s)
- Yoshie Tsujiya
- Department of Clinical Pharmacy, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Ai Hasegawa
- Department of Clinical Pharmacy, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Motohiro Yamamori
- Department of Clinical Pharmacy, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Noboru Okamura
- Department of Clinical Pharmacy, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| |
Collapse
|
15
|
Avsar T, Yigit BN, Turan G, Altunsu D, Calis S, Kurt B, Kilic T, Yavuz Ergun M, Durdagi S, Acar M. Development of imidazolone based angiotensin II receptor type I inhibitor small molecule as a chemotherapeutic agent for cell cycle inhibition. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1954098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Timucin Avsar
- Department of Medical Biology, School of Medicine, Bahcesehir University, Istanbul, Turkey
- Neuroscience Program, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
| | - Berfu Nur Yigit
- Neuroscience Program, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
| | - Gizem Turan
- Neuroscience Program, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
| | - Deniz Altunsu
- Neuroscience Program, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
| | - Seyma Calis
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Graduate School of Science, Engineering and Technology, Molecular Biology, Genetics and Biotechnology Graduate Program, Istanbul Technical University, Istanbul, Turkey
| | - Bahar Kurt
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
| | - Turker Kilic
- Neuroscience Program, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Department of Neurosurgery, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - M. Yavuz Ergun
- Department of Chemistry, Dokuz Eylul University, Izmir, Turkey
| | - Serdar Durdagi
- Neuroscience Program, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
- Department of Biophysics, Computational Biology and Molecular Simulations Laboratory, School of Medicine, Bahcesehir University Istanbul, Turkey
| | - Melih Acar
- Department of Medical Biology, School of Medicine, Bahcesehir University, Istanbul, Turkey
- Neuroscience Laboratory, Health Sciences Institute, Bahcesehir University, Istanbul, Turkey
| |
Collapse
|
16
|
Morsy MA, Abdelraheem WM, El-Hussieny M, Refaie MMM. Protective Effects of Irbesartan, an Angiotensin Receptor Blocker with PPARγ Agonistic Activity, against Estradiol Benzoate-Induced Endometrial Hyperplasia and Atypia in Female Rats via Modulation of TNFα/Survivin Pathway. Pharmaceuticals (Basel) 2021; 14:ph14070649. [PMID: 34358075 PMCID: PMC8308641 DOI: 10.3390/ph14070649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022] Open
Abstract
Endometrial hyperplasia (EH) is a common gynecological problem and may progress to carcinoma. Early detection and management of EH are mandatory for the prevention of endometrial cancer. Activation of the renin-angiotensin system and angiotensin II signaling are involved in the progression of precancerous and cancerous lesions. However, no studies have evaluated the role of this system in estradiol benzoate (EB)-induced EH and atypia. Irbesartan (IRB), an angiotensin II receptor blocker with peroxisome proliferator-activated receptor gamma (PPARγ) agonistic activity was administered (30 mg/kg/d) in EB-treated (60 µg/100 g bodyweight, intramuscularly, three times per week) or untreated rats for 4 weeks. Uterine weight changes, malondialdehyde, superoxide dismutase (SOD), tumor necrosis factor-alpha (TNFα), survivin, cleaved caspase 3, interleukin-10 (IL10), and PPARγ were measured in addition to undergoing histopathological examination. Results showed that EB-induced EH and atypia significantly increased the uterine body weight, malondialdehyde, TNFα, and survivin, accompanied with significantly decreased SOD, cleaved caspase 3, IL10, and PPARγ, with typical histopathological changes of EH and atypia. Coadministration of IRB significantly prevented EB-induced biochemical and histopathological changes. The protective effects of IRB may be attributed to its anti-inflammatory and antioxidant properties, reduction of survivin, and increased levels of cleaved caspase 3.
Collapse
Affiliation(s)
- Mohamed A. Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt;
- Correspondence: ; Tel.: +966-5496-72245
| | - Wedad M. Abdelraheem
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt;
| | - Maram El-Hussieny
- Department of Pathology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt;
| | - Marwa M. M. Refaie
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt;
| |
Collapse
|
17
|
Taghvaei S, Sabouni F, Minuchehr Z. Evidence of Omics, Immune Infiltration, and Pharmacogenomic for SENP1 in the Pan-Cancer Cohort. Front Pharmacol 2021; 12:700454. [PMID: 34276383 PMCID: PMC8280523 DOI: 10.3389/fphar.2021.700454] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/14/2021] [Indexed: 12/20/2022] Open
Abstract
Sentrin specific-protease 1 (SENP1) is a protein involved in deSUMOylation that is almost overexpressed in cancer. SENP1 has a determinative role in the activation of transcription programs in the innate immune responses and the development B of and C lymphocytes. We found, SENP1 possibly plays a critical role in immune infiltration and acts as an expression marker in PAAD, ESCA, and THYM. CD4+ T cells, CD8+ T cells, and macrophages were more key-related immune cells, indicating that SENP1 might be introduced as a potential target for cancer immunotherapy. We further showed that dysregulation of SENP1 is powerfully associated with decreased patient survival and clinical stage. Total SENP1 protein also increases in cancer. SENP1 is also controlled by transcription factors (TFs) CREB1, KDM5A, REST, and YY1 that regulates apoptosis, cell cycle, cell proliferation, invasion, tumorigenesis, and metastasis. These TFs were in a positive correlation with SENP1. MiR-138-5p, miR-129-1-3p, and miR-129-2-3p also inhibit tumorigenesis through targeting of SENP1. The SENP1 expression level positively correlated with the expression levels of UBN1, SP3, SAP130, NUP98, NUP153 in 32 tumor types. SENP1 and correlated and binding genes: SAP130, NUP98, and NUP153 activated cell cycle. Consistent with this finding, drug analysis was indicated SENP1 is sensitive to cell cycle, apoptosis, and RTK signaling regulators. In the end, SENP1 and its expression-correlated and functional binding genes were enriched in cell cycle, apoptosis, cellular response to DNA damage stimulus. We found that the cell cycle is the main way for tumorigenesis by SENP1. SENP1 attenuates the effect of inhibitory drugs on the cell cycle. We also introduced effective FDA-Approved drugs that can inhibit SENP1. Therefore in the treatments in which these drugs are used, SENP1 inhibition is a suitable approach. This study supplies a wide analysis of the SENP1 across The Cancer Genome Atlas (CGA) cancer types. These results suggest the potential roles of SENP1 as a biomarker for cancer. Since these drugs and the drugs that cause to resistance are applied to cancer treatment, then these two class drugs can use to inhibition of SENP1.
