1
|
Kalupahana NS, Moustaid-Moussa N. Beyond blood pressure, fluid and electrolyte homeostasis - Role of the renin angiotensin aldosterone system in the interplay between metabolic diseases and breast cancer. Acta Physiol (Oxf) 2024; 240:e14164. [PMID: 38770946 DOI: 10.1111/apha.14164] [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: 01/16/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The classical renin angiotensin aldosterone system (RAAS), as well as the recently described counter-regulatory or non-canonical RAAS have been well characterized for their role in cardiovascular homeostasis. Moreover, extensive research has been conducted over the past decades on both paracrine and the endocrine roles of local RAAS in various metabolic regulations and in chronic diseases. Clinical evidence from patients on RAAS blockers as well as pre-clinical studies using rodent models of genetic manipulations of RAAS genes documented that this system may play important roles in the interplay between metabolic diseases and cancer, namely breast cancer. Some of these studies suggest potential therapeutic applications and repurposing of RAAS inhibitors for these diseases. In this review, we discuss the mechanisms by which RAAS is involved in the pathogenesis of metabolic diseases such as obesity and type-2 diabetes as well as the role of this system in the initiation, expansion and/or progression of breast cancer, especially in the context of metabolic diseases.
Collapse
Affiliation(s)
- Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
2
|
Fatehi R, Nouraei M, Panahiyan M, Rashedinia M, Firouzabadi N. Modulation of ACE2/Ang1-7/Mas and ACE/AngII/AT1 axes affects anticancer properties of sertraline in MCF-7 breast cancer cells. Biochem Biophys Rep 2024; 38:101738. [PMID: 38831897 PMCID: PMC11145238 DOI: 10.1016/j.bbrep.2024.101738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
The renin-angiotensin system (RAS) is best known for playing a major role in maintaining the physiology of the cardiovascular system. Dysregulation of the RAS pathway has been proposed as a link to some malignancies and contributes to cancer metastasis. Breast cancer is considered as one of the leading causes of cancer death in women and its prevention remains yet a challenge. Elements of RAS are expressed in both normal breast tissue and cancerous cells, signifying the essential role of RAS in breast cancer pathology. Sertraline, a widely used antidepressant, has shown anti-proliferative properties on a variety of malignancies. This study aimed to investigate the effect of sertraline and its combination with agonists and antagonists of RAS (A779, Ang 1-7 and losartan) on viability of MCF-7 cells along with their effect on apoptosis and distribution of cell cycle. Our results indicated that sertraline, losartan and Ang 1-7 significantly decreased cell viability, induced apoptosis and cell cycle arrest. A779 blunted the effect of sertraline on cell viability, ROS generation and cell cycle arrest. Combination treatment of sertraline with losartan as well as Ang 1-7 caused a remarkable decline in cell viability. In conclusion, results of the present study support the anti-cancer properties of sertraline, losartan and Ang 1-7 via induction of apoptosis and cell cycle arrest.
Collapse
Affiliation(s)
- Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Nouraei
- Student Research Comittee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Panahiyan
- Student Research Comittee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Rashedinia
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Food and Supplements Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Sayour NV, Paál ÁM, Ameri P, Meijers WC, Minotti G, Andreadou I, Lombardo A, Camilli M, Drexel H, Grove EL, Dan GA, Ivanescu A, Semb AG, Savarese G, Dobrev D, Crea F, Kaski JC, de Boer RA, Ferdinandy P, Varga ZV. Heart failure pharmacotherapy and cancer: pathways and pre-clinical/clinical evidence. Eur Heart J 2024; 45:1224-1240. [PMID: 38441940 PMCID: PMC11023004 DOI: 10.1093/eurheartj/ehae105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 04/08/2024] Open
Abstract
Heart failure (HF) patients have a significantly higher risk of new-onset cancer and cancer-associated mortality, compared to subjects free of HF. While both the prevention and treatment of new-onset HF in patients with cancer have been investigated extensively, less is known about the prevention and treatment of new-onset cancer in patients with HF, and whether and how guideline-directed medical therapy (GDMT) for HF should be modified when cancer is diagnosed in HF patients. The purpose of this review is to elaborate and discuss the effects of pillar HF pharmacotherapies, as well as digoxin and diuretics on cancer, and to identify areas for further research and novel therapeutic strategies. To this end, in this review, (i) proposed effects and mechanisms of action of guideline-directed HF drugs on cancer derived from pre-clinical data will be described, (ii) the evidence from both observational studies and randomized controlled trials on the effects of guideline-directed medical therapy on cancer incidence and cancer-related outcomes, as synthetized by meta-analyses will be reviewed, and (iii) considerations for future pre-clinical and clinical investigations will be provided.