Collapse
Affiliation(s)
- Somayye Taghvaei
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farzaneh Sabouni
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zarrin Minuchehr
- Department of Systems Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
18
|
Wang Y, Zhang T, Li C, Guo J, Xu B, Xue L. Telmisartan attenuates human glioblastoma cells proliferation and oncogenicity by inducing the lipid oxidation. Asia Pac J Clin Oncol 2021; 18:217-223. [PMID: 33945216 PMCID: PMC9290901 DOI: 10.1111/ajco.13574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
Background Glioblastoma (GBM) is one of the most common primary brain tumors, which accounts up to 80% of malignant brain tumors and the 5‐year relative survival rate is below 5%. Recent studies showed that the lipid metabolism played an essential role in GBM development. As a peroxisome proliferators‐activated receptors γ (PPAR‐γ) agonist, telmisartan improves the lipid metabolism and has been used to treat hypertension for long time. It has also been shown to have anticancer function, such as in lung cancer and melanoma. Methods Incucyte real‐time live cell imaging system was used to assess the effect of telmisartan on glioma cell lines U87 and U251 proliferation. Transwell assay and colony formation assay were conducted to detect the effect of telmisartan on oncogenicity of GBM cell lines. Western blot and immunofluorescence analysis were used to detect the effect of telmisartan on the expression of PPAR‐γ and hydroxyacyl‐coenzyme A dehydrogenase alpha subunit (HADHA). Results We demonstrate that telmisartan inhibits two glioma cell lines U87 and U251 proliferation in a time‐ and dose‐dependent manner, and arrests the cell cycle at S phase. We further show that telmisartan decreases the oncogenicity of GBM cell lines. Our data show that telmisartan treatment significantly increases the PPAR‐γ expression level, enhances the lipid oxidation, and upregulates the level of fatty acid oxidation key enzyme HADHA. Conclusions Telmisartan inhibits the proliferation and oncogenicity while it also increases the lipid oxidation of human GBM cells.
Collapse
Affiliation(s)
- Yan Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.,Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Tengrui Zhang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.,Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Chen Li
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.,Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, China.,Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Baohui Xu
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.,Medical Research Center, Peking University Third Hospital, Beijing, China.,Biobank, Peking University Third Hospital, Beijing, China
| |
Collapse
|
19
|
IL13Rα2 Is Involved in the Progress of Renal Cell Carcinoma through the JAK2/FOXO3 Pathway. J Pers Med 2021; 11:jpm11040284. [PMID: 33917914 PMCID: PMC8068290 DOI: 10.3390/jpm11040284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
Previously, we reported a close relationship between type II IL4Rα and IL13Rα1 complex and poor outcomes in renal cell carcinoma (RCC). In this study, we investigated the clinicopathologically significant oncogenic role of IL13Rα2, a kind of the independent receptor for IL13, in 229 RCC patients. The high expression of IL13Rα2 was closely related to relapse-free survival in specific cancers in univariate and multivariate analysis. Then, the oncogenic role of IL13Rα2 was evaluated by performing in vitro assays for cell proliferation, cell cycle arrest, and apoptosis in A498, ACHN, Caki1, and Caki2, four kinds of RCC cells after transfection of siRNA against IL13Rα2. Cell proliferation was suppressed, and apoptosis was induced in A498, ACHN, Caki1, and Caki2 cells by knockdown of IL13Rα2. Interestingly, the knockdown of IL13Rα2 decreased the phosphorylation of JAK2 and increased the expression of FOXO3. Furthermore, the knockdown of IL13Rα2 reduced the protein interaction among IL13Rα2, phosphorylated JAK2, and FOXO3. Since phosphorylation of JAK2 was regulated by IL13Rα2, we tried to screen a novel JAK2 inhibitor from the FDA-approved drug library and selected telmisartan, a clinically used medicine against hypertension, as one of the strongest candidates. Telmisartan treatment decreased the cell proliferation rate and increased apoptosis in A498, ACHN, Caki1, and Caki2 cells. Mechanistically, telmisartan treatment decreased the phosphorylation of JAK2 and increased the expression of FOXO3. Taken together, these results suggest that IL13Rα2 regulates the progression of RCC via the JAK2/FOXO3-signaling path pathway, which might be targeted as the novel therapeutic option for RCC patients.
Collapse
|
20
|
Ziaja M, Urbanek KA, Kowalska K, Piastowska-Ciesielska AW. Angiotensin II and Angiotensin Receptors 1 and 2-Multifunctional System in Cells Biology, What Do We Know? Cells 2021; 10:cells10020381. [PMID: 33673178 PMCID: PMC7917773 DOI: 10.3390/cells10020381] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
For years, the renin-angiotensin system (RAS) has been perceived as a system whose role is to primarily modulate the functioning of the cardiovascular system. Years of research into the role of RAS have provided the necessary data to confirm that the role of RAS is very complex and not limited to the cardiovascular system. The presence of individual elements of the renin-angiotensin (RA) system allows to control many processes, ranging from the memorization to pro-cancer processes. Maintaining the proportions between the individual axes of the RA system allows for achieving a balance, often called homeostasis. Thus, any disturbance in the expression or activity of individual RAS elements leads to pathophysiological processes.
Collapse
|
21
|
ÖztÜrk N, Kara A, Vural İ. Formulation and In Vitro Evaluation of Telmisartan Nanoparticles Prepared by Emulsion-Solvent Evaporation Technique. Turk J Pharm Sci 2020; 17:492-499. [PMID: 33177929 DOI: 10.4274/tjps.galenos.2019.76402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 07/30/2019] [Indexed: 12/01/2022]
Abstract
Objectives Telmisartan (TLM) is an antihypertensive drug that has been shown to have antiproliferative effects on cancer cells. It has low solubility and suboptimal oral bioavailability. To investigate the potential anticancer effect of TLM on breast cancer cells, poly (D, L-lactide) (PLA) nanoparticles were formulated with the benefit of improving its solubility. Materials and Methods TLM-loaded PLA nanoparticles were prepared by emulsion solvent evaporation. The effects of sonication time and polymer:drug ratio on nanoparticle size and drug encapsulation were investigated. TLM-loaded nanoparticles were tested against MCF-7 and MD-AMB-231 breast cancer cell lines for antiproliferative effects. Results Nanoparticles with mean particle size 272 nm and 79% encapsulation efficiency were obtained. Sustained release TLM nanoparticles (40% in 24 h) decreased cell viability to 45% for MCF-7 cells at 72 h, even at the lowest TLM concentration, indicating better anticancer efficiency than TLM solution. Conclusion TLM nanoparticles could be potential anticancer agents for breast cancer and deserve further studies.
Collapse
Affiliation(s)
- Naile ÖztÜrk
- İnönü University Faculty of Pharmacy, Department of Pharmaceutical Technology, Malatya, Turkey
| | - Aslı Kara
- Hitit University Sungurlu Vocational High School, Department of Medical Services and Techniques, Çorum, Turkey
| | - İmran Vural
- Hacettepe University Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara, Turkey
| |
Collapse
|
22
|
Martínez VR, Aguirre MV, Todaro JS, Ferrer EG, Williams PAM. Improvement of the Anticancer Activities of Telmisartan by Zn(II) Complexation and Mechanisms of Action. Biol Trace Elem Res 2020; 197:454-463. [PMID: 31863274 DOI: 10.1007/s12011-019-02013-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
To improve the anticancer activity of telmisartan, its structure has been modified by Zn(II) complexation giving [Zn(Telm)2(H2O)2]·2H2O (ZnTelm). The cytotoxic effect was measured on the human lung cancer cells (A549) and on the lung fibroblast cells (MRC-5). The complex markedly improved anticancer activity (IC50 75 μM) of telmisartan (IC50 125 μM) or ZnSO4 (IC50 225 μM) and did not show toxicity on non-cancer cells, inducing oxidative stress with cellular ROS generation and GSH/GSSG decrease. Apoptosis was the dominant form of cell death for the complex. The Bax/Bcl-XL ratio was significantly increased as well as caspase-3 activation. Both the complex and the ligand bind to bovine serum albumin (BSA) and can be stored and transported by the protein but the interaction with the complex is greater. Telmisartan binds BSA by hydrophobic interactions while the interaction of ZnTelm occurs through van der Waals forces and hydrogen bonding. Therefore, it can be shown that the coordination complex ZnTelm improved the anticancer activity of the antihypertensive drug telmisartan (IC50 75 μM and 125 μM, respectively) and the interaction with BSA. Graphical Abstract Improvement of the anticancer activities of telmisartan by Zn(II) complexation and mechanisms of action. Intrinsic apoptotic pathway: induction ofoxidative stress and regulation of proteins related to apoptosis. The complex interacted with bovine serum albumin (BSA) and can be stored and transported by the protein.
Collapse
Affiliation(s)
- Valeria R Martínez
- Centro de Química Inorgánica (CEQUINOR-CONICET-CICPBA-UNLP), 120 N° 1465, La Plata, Argentina
| | - María V Aguirre
- Laboratorio de Investigaciones Bioquímicas. Facultad de Medicina. UNNE, Moreno 1240, Corrientes, Argentina
| | - Juan S Todaro
- Laboratorio de Investigaciones Bioquímicas. Facultad de Medicina. UNNE, Moreno 1240, Corrientes, Argentina
| | - Evelina G Ferrer
- Centro de Química Inorgánica (CEQUINOR-CONICET-CICPBA-UNLP), 120 N° 1465, La Plata, Argentina
| | - Patricia A M Williams
- Centro de Química Inorgánica (CEQUINOR-CONICET-CICPBA-UNLP), 120 N° 1465, La Plata, Argentina.
| |
Collapse
|
23
|
Yang Y, Li X, Wang T, Guo Q, Xi T, Zheng L. Emerging agents that target signaling pathways in cancer stem cells. J Hematol Oncol 2020; 13:60. [PMID: 32456660 PMCID: PMC7249421 DOI: 10.1186/s13045-020-00901-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/18/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) contribute to the initiation, recurrence, and metastasis of cancer; however, there are still no drugs targeting CSCs in clinical application. There are several signaling pathways playing critical roles in CSC progression, such as the Wnt, Hedgehog, Notch, Hippo, and autophagy signaling pathways. Additionally, targeting the ferroptosis signaling pathway was recently shown to specifically kill CSCs. Therefore, targeting these pathways may suppress CSC progression. The structure of small-molecule drugs shows a good spatial dispersion, and its chemical properties determine its good druggability and pharmacokinetic properties. These characteristics make small-molecule drugs show a great advantage in drug development, which is increasingly popular in the market. Thus, in this review, we will summarize the current researches on the small-molecule compounds suppressing CSC progression, including inhibitors of Wnt, Notch, Hedgehog, and autophagy pathways, and activators of Hippo and ferroptosis pathways. These small-molecule compounds emphasize CSC importance in tumor progression and propose a new strategy to treat cancer in clinic via targeting CSCs.
Collapse
Affiliation(s)
- Yue Yang
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Ting Wang
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Qianqian Guo
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450003, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
24
|
Cheng Z, Liu Z. Renin-angiotensin system gene polymorphisms and colorectal cancer risk: a meta-analysis. J Renin Angiotensin Aldosterone Syst 2020; 20:1470320319881932. [PMID: 31642377 PMCID: PMC6811762 DOI: 10.1177/1470320319881932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE The renin-angiotensin system gene has been implicated in the progression of colorectal cancer. Nevertheless, the details of that role remain controversial. We performed a meta-analysis to investigate the correlation between renin-angiotensin system gene polymorphisms and colorectal cancer. METHODS We retrieved relevant studies from PubMed and Embase. Subsequently, fixed or random-effects models were used to calculate pooled odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS We identified six studies of the angiotensin-converting enzyme insertion/deletion (I/D) polymorphism, and two studies of the angiotensinogen M235T polymorphism. The angiotensin-converting enzyme I/D polymorphism did not significantly correlate with colorectal cancer risk in the total population (DD vs. II: OR 0.77, 95% CI 0.39-1.50; DI vs. II: OR 1.05, 95% CI 0.85-1.30; dominant model: OR 0.94, 95% CI 0.68-1.31; recessive model: OR 1.01, 95% CI 0.80-1.27). Similarly, the angiotensinogen M235T polymorphism was not associated with colorectal cancer risk (TT vs. MM: OR 1.38, 95% CI 0.52-3.67; TM vs. MM: OR 1.19, 95% CI 0.96-1.47; dominant model: OR 1.28, 95% CI 0.77-2.14; recessive model: OR 1.17, 95% CI 0.53-2.59). CONCLUSION Our findings suggest that the angiotensin-converting enzyme I/D and angiotensinogen M235T polymorphisms are unlikely to correlate with colorectal cancer.