Collapse
Affiliation(s)
- Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Ágnes M Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Italian IRCCS Cardiology Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Wouter C Meijers
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Giorgio Minotti
- University Campus Bio-Medico, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation & Treatment (VIVIT), Carinagasse 47, A-6800 Feldkirch, Austria
| | - Erik Lerkevang Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Gheorghe Andrei Dan
- Carol Davila University of Medicine and Pharmacy, Colentina University Hospital, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Andreea Ivanescu
- Carol Davila University of Medicine and Pharmacy, Colentina University Hospital, Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Anne Grete Semb
- Division of Research and Innovation, REMEDY-Centre for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway
| | - Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular and Neuro Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Juan-Carlos Kaski
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Rudolf A de Boer
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Üllői út 26, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| |
Collapse
|
4
|
Silva JR, Souza-Fabjan JMG, Bento TFM, Silva RC, Moura CRF, Bartlewski PM, Batista RITP. The effects of heat stress on intrauterine development, reproductive function, and ovarian gene expression of F1 female mice as well as gene expression of F2 embryos†. Biol Reprod 2024; 110:33-47. [PMID: 37812452 DOI: 10.1093/biolre/ioad133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/30/2023] [Accepted: 10/01/2023] [Indexed: 10/10/2023] Open
Abstract
Exposure to heat stress (HS) in utero was postulated to trigger an adaptive molecular response that can be transmitted to the next generation. Hence, this study assessed the impact of HS exposure at different stages of the gestational period of mice on the female F1 population and their offspring. Heat stress exposure (41°C and 65% relative humidity-RH) occurred during the first half (FP), the second half (SP), or the entire pregnancy (TP). A control group (C) was maintained in normothermic conditions (25°C, 45% RH) throughout the experiment. Heat stress had a significant negative effect on intrauterine development, mainly when HS exposure occurred in the first half of pregnancy (FP and TP groups). Postnatal growth of FP and TP mice was hindered until 4 weeks of age. The total number of follicles per ovary did not vary (P > 0.05) between the control and HS-exposed groups. Mean numbers of primordial follicles were lower (P < 0.05) in the sexually mature FP than those in SP and TP F1 females. However, the mean number of viable embryos after superovulation was lower (P < 0.05) in TP compared with C group. The expression of genes associated with physiological and cellular response to HS, autophagy, and apoptosis was significantly affected in the ovarian tissue of F1 females and F2 in vivo-derived blastocysts in all HS-exposed groups. In conclusion, exposure to HS during pregnancy compromised somatic development and reproductive parameters as well as altered gene expression profile that was then transmitted to the next generation of mice.
Collapse
Affiliation(s)
- José R Silva
- Programa de Pós-Graduação em Ciência e Biotecnologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Joanna M G Souza-Fabjan
- Programa de Pós-Graduação em Ciência e Biotecnologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
- Faculdade de Veterinária, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Tays F M Bento
- Programa de Pós-Graduação em Ciência e Biotecnologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Robson Campos Silva
- Departamento de Ciências Básicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Cristiane R F Moura
- Departamento de Ciências Básicas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil
| | - Pawel M Bartlewski
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Ribrio I T P Batista
- Programa de Pós-Graduação em Ciência e Biotecnologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
- Faculdade de Veterinária, Universidade Federal Fluminense, Niterói, RJ, Brazil
| |
Collapse
|
5
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
6
|
Hwang HJ, Lee TG. Impact on clinical outcomes of renin-angiotensin system inhibitors against doxorubicin-related toxicity in patients with breast cancer and hypertension: A nationwide cohort study in South Korea. PLoS One 2023; 18:e0294649. [PMID: 37983233 PMCID: PMC10659172 DOI: 10.1371/journal.pone.0294649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Although doxorubicin (DOX) is a commonly used potent chemotherapeutic agent in patients with breast cancer, its cardiotoxic effect is a concern, particularly in patients with hypertension. Antihypertensive renin-angiotensin system (RAS) inhibitors may potentially play a role in preventing overt heart failure (HF) due to DOX toxicity. This study aimed to evaluate whether the use of RAS inhibitors improves clinical outcomes in patients with hypertension and breast cancer undergoing DOX-containing chemotherapy. METHODS A total of 54,344 female patients who were first diagnosed with breast cancer and initiated into DOX therapy between 2008 and 2015 were recruited from a nationwide Korean cohort. Patients were divided into two groups: with and without hypertension (HT, n = 10,789; non-HT, n = 43,555), and the RAS inhibitor group (n = 1,728) was sub-classified from the HT group. Two propensity score-matched cohorts were constructed to compare the clinical outcomes between non-HT and HT groups and between non-HT and RAS inhibitor groups. The primary outcome was the composite of HF and death. RESULTS After propensity score matching, the HT group had a higher risk for HF (adjusted hazard ratio [HR] = 1.30, 95% confidence intervals [95% CI] = 1.09-1.55) compared to the non-HT group, but there was no significant difference in primary outcome between the two groups. The RAS inhibitor group had a lower risk for primary outcome (adjusted HR = 0.78, 95% CI = 0.65-0.94) and death (adjusted HR = 0.81, 95% CI = 0.66-0.99) compared to the non-HT group. CONCLUSIONS Hypertension is a risk factor for HF in patients with breast cancer undergoing DOX chemotherapy. However, the RAS inhibitors used to treat hypertension may contribute to decreased mortality and improved clinical outcomes.
Collapse
Affiliation(s)
- Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Taek-Gu Lee
- Department of Surgery, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| |
Collapse
|
7
|
Okarvi SM. Preparation, Radiolabeling with 68Ga/ 177Lu and Preclinical Evaluation of Novel Angiotensin Peptide Analog: A New Class of Peptides for Breast Cancer Targeting. Pharmaceuticals (Basel) 2023; 16:1550. [PMID: 38004416 PMCID: PMC10675340 DOI: 10.3390/ph16111550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 11/26/2023] Open
Abstract
AIM Angiotensin II (AngII) is known to play a significant part in the development of breast cancer by triggering cell propagation of breast cancer, tumor angiogenesis, and regulating tumor invasion and cell migration. AngII arbitrates its action via two G-protein-coupled receptors, AngII type 1 receptor (AT1) and AngII type 2 receptor (AT2). Overexpression of the AT1 receptor in breast cancer cells seems to promote tumor growth and angiogenesis, thus targeting the AT1 receptor using AngII peptide would facilitate the detection of breast carcinoma. We developed an AngII peptide intending to assess whether the peptide of the renin-angiotensin system holds the ability to target AT1 receptor-overexpressing breast cancer in vivo. METHODS DOTA-coupled AngII peptide was synthesized by conventional solid-phase peptide synthesis according to Fmoc/HATU chemistry. 68Ga/177Lu labeled AngII peptide was evaluated for its binding with TNBC MDA-MB-231 and ER+ MCF7 cell lines. Pharmacokinetics was studied in healthy balb/c mice and in vivo tumor targeting in nude mice with MDA-MB-231 tumors xenografts. RESULTS DOTA-AngII peptide was labeled efficiently with 68Ga/177Lu with high labeling efficiency (≥90%). The stability of the radiopeptide in human plasma was found to be high. The AngII peptide analog showed nanomolar (<40 nM) AT1 receptor-specific binding affinity. The radioactivity internalized into MDA-MBA-231 and MCF7 cells were 14.97% and 11.75%, respectively. In vivo, biodistribution in balb/c mice exhibited efficient clearance of 68Ga/177Lu-DOTA-AngII peptide from the blood and elimination predominantly by the renal system due to its hydrophilic nature. A low amount of radioactivity was seen in the major organs including lungs, liver, stomach, spleen, and intestines (<3% ID/g) except the kidneys. A high renal-urinary excretion was observed for the radiotracer. In the TNBC MDA-MB-231 xenografts model, radiolabeled AngII peptide exhibited specific and effective AT1-based targeting in vivo. A rapid and efficient tumor targeting (2.18% ID/g at 45 min p.i.) together with fast renal excretion (~67% ID) highlights the tumor-targeting potential of the radiotracer. The AT1 receptor specificity of the radiotracer was validated by blocking assays. Furthermore, PET imaging provided sufficient visualization of MDA-MB-231 tumors in nude mice. CONCLUSION Our findings suggest that 68Ga/177Lu-DOTA-AngII peptide can be useful for the theranostic application of breast carcinomas. This study suggests the potential of this innovative class of peptides for rapid and efficient targeting of tumors and warrants further evaluation.