Collapse
Affiliation(s)
- Zhen Cheng
- International Healthcare Center, Sir Run Run Shaw Hospital, China
| | - Zhiwei Liu
- Laboratory Department, Sir Run Run Shaw Hospital, China
| |
Collapse
|
25
|
Zhu Y, Yu F, Tan Y, Hong Y, Meng T, Liu Y, Dai S, Qiu G, Yuan H, Hu F. Reversing activity of cancer associated fibroblast for staged glycolipid micelles against internal breast tumor cells. Am J Cancer Res 2019; 9:6764-6779. [PMID: 31660067 PMCID: PMC6815968 DOI: 10.7150/thno.36334] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 07/15/2019] [Indexed: 01/01/2023] Open
Abstract
Rationale: Nano-carrier based combinational therapies for tumor cells hold great potential to improve the outcomes of patients. However, cancer associated fibroblasts (CAFs) in desmoplastic tumors and the derived pathological tumor stroma severely impede the access and sensitibity of tumor cells to antitumor therapies. Methods: Glycolipid-based polymeric micelles (GLPM) were developed to encapsulate an angiotensin II receptor I inhibitor (telmisartan, Tel) and a cytotoxic drug (doxorubicin, DOX) respectively, which could exert combinational antitumor efficacy by reprogramming tumor microenvironment to expose the vulnerability of internal tumor cells. Results: As demonstrated, α-SMA positive CAFs significantly decreased after the pre-administration of GLPM/Tel in vitro, which accordingly inhibited the secretion of the CAFs derived stroma. The tumor vessels were further decompressed as a result of the alleviated solid stress inside the tumor masses, which promoted more intratumoral drug delivery and penetration. Ultimately, staged administration of the combined GLPM/Tel and GLPM/DOX at the screened molar ratio not only inhibited the stroma continuously, but also achieved a synergistic antitumor effect through the apoptosis-related peroxisome proliferator-activated receptor-gamma (PPAR-γ) pathway. Conclusion: In summary, the strategy of suppressing tumor stroma for subsequent combinational therapies against internal breast tumor cells could provide avenues for management of intractable desmoplastic tumors.
Collapse
|
26
|
Saber S, Basuony M, Eldin AS. Telmisartan ameliorates dextran sodium sulfate-induced colitis in rats by modulating NF-κB signalling in the context of PPARγ agonistic activity. Arch Biochem Biophys 2019; 671:185-195. [DOI: 10.1016/j.abb.2019.07.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 01/09/2023]
|
27
|
Telmisartan attenuates N-nitrosodiethylamine-induced hepatocellular carcinoma in mice by modulating the NF-κB-TAK1-ERK1/2 axis in the context of PPARγ agonistic activity. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1591-1604. [PMID: 31367864 DOI: 10.1007/s00210-019-01706-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/23/2019] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is characterized by bad prognosis and is the second most common reason for cancer-linked mortality. Treatment with sorafenib (SRF) alone increases patient survival by only a few months. A causal link has been determined between angiotensin II (Ang-II) and HCC. However, the mechanisms underlying the tumorigenic effects of Ang-II remain to be elucidated. N-Nitrosodiethylamine was utilized to examine the effects of telmisartan (TEL) (15 mg/kg), SRF (30 mg/kg), and a combination of these two agents on HCC mice. Downregulation of NF-кBP65 mRNA expression and inhibition of the phosphorylation-induced activation of both ERK1/2 and NF-кB P65 were implicated in the anti-tumor effects of TEL and SRF. Consequent regression of malignant changes and improvements in liver function associated with reduced levels of AFP, TNF-α, and TGF-β1 were also confirmed. Anti-proliferative, anti-metastatic, and anti-angiogenic effects of treatment were indicated by reduced hepatic cyclin D1 mRNA expression, reduced MMP-2 levels, and reduced VEGF levels, respectively. TEL, but not SRF, demonstrated agonistic activity for PPARγ receptors, as evidenced by increased PPARγ DNA binding activity, upregulation of CD36, and HO-1 mRNA expression followed by increased liver antioxidant capacity. Both TEL and SRF inhibited TAK1 phosphorylation-induced activation, indicating that TAK1 might act as a central mediator in the interaction between ERK1/2 and NF-кB. TEL, by modulating the ERK1/2, TAK1, and NF-кB signaling axis in the context of PPARγ agonistic activity, exerted anti-tumor effects and increased tumor sensitivity to SRF. Therefore, TEL is an encouraging agent for further clinical trials regarding the management of HCC.
Collapse
|
28
|
Matsui T, Chiyo T, Kobara H, Fujihara S, Fujita K, Namima D, Nakahara M, Kobayashi N, Nishiyama N, Yachida T, Morishita A, Iwama H, Masaki T. Telmisartan Inhibits Cell Proliferation and Tumor Growth of Esophageal Squamous Cell Carcinoma by Inducing S-Phase Arrest In Vitro and In Vivo. Int J Mol Sci 2019; 20:ijms20133197. [PMID: 31261874 PMCID: PMC6651359 DOI: 10.3390/ijms20133197] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common primary esophageal malignancy. Telmisartan, an angiotensin II type 1 (AT1) receptor blocker (ARB) and a widely used antihypertensive, has been shown to inhibit proliferation of various cancer types. This study evaluated the effects of telmisartan on human ESCC cell proliferation in vitro and in vivo and sought to identify the microRNAs (miRNAs) involved in these antitumor effects. We examined the effects of telmisartan on three human ESCC cell lines (KYSE150, KYSE180, and KYSE850). Telmisartan inhibited proliferation of these three cell lines by inducing S-phase arrest, which was accompanied by decreased expression of cyclin A2, cyclin-dependent kinase 2, and other cell cycle-related proteins. Additionally, telmisartan reduced levels of phosphorylated ErbB3 and thrombospondin-1 in KYSE180 cells. Furthermore, expression of miRNAs was remarkably altered by telmisartan in vitro. Telmisartan also inhibited tumor growth in vivo in a xenograft mouse model. In conclusion, telmisartan inhibited cell proliferation and tumor growth in ESCC cells by inducing cell-cycle arrest.
Collapse
Affiliation(s)
- Takanori Matsui
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Taiga Chiyo
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Shintaro Fujihara
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Koji Fujita
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Daisuke Namima
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Mai Nakahara
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Nobuya Kobayashi
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Noriko Nishiyama
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Tatsuo Yachida
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan
| | - Hisakazu Iwama
- Life Science Research Center, Kagawa University, Kagawa 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa 761-0793, Japan.
| |
Collapse
|
29
|
Kwon YJ, Suh GH, Kang SS, Kim HJ. Successful management of proteinuria and systemic hypertension in a dog with renal cell carcinoma with surgery, telmisartan, and amlodipine. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2018; 59:759-762. [PMID: 30026623 PMCID: PMC6005071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An 11-year-old neutered male Yorkshire terrier dog was presented with a 3-week history of hematuria and anorexia. A unilateral renal mass was detected and surgically removed. The renal mass was diagnosed on histopathologic examination as a renal carcinoma. Supportive medical therapy was carried out and persistent systemic hypertension was managed using telmisartan.
Collapse
Affiliation(s)
- Yong-Jin Kwon
- College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea
| | - Guk-Hyun Suh
- College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea
| | - Seong-Soo Kang
- College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea
| | - Ha-Jung Kim
- College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea
| |
Collapse
|
30
|
Casalechi M, Dela Cruz C, Lima LC, Maciel LP, Pereira VM, Reis FM. Angiotensin peptides in the non-gravid uterus: Paracrine actions beyond circulation. Peptides 2018; 101:145-149. [PMID: 29367076 DOI: 10.1016/j.peptides.2018.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
Abstract
The renin-angiotensin system (RAS) involves a complex network of precursors, peptides, enzymes and receptors comprising a systemic (endocrine) and a local (paracrine/autocrine) system. The local RAS plays important roles in tissue modulation and may operate independently of or in close interaction with the circulatory RAS, acting in a complementary fashion. Angiotensin (Ang) II, its receptor AT1 and Ang-(1-7) expression in the endometrium vary with menstrual cycle, and stromal cell decidualization in vitro is accompanied by local synthesis of angiotensinogen and prorenin. Mas receptor is unlikely to undergo marked changes accompanying the cyclic ovarian steroid hormone fluctuations. Studies investigating the functional relevance of the RAS in the non-gravid uterus show a number of paracrine effects beyond circulation and suggest that RAS peptides may be involved in the pathophysiology of proliferative and fibrotic diseases. Endometrial cancer is associated with increased expression of Ang II, Ang-converting enzyme 1 and AT1 in the tumoral tissue compared to neighboring non-neoplastic endometrium, and also with a gene polymorphism that enhances AT1 signal. Ang II induces human endometrial cells to transdifferentiate into cells with myofibroblast phenotype and to synthetize extracellular matrix components that might contribute to endometrial fibrosis. Altogether, these findings point to a fully operating RAS within the uterus, but since many concepts rely on preliminary evidence further studies are needed to clarify the role of the local RAS in uterine physiology and pathophysiology.
Collapse
Affiliation(s)
- Maíra Casalechi
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cynthia Dela Cruz
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza C Lima
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana P Maciel
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Virgínia M Pereira
- Department of Veterinary Medicine, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Fernando M Reis
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
31
|
The angiotensin II type 1 receptor antagonist telmisartan inhibits cell proliferation and tumor growth of esophageal adenocarcinoma via the AMPKα/mTOR pathway in vitro and in vivo. Oncotarget 2018; 8:8536-8549. [PMID: 28052030 PMCID: PMC5352420 DOI: 10.18632/oncotarget.14345] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/05/2016] [Indexed: 01/18/2023] Open
Abstract
Telmisartan, a widely used antihypertensive drug, is an angiotensin II type 1 (AT1) receptor blocker (ARB). This drug inhibits cancer cell proliferation, but the underlying mechanisms in various cancers, including esophageal cancer, remain unknown. The aim of the present study was to evaluate the effects of telmisartan on human esophageal cancer cell proliferation in vitro and in vivo. We assessed the effects of telmisartan on human esophageal adenocarcinoma (EAC) cells using the cell lines OE19, OE33, and SKGT-4. Telmisartan inhibited the proliferation of these three cell lines via blockade of the G0 to G1 cell cycle transition. This blockade was accompanied by a strong decrease in cyclin D1, cyclin E, and other cell cycle-related proteins. Notably, the AMP-activated protein kinase (AMPK) pathway, a fuel sensor signaling pathway, was enhanced by telmisartan. Compound C, which inhibits the two catalytic subunits of AMPK, enhanced the expression of cyclin E, leading to G0/G1 arrest in human EAC cells. In addition, telmisartan reduced the phosphorylation of epidermal growth factor receptor (p-EGFR) and ERBB2 in vitro. In our in vivo study, intraperitoneal injection of telmisartan led to a 73.2% reduction in tumor growth in mice bearing xenografts derived from OE19 cells. Furthermore, miRNA expression was significantly altered by telmisartan in vitro and in vivo. In conclusion, telmisartan suppressed human EAC cell proliferation and tumor growth by inducing cell cycle arrest via the AMPK/mTOR pathway.
Collapse
|
32
|
Samukawa E, Fujihara S, Oura K, Iwama H, Yamana Y, Tadokoro T, Chiyo T, Kobayashi K, Morishita A, Nakahara M, Kobara H, Mori H, Okano K, Suzuki Y, Himoto T, Masaki T. Angiotensin receptor blocker telmisartan inhibits cell proliferation and tumor growth of cholangiocarcinoma through cell cycle arrest. Int J Oncol 2017; 51:1674-1684. [PMID: 29075786 PMCID: PMC5673010 DOI: 10.3892/ijo.2017.4177] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022] Open
Abstract
Cholangiocarcinoma (CCA) is at an advanced stage at the time of its diagnosis, and developing a more effective treatment of CCA would be desirable. Angiotensin II type 1 (AT1) receptor blocker (ARB), telmisartan may inhibit cancer cell proliferation, but the mechanisms by which telmisartan affects various cancers remain unknown. In this study, we evaluated the effects of telmisartan on human CCA cells and to assess the expression of microRNAs (miRNAs). We studied the effects of telmisartan on CCA cells using two cell lines, HuCCT-1 and TFK-1. In our experiments, telmisartan inhibited the proliferation of HuCCT-1 and TFK-1 cells. Additionally, telmisartan induced G0/G1 cell cycle arrest via blockade of the G0 to G1 cell cycle transition. Notably, telmisartan did not induce apoptosis in HuCCT-1 cells. This blockade was accompanied by a strong decrease in cell cycle-related protein, especially G1 cyclin, cyclin D1, and its catalytic subumits, Cdk4 and Cdk6. Telmisartan reduced the phosphorylation of EGFR (p-EGFR) and TIMP-1 by using p-RTK and angiogenesis array. Furthermore, miRNA expression was markedly altered by telmisartan in HuCCT-1. Telmisartan inhibits tumor growth in CCA xenograft model in vivo. In conclusion, telmisartan was shown to inhibit human CCA cell proliferation by inducing cell cycle arrest.