Collapse
Affiliation(s)
- Subhani M Okarvi
- Cyclotron and Radiopharmaceuticals Department, King Faisal Specialist Hospital and Research Centre, MBC-03, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| |
Collapse
|
8
|
Krishnarao K, Bruno KA, Di Florio DN, Edenfield BH, Whelan ER, Macomb LP, McGuire MM, Hill AR, Ray JC, Cornell LF, Tan W, Geiger XJ, Salomon GR, Douglass EJ, Fairweather D, Yamani MH. Upregulation of Endothelin-1 May Predict Chemotherapy-Induced Cardiotoxicity in Women with Breast Cancer. J Clin Med 2022; 11:jcm11123547. [PMID: 35743613 PMCID: PMC9224558 DOI: 10.3390/jcm11123547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 02/04/2023] Open
Abstract
As survival in breast cancer patients from newer therapies increases, concerns for chemotherapy-induced cardiotoxicity (CIC) have offset some of these benefits, manifesting as a decline in left ventricular ejection fraction (LVEF). Patients receiving anthracycline-based chemotherapy followed by trastuzumab are at risk for CIC. Previous research evaluating whether clinical biomarkers predict cardiotoxicity has been inconsistent. Recently, angiotensin II type 1 receptor (ATR1) and endothelin 1 (ET1) have been shown to play a role in breast tumor growth. We evaluated ATR1 and ET1 expression in breast cancer tissue and its association with CIC. A total of 33 paraffin-embedded breast tissue specimens from women with breast cancer treated with anthracycline-based chemotherapy and trastuzumab were analyzed by immunohistochemistry (IHC) and qRT-PCR. We found that ET1 expression was increased in patients with an LVEF ≤ 50% (p = 0.032) with a lower LVEF correlating with higher ET1 expression (r = 0.377, p = 0.031). In patients with a change in LVEF of greater than 10%, greater ET1 expression was noted compared to those without a change in LVEF (p = 0.017). Increased ET1 expression in breast tumor tissue is associated with reduced LVEF. Future studies need to examine whether ET1 may be a tissue biomarker that helps predict the risk of developing CIC in women with breast cancer.
Collapse
Affiliation(s)
- Krithika Krishnarao
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Department of Cardiovascular Medicine, Ochsner Health, New Orleans, LA 70121, USA
- Correspondence: ; Tel.: +1-504-842-9780
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Damian N. Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Emily R. Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Logan P. Macomb
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Molly M. McGuire
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Anneliese R. Hill
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Jordan C. Ray
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Lauren F. Cornell
- Department of Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (L.F.C.); (W.T.)
| | - Winston Tan
- Department of Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (L.F.C.); (W.T.)