Collapse
Affiliation(s)
- Eri Samukawa
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Shintaro Fujihara
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Kyoko Oura
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hisakazu Iwama
- Life Science Research Center, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Yoshimi Yamana
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Tomoko Tadokoro
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Taiga Chiyo
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Kiyoyuki Kobayashi
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Mai Nakahara
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hirohito Mori
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Keiichi Okano
- Gastroenterological Surgery, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Yasuyuki Suzuki
- Gastroenterological Surgery, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Takashi Himoto
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| |
Collapse
|
33
|
Oura K, Tadokoro T, Fujihara S, Morishita A, Chiyo T, Samukawa E, Yamana Y, Fujita K, Sakamoto T, Nomura T, Yoneyama H, Kobara H, Mori H, Iwama H, Okano K, Suzuki Y, Masaki T. Telmisartan inhibits hepatocellular carcinoma cell proliferation in vitro by inducing cell cycle arrest. Oncol Rep 2017; 38:2825-2835. [PMID: 29048654 PMCID: PMC5780034 DOI: 10.3892/or.2017.5977] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and the third leading cause of cancer-related death. Telmisartan, a widely used antihypertensive drug, is an angiotensin II type 1 (AT1) receptor blocker (ARB) that might inhibit cancer cell proliferation, but the mechanisms through which telmisartan affects various cancers remain unknown. The aim of the present study was to evaluate the effects of telmisartan on human HCC and to assess the expression of microRNAs (miRNAs). We studied the effects of telmisartan on HCC cells using the HLF, HLE, HepG2, HuH-7 and PLC/PRF/5 cell lines. In our experiments, telmisartan inhibited the proliferation of HLF, HLE and HepG2 cells, which represent poorly differentiated types of HCC cells. However, HuH-7 and PLC/PRF/5 cells, which represent well-differentiated types of HCC cells, were not sensitive to telmisartan. Telmisartan induced G0/G1 cell cycle arrest of HLF cells by inhibiting the G0-to-G1 cell cycle transition. This blockade was accompanied by a marked decrease in the levels of cyclin D1, cyclin E and other cell cycle-related proteins. Notably, the activity of the AMP-activated protein kinase (AMPK) pathway was increased, and the mammalian target of rapamycin (mTOR) pathway was inhibited by telmisartan treatment. Additionally, telmisartan increased the level of caspase-cleaved cytokeratin 18 (cCK18), partially contributed to the induction of apoptosis in HLF cells and reduced the phosphorylation of ErbB3 in HLF cells. Furthermore, miRNA expression was markedly altered by telmisartan in vitro. In conclusion, telmisartan inhibits human HCC cell proliferation by inducing cell cycle arrest.
Collapse
Affiliation(s)
- Kyoko Oura
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Tomoko Tadokoro
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Shintaro Fujihara
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Taiga Chiyo
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Eri Samukawa
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Yoshimi Yamana
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Koji Fujita
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Teppei Sakamoto
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Takako Nomura
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Hirohito Yoneyama
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Hirohito Mori
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Hisakazu Iwama
- Life Science Research Center, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Keiichi Okano
- Gastroenterological Surgery, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Yasuyuki Suzuki
- Gastroenterological Surgery, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Kagawa 761-0793, Japan
| |
Collapse
|
34
|
Nenicu A, Gu Y, Körbel C, Menger MD, Laschke MW. Combination therapy with telmisartan and parecoxib induces regression of endometriotic lesions. Br J Pharmacol 2017; 174:2623-2635. [PMID: 28548231 DOI: 10.1111/bph.13874] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/24/2017] [Accepted: 05/18/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Telmisartan suppresses the development of endometriotic lesions. However, the drug also up-regulates the expression of COX-2, which has been suggested to promote the progression of endometriosis. Accordingly, in the present study we analysed whether a combination therapy with telmisartan and a COX-2 inhibitor may be more effective in the treatment of endometriotic lesions than the application of telmisartan alone. EXPERIMENTAL APPROACH Endometriotic lesions were induced in the peritoneal cavity of C57BL/6 mice, which were treated daily with an i.p. injection of telmisartan (10 mg·kg-1 ), parecoxib (5 mg·kg-1 ), a combination of telmisartan and parecoxib or vehicle. Therapeutic effects on lesion survival, growth, vascularization, innervation and protein expression were studied over 4 weeks by high-resolution ultrasound imaging as well as immunohistochemical and Western blot analyses. KEY RESULTS Telmisartan-treated lesions exhibited a significantly reduced lesion volume when compared with vehicle-treated controls and parecoxib-treated lesions. This inhibitory effect of telmisartan was even more pronounced when it was used in combination with parecoxib. The combination therapy resulted in a reduced microvessel density as well as lower numbers of proliferating Ki67-positive cells and higher numbers of apoptotic cleaved caspase-3-positive stromal cells within the lesions. This was associated with a lower expression of COX-2, MMP-9 and p-Akt/Akt when compared with controls. The application of the two drugs further inhibited the ingrowth of nerve fibres into the lesions. CONCLUSIONS AND IMPLICATIONS Combination therapy with telmisartan and a COX-2 inhibitor represents a novel, effective pharmacological strategy for the treatment of endometriosis.
Collapse
Affiliation(s)
- Anca Nenicu
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Yuan Gu
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
35
|
LY333531, a PKCβ inhibitor, attenuates glomerular endothelial cell apoptosis in the early stage of mouse diabetic nephropathy via down-regulating swiprosin-1. Acta Pharmacol Sin 2017; 38:1009-1023. [PMID: 28414198 DOI: 10.1038/aps.2016.172] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/28/2016] [Indexed: 12/16/2022]
Abstract
Glomerular endothelial cell (GEC) injury plays an important role in the early stage of diabetic nephropathy (DN). Previous studies show that a PKCβ inhibitor is effective for treating DN. In the current study we further explored the effects and molecular mechanisms of PKCβ inhibitors on GEC apoptosis in DN in streptozotocin-induced diabetic mice in vivo and high glucose- or PMA-treated human renal glomerular endothelial cells (HRGECs) in vitro. In the diabetic mice, hyperglycemia caused aggravated nephropathy and GEC apoptosis accompanied by significantly increased expression of swiprosin-1, a potentally pro-apoptotic protein. Administration of LY333531 (1 mg·kg-1·d-1 for 8 weeks) significantly attenuated both GEC apoptosis and swiprosin-1 upregulation in the diabetic mice. Similar results were observed in high glucose- or PMA-treated HRGECs in vitro. The pro-apoptotic role of swiprosin-1 was further examined using HRGECs treated with lentivirus mediating RNA interference or over-expression and swiprosin-1-knockout mice. Over-expression of swiprosin-1 in HRGECs resulted in increases in apoptosis and in caspase-9, caspase-3 and Bax expression. In contrast, knockdown of swiprosin-1 attenuated high glucose- or PMA-induced HRGECs apoptosis. Furthermore, over-expression of swiprosin-1 promoted interaction between swiprosin-1 and caspase-9 and increased the formation of apoptosomes. In diabetic swiprosin-1-/- mice, the kidney/body weight, urinary albumin, glomerular hypertrophy, mitochondrial apoptotic-associated proteins and GEC apoptosis were significantly attenuated as compared with those in diabetic swiprosin-1+/+ mice. These results demonstrate that swiprosin-1 is up-regulated by PKCβ in the early stage of DN, and that PKCβ facilitates GEC apoptosis through the mitochondrial-dependent pathway.
Collapse
|
36
|
Pringle KG, Delforce SJ, Wang Y, Ashton KA, Proietto A, Otton G, Blackwell CC, Scott RJ, Lumbers ER. Renin-angiotensin system gene polymorphisms and endometrial cancer. Endocr Connect 2016; 5:128-35. [PMID: 27068935 PMCID: PMC5002951 DOI: 10.1530/ec-15-0112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/11/2016] [Indexed: 12/11/2022]
Abstract
Endometrial cancer (EC) is the most common gynaecological malignancy and its incidence is increasing. Dysregulation of the endometrial renin-angiotensin system (RAS) could predispose to EC; therefore, we studied the prevalence of RAS single nucleotide polymorphisms (SNPs) in Australian women with EC. SNPs assessed were AGT M235T (rs699); AGTR1 A1166C (rs5186); ACE A240T and T93C (rs4291, rs4292) and ATP6AP2 (rs2968915). They were identified using TaqMan SNP Genotyping Assays. The C allele of the AGTR1 SNP (rs5186) was more prevalent in women with EC (odds ratio (OR) 1.7, 95% confidence interval (CI) (1.2-2.3), P=0.002). The CC genotype of this SNP is associated with upregulation of the angiotensin II type 1 receptor (AGTR1). The G allele of AGT rs699, which is associated with higher angiotensinogen (AGT) levels, was less prevalent in women with EC (OR 0.54, 95% CI (0.39-0.74), P<0.001) compared with controls. AGT and AGT formed by removal of angiotensin I (des(Ang I)AGT) are both anti-angiogenic. In women with EC who had had hormone replacement therapy (HRT), the prevalence of the AGTR1 SNP (rs5186) and the ACE SNPs (rs4291 and rs4292) was greater than in women who had no record of HRT; SNP rs4291 is associated with increased plasma ACE activity. These data suggest there is an interaction between genotype, oestrogen replacement therapy and EC. In conclusion, the prevalence of two SNPs that enhance RAS activity was different in women with EC compared with healthy controls. These genetic factors may interact with obesity and hyperoestrogenism, predisposing ageing, obese women to EC.
Collapse
Affiliation(s)
- Kirsty G Pringle
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia
| | - Sarah J Delforce
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia
| | - Yu Wang
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia
| | - Katie A Ashton
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia
| | - Anthony Proietto
- Hunter Centre for Gynaecological CancerJohn Hunter Hospital, Newcastle, New South Wales, Australia
| | - Geoffrey Otton
- Hunter Centre for Gynaecological CancerJohn Hunter Hospital, Newcastle, New South Wales, Australia
| | - C Caroline Blackwell
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia Division of Molecular MedicinePathology North, Newcastle, New South Wales, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and PharmacyUniversity of Newcastle, Callaghan, New South Wales, Australia
Hunter Medical Research InstituteNew Lambton, Newcastle, New South Wales, Australia
| |
Collapse
|
37
|
Increase of human prostate cancer cell (DU145) apoptosis by telmisartan through PPAR-delta pathway. Eur J Pharmacol 2016; 775:35-42. [PMID: 26852954 DOI: 10.1016/j.ejphar.2016.02.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 12/23/2022]
Abstract
The effect of telmisartan on prostate cancer DU145 cell survival and the underlying mechanism of apoptosis involving peroxisome proliferator-activated receptor (PPAR) pathway were investigated. Cultured DU145 cells were treated pharmacologically with telmisartan and GSK0660 (a PPAR-delta antagonist); or by RNA interference with siRNA of PPAR-delta. The treatment effects on cell survival were evaluated with cell viability assay, life and dead cell staining and flow cytometry. Western blot analysis for PPAR-delta protein expression was also performed. The results showed that telmisartan (0-80 µm) dose-dependently reduced DU145 cell survival. Flow cytometry demonstrated cancer cell cycle arrest with increase of sub-G1 phase. GSK0660 partially but significantly restored the telmisartan-treated cell viability. Similarly, siRNA of PPAR-delta significantly reversed the telmisartan-induced apoptosis. Western blot showed that telmisartan significantly increased DU145 cell PPAR-delta protein expression. Co-incubation with siRNA of PPAR-delta inhibited the telmisartan effect of PPAR-delta up-regulation. In conclusion, telmisartan induces prostate cancer DU145 cells apoptosis through the up-regulation of PPAR-delta protein expression. Pharmacological inhibition or genetic silencing of PPAR-delta activity can both reverse the telmisartan-induced apoptotic effect. Thus the PPAR-delta pathway might be a potential target for the treatment of prostate cancer.
Collapse
|
38
|
Pu Z, Zhu M, Kong F. Telmisartan prevents proliferation and promotes apoptosis of human ovarian cancer cells through upregulating PPARγ and downregulating MMP‑9 expression. Mol Med Rep 2015; 13:555-9. [PMID: 26548340 DOI: 10.3892/mmr.2015.4512] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 09/25/2015] [Indexed: 11/06/2022] Open
Abstract
The mortality rate of ovarian cancer is the highest of all gynecological malignancies. Telmisartan is a commonly used clinical angiotensin receptor blocker, which has antihypertensive, anti‑inflammatory and antithrombotic effects. In the present study, it was investigated whether telmisartan could exert anticancer effects on ovarian cancer cells through upregulating peroxisome proliferator‑activated receptor γ (PPARγ) and downregulating matrix metalloproteinase‑9 (MMP‑9) expression. A 3.3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was conducted to analyze the proliferation of HEY cells. A Caspase‑3 Activity Assay kit and an Annexin V‑fluorescein isothiocyanate/propidium iodide kit were used to analyze the apoptosis of HEY cells. In addition, a gelatin zymography assay and reverse trancription‑quantitative polymerase chain reaction were included to analyze the expression of PPARγ and MMP‑9 in HEY cells. The data showed that telmisartan could significantly decrease cell viability and induce the apoptosis of HEY cells in a time‑ and dose‑dependent manner. Furthermore, telmisartan could also dose‑dependently increase the expression of PPARγ and decrease the expression of MMP‑9 in HEY cells. In addition, downregulation of the expression of PPARγ by small interfering (si)RNA could reduce the effect of telmisartan on ovarian cancer cells and increase the expression of MMP‑9. In conclusion, the results indicated that telmisartan prevents proliferation and promotes apoptosis of human ovarian cancer cells by upregulating PPARγ and downregulating MMP‑9 expression.