| | | | - Gary R. Salomon
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Erika J. Douglass
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Mohamad H. Yamani
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| |
Collapse
|
9
|
Balasundaram A, Udhaya Kumar S, George Priya Doss C. A computational model revealing the immune-related hub genes and key pathways involved in rheumatoid arthritis (RA). ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 129:247-273. [PMID: 35305721 DOI: 10.1016/bs.apcsb.2021.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Rheumatoid arthritis (RA) has one of the highest disability rates among inflammatory joint disorders. However, the reason and possible molecular events are still unclear. There are various treatment options available, but no complete cure. To obtain early diagnosis and successful medication in RA, it is necessary to explore gene susceptibility and pathogenic factors. The main intend of our work is to explore the immune-related hub genes with similar functions that are differentially expressed in RA patients. Three datasets such as GSE21959, GSE55457, and GSE77298, were taken to analyze the differently expressed genes (DEGs) among 55 RA and 33 control samples. We obtained 331 upregulated and 275 downregulated DEGs from three Gene Expression Omnibus (GEO) datasets using the R package. Furthermore, a protein-protein interaction network was built for upregulated and downregulated DEGs using Cytoscape. Subsequently, MCODE analysis was performed and obtained the top two modules in each DEG's upregulated and downregulated protein-protein interactions (PPIs) network. CytoNCA and cytoHubba were performed and identified overlapping DEGs. In addition, we narrowed down DEGs by filtering with immune-related genes and identified DE-IRGs. Gene ontology (GO) and KEGG pathway analysis in upregulated and downregulated DEGs were executed with the DAVID platform. Our study obtained the nine most significant DE-IRGs in RA such as CXCR4, CDK1, BUB1, BIRC5, AGTR1, EGFR, EDNRB, KALRN, and GHSR. Among them, CXCR4, CDK1, BUB1, and BIRC5 are overexpressed in RA and may contribute to the pathophysiology of the disease. Similarly, AGTR1, EGFR, EDNRB, KALRN, and GHSR are all low expressed in RA and may have a contribution to pathogenesis. GO, KEGG functional enrichment, and GeneMANIA showed that the dysregulated process of DE-IRGs causes RA development and progression. These findings may be helpful in future studies in RA diagnosis and therapy.
Collapse
Affiliation(s)
- Ambritha Balasundaram
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, TN, India
| | - S Udhaya Kumar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, TN, India
| | - C George Priya Doss
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, TN, India.
| |
Collapse
|
10
|
Asgharzadeh F, Geraylow KR, Khazaei M, Nassiri M, Hassanian SM, Ferns GA, Avan A. Angiotensin-converting Enzyme Inhibitors and Angiotensin Receptor Blockers as Potential Therapeutic Options for Pancreatic Cancer. Curr Cancer Drug Targets 2022; 22:785-795. [PMID: 35585824 DOI: 10.2174/1568009622666220517104411] [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: 10/06/2021] [Revised: 01/27/2022] [Accepted: 02/15/2022] [Indexed: 11/22/2022]
Abstract
The renin-angiotensin system (RAS) has been reported to have a role in carcinogenesis, and therefore it may be of value as a potential therapeutic target in inhibiting tumor growth. It has been shown that inhibition of RAS via angiotensin I-converting enzyme inhibitors (ACEIs) and angiotensin II type-1 receptor (ARBs) inhibitors may have a protective effect against several malignancies. Here, we provide an overview of the potential value of the RAS pathway and targeting via ACE/ARB inhibitors in pancreatic cancer. Whilst the potential role of RAS as a target for the treatment of pancreatic cancer has been reported, the use of candesartan with gemcitabine failed to improve outcomes in pancreatic cancer. Another study of 1-3 years using ARB was found to reduce the risk of pancreatic cancer. In line with these trials, others have demonstrated that the ARBs in combination with gemcitabine might improve clinical outcomes in patients with advanced pancreatic cancer. Prospective trials are warranted to investigate this hypothesis.
Collapse
Affiliation(s)
- Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Moschetta-Pinheiro MG, Colombo J, de Godoy BLV, Balan JF, Nascimento BC, Zuccari DAPDC. Modulation of Epithelial Mesenchymal Transition after AGTR-1 Gene Edition by Crispr/Cas9 and Losartan Treatment in Mammary Tumor Cell Line: A Comparative Study between Human and Canine Species. Life (Basel) 2021; 11:life11121427. [PMID: 34947958 PMCID: PMC8705831 DOI: 10.3390/life11121427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most prevalent tumor type among women and female dogs. Tumor malignancy is characterized by the epithelial-to-mesenchymal transition (EMT) which leads to the metastasis formation. The inhibition of angiotensin II type I receptor (AGTR1) by an antagonist such as losartan can suppress angiogenesis, consequently contributing to the metastasis control. The aim of this study was to analyze the capacity of losartan and AGTR-1 gene edition to modulate the EMT process in triple negative/metastatic mammary tumor cells, compared to existing treatment protocols such as carboplatin. The cell lines CF41.Mg and MDA-MB-468, were cultured and treated with carboplatin, losartan, or submitted to AGTR-1 gene edition by CRISPR/Cas9. EMT markers and PARP-1 protein and gene expression were evaluated by immunofluorescence or immunocytochemistry and qRT-PCR, respectively. Cell migration capacity was also evaluated. For CF41.Mg and MDA-MB-468 cell lines, there was an increase in E-cadherin and a decrease in N-cadherin and PARP-1 protein and gene expression after treatment with carboplatin, losartan, both in combination and after AGTR-1 gene edition. There was a decrease in VEGF and PARP-1 protein and gene expression after AGTR-1 gene edition. Moreover, in both lines, reduction in invasion rate was observed after all treatments. Our data suggest that losartan and the gene edition of AGTR-1 by CRISPR/Cas9 were able to block the DNA repair and control the EMT process, such as carboplatin. The results in the canine species are unprecedented, as there are no data in the literature that demonstrate the action of losartan in this tumor type.
Collapse
Affiliation(s)
- Marina Gobbe Moschetta-Pinheiro
- PostGraduate Program in Health Sciences, Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil;
- Department of Health Sciences, Universidade Paulista (UNIP), Avenida Juscelino K. de Oliveira, s/n, São José do Rio Preto 15091-450, Brazil
- Correspondence: ; Fax: +55-17-3201-5885
| | - Jucimara Colombo
- Laboratório de Investigação Molecular no Câncer (LIMC), Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil; (J.C.); (J.F.B.); (B.C.N.); (D.A.P.d.C.Z.)
| | - Bianca Lara Venâncio de Godoy
- PostGraduate Program in Health Sciences, Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil;
- Laboratório de Investigação Molecular no Câncer (LIMC), Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil; (J.C.); (J.F.B.); (B.C.N.); (D.A.P.d.C.Z.)
| | - Julia Ferreira Balan
- Laboratório de Investigação Molecular no Câncer (LIMC), Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil; (J.C.); (J.F.B.); (B.C.N.); (D.A.P.d.C.Z.)
| | - Bianca Carlos Nascimento
- Laboratório de Investigação Molecular no Câncer (LIMC), Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil; (J.C.); (J.F.B.); (B.C.N.); (D.A.P.d.C.Z.)
| | - Debora Aparecida Pires de Campos Zuccari
- Laboratório de Investigação Molecular no Câncer (LIMC), Faculdade de Medicina de São José do Rio Preto (FAMERP), Avenida Brigadeiro Faria Lima, 5416, São José do Rio Preto 15090-000, Brazil; (J.C.); (J.F.B.); (B.C.N.); (D.A.P.d.C.Z.)