Collapse
Affiliation(s)
- Zhichen Pu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Min Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Fandou Kong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
39
|
Te Riet L, van Esch JHM, Roks AJM, van den Meiracker AH, Danser AHJ. Hypertension: renin-angiotensin-aldosterone system alterations. Circ Res 2015; 116:960-75. [PMID: 25767283 DOI: 10.1161/circresaha.116.303587] [Citation(s) in RCA: 508] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Blockers of the renin-angiotensin-aldosterone system (RAAS), that is, renin inhibitors, angiotensin (Ang)-converting enzyme (ACE) inhibitors, Ang II type 1 receptor antagonists, and mineralocorticoid receptor antagonists, are a cornerstone in the treatment of hypertension. How exactly they exert their effect, in particular in patients with low circulating RAAS activity, also taking into consideration the so-called Ang II/aldosterone escape that often occurs after initial blockade, is still incompletely understood. Multiple studies have tried to find parameters that predict the response to RAAS blockade, allowing a personalized treatment approach. Consequently, the question should now be answered on what basis (eg, sex, ethnicity, age, salt intake, baseline renin, ACE or aldosterone, and genetic variance) a RAAS blocker can be chosen to treat an individual patient. Are all blockers equal? Does optimal blockade imply maximum RAAS blockade, for example, by combining ≥2 RAAS blockers or by simply increasing the dose of 1 blocker? Exciting recent investigations reveal a range of unanticipated extrarenal effects of aldosterone, as well as a detailed insight in the genetic causes of primary aldosteronism, and mineralocorticoid receptor blockers have now become an important treatment option for resistant hypertension. Finally, apart from the deleterious ACE-Ang II-Ang II type 1 receptor arm, animal studies support the existence of protective aminopeptidase A-Ang III-Ang II type 2 receptor and ACE2-Ang-(1 to 7)-Mas receptor arms, paving the way for multiple new treatment options. This review provides an update about all these aspects, critically discussing the many controversies and allowing the reader to obtain a full understanding of what we currently know about RAAS alterations in hypertension.
Collapse
Affiliation(s)
- Luuk Te Riet
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Joep H M van Esch
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Anton J M Roks
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Anton H van den Meiracker
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - A H Jan Danser
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Izzedine H, Derosa L, Le Teuff G, Albiges L, Escudier B. Hypertension and angiotensin system inhibitors: impact on outcome in sunitinib-treated patients for metastatic renal cell carcinoma. Ann Oncol 2015; 26:1128-1133. [PMID: 25795198 DOI: 10.1093/annonc/mdv147] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/12/2015] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND To examine the association between hypertension (HTN), angiotensin system inhibitors (ASI) use and survival outcomes in patients with metastatic renal cell carcinoma (mRCC) treated with sunitinib (SU). METHODS We retrospectively reviewed all patients with mRCC who received SU as first-line treatment in Gustave Roussy from April 2004 to November 2013. The HTN (either pre-existing or secondary to SU), use of ASI (either before or during SU) were analysed. Overall survival (OS) and progression-free survival (PFS) of different exposures were compared with log-rank test. The associations between exposures and survival outcomes were estimated with hazard ratios (HRs) and 95% confidence interval (CI) through a multivariable Cox model adjusted for age, gender, International mRCC Database Consortium risk group and histology. RESULTS Among 213 patients with a 3.6-year median follow-up, 134 were hypertensive and 105 were ASI users with a significant association between the two exposures (P < 0.0001). Hypertensive patients have longer OS (median: 41.6 versus 16.4 months, P < 0.0001) and longer PFS (median: 12.9 versus 5.6 months, P < 0.0001) than non-hypertensive patients (n = 79). ASI users (n = 105) had more HTN_PRE compared with those (n = 108) who did not (65% versus 19%, P < 0.001). Multivariable analysis showed that hypertensive patients were significantly associated with OS (P = 0.05) and marginally with PFS (P = 0.06) while ASI intake was significantly associated with better OS [HR = 0.40; 95% CI (0.24-0.66), P < 0.001] and PFS [HR = 0.55 (0.35-0.86), P = 0.009]. The latter remain statistically significantly associated after controlling for the number of metastases. There is no difference on outcome between patients who receive ASI before starting SU and those who received ASI during SU treatment. CONCLUSION Concomitant use of ASI may significantly improve OS and PFS in mRCC patients receiving SU. HTN is marginally associated with the outcome in these patients.
Collapse
Affiliation(s)
- H Izzedine
- Department of Nephrology, Monceau Park International Clinic, Paris.
| | - L Derosa
- Department of Medical Oncology, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Santa Chiara Hospital, Pisa, Italy
| | - G Le Teuff
- Department of Biostatistics and Epidemiology, Gustave Roussy, Villejuif, France
| | - L Albiges
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - B Escudier
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
41
|
Abstract
Recently controversial data emerged regarding the cancer inducing activity of angiotensin-receptor blockers. There may be several reasons which may explain the controversial data published in the scientific literature including wrong trial design or misinterpretation of data. Considering the large number of patients receiving treatment for hypertension, it is essential to have a clear view of the cancer-related safety of these drugs. This paper tries to give an overview on this issue based on data available in the literature. According to our present knowledge, angiotensin-receptor blockers exert more likely anticancer activity rather than carcinogenesis inducing effect. In fact, some oncologic trials point to this direction, because angiotensin-receptor blockers are suggested as co-treatment to chemotherapy in cases of pancreatic, oesophageal and gastric cancers.
Collapse
Affiliation(s)
- András Telekes
- Bajcsy-Zsilinszky Kórház Onkológiai Osztály Budapest Maglódi út 89-91. 1106
| | - István Kiss
- Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika, Geriátriai Tanszéki Csoport Budapest Szent Imre Egyetemi Oktatókórház Nephrologia-Hypertonia Profil és Aktív Geriátriai Részleg Budapest B. Braun Avitum Zrt. Dialízis Hálózat 1. Sz. Dialízisközpont Budapest
| |
Collapse
|
42
|
Ren P, Zhang Y, Huang Y, Yang Y, Jiang M. Functions of Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) in Gynecologic Disorders. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2015; 9:43-9. [PMID: 25987855 PMCID: PMC4412418 DOI: 10.4137/cmo.s23527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 12/24/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of a class of nuclear hormone receptors intimately involved in the regulation of expression of myriad genes that regulate energy metabolism, cell differentiation, apoptosis, and inflammation. Although originally discovered as a pivotal regulator of adipocyte differentiation, the roles that PPARγ plays in gynecological disorders are still unknown. There are a number of studies on the functions of PPARγ and its agonists in gynecological disorders. In this mini-review, we provide a brief summary of the advances in recent years.
Collapse
Affiliation(s)
- Ping Ren
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China ; Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yuquan Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yan Huang
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China ; Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yingli Yang
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China ; Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ming Jiang
- Laboratory of Nuclear Receptors and Cancer Research, Basic Medical Research Center, Nantong University School of Medicine, Nantong, Jiangsu, China
| |
Collapse
|