- PostGraduate Program in Genetics, Instituto de Biociências, Letras e Ciências Exatas (UNESP/IBILCE), Rua Cristovão Colombo, 2265, São José do Rio Preto 15054-000, Brazil
| |
Collapse
|
12
|
Xiong L, Wei Y, Zhou X, Dai P, Cai Y, Zhou X, Xu M, Zhao J, Tang H. AGTR1 Inhibits the Progression of Lung Adenocarcinoma. Cancer Manag Res 2021; 13:8535-8550. [PMID: 34803402 PMCID: PMC8598130 DOI: 10.2147/cmar.s335543] [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: 08/25/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose The occurrence and development of lung adenocarcinoma (LUAD) are related to many factors. Multiple researches showed that the renin-angiotensin system (RAS) plays an important role in lung cancer. This research mainly focuses on angiotensin II receptor 1 (AT1R) encoding gene AGTR1, an important part of the RAS. Methods We comprehensively evaluated the expression of AGTR1 in pan-cancer based on RNA sequencing data obtained from The Cancer Genome Atlas (TCGA). We explored the correlation of AGTR1 with clinicopathological features, prognosis and tumor microenvironment in LUAD. We also explored the mechanism through enrichment analysis and verified it with cell lines and tissue samples. Results We found that AGTR1 was less expressed in most tumors and related to prognosis based on the TCGA database. To further explore its mechanism, we mainly focused on LUAD. Combined with the verification results in the GEO database, AGTR1 was associated with a better prognosis in LUAD. High expression of AGTR1 was associated with less lymph node metastasis (P=0.007) and MET mutation (P=0.019). High expression of AGTR1 was related to the anti-tumor immune microenvironment with high infiltration of B cells, myeloid dendritic cells, monocytes, and low infiltration of myeloid-derived suppressor cells (all P<0.05). Enrichment analysis and in vitro verification results showed that AGTR1 was likely to play a role in LUAD through the PI3K/AKT3 pathway. Finally, we verified the above results through tissue samples and the construction of AGTR1 overexpressing cells. Conclusion AGTR1 inhibits the progression of lung adenocarcinoma through the PI3K/AKT3 pathway.
Collapse
Affiliation(s)
- Lecai Xiong
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Xiao Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Peng Dai
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Yi Cai
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Xuefeng Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jinping Zhao
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| |
Collapse
|
13
|
The C-terminal HSP90 inhibitor NCT-58 kills trastuzumab-resistant breast cancer stem-like cells. Cell Death Dis 2021; 7:354. [PMID: 34775489 PMCID: PMC8590693 DOI: 10.1038/s41420-021-00743-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022]
Abstract
N-terminal HSP90 inhibitors in development have had issues arising from heat shock response (HSR) induction and off-target effects. We sought to investigate the capacity of NCT-58, a rationally-synthesized C-terminal HSP90 inhibitor, to kill trastuzumab-resistant HER2-positive breast cancer stem-like cells. NCT-58 does not induce the HSR due to its targeting of the C-terminal region and elicits anti-tumor activity via the simultaneous downregulation of HER family members as well as inhibition of Akt phosphorylation. NCT-58 kills the rapidly proliferating bulk tumor cells as well as the breast cancer stem-like population, coinciding with significant reductions in stem/progenitor markers and pluripotent transcription factors. NCT-58 treatment suppressed growth and angiogenesis in a trastuzumab-resistant xenograft model, concomitant with downregulation of ICD-HER2 and HSF-1/HSP70/HSP90. These findings warrant further investigation of NCT-58 to address trastuzumab resistance in heterogeneous HER2-positive cancers.
Collapse
|
14
|
Lee JYL, Ekambaram P, Carleton NM, Hu D, Klei LR, Cai Z, Myers MI, Hubel NE, Covic L, Agnihotri S, Krappmann D, Bornancin F, Lee AV, Oesterreich S, McAllister-Lucas L, Lucas PC. MALT1 is a Targetable Driver of Epithelial-to-Mesenchymal Transition in Claudin-low, Triple-Negative Breast Cancer. Mol Cancer Res 2021; 20:373-386. [PMID: 34753803 DOI: 10.1158/1541-7786.mcr-21-0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/03/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022]
Abstract
MALT1 is the effector protein of the CARMA/Bcl10/MALT1 (CBM) signalosome, a multi-protein complex that drives pro-inflammatory signaling pathways downstream of a diverse set of receptors. While CBM activity is best known for its role in immune cells, emerging evidence suggests that it plays a key role in the pathogenesis of solid tumors, where it can be activated by selected G protein-coupled receptors (GPCRs). Here, we demonstrated that overexpression of GPCRs implicated in breast cancer pathogenesis, specifically the receptors for Angiotensin II and thrombin (AT1R and PAR1), drove a strong epithelial-to-mesenchymal transition (EMT) program in breast cancer cells that is characteristic of claudin-low, triple-negative breast cancer (TNBC). In concert, MALT1 was activated in these cells and contributed to the dramatic EMT phenotypic changes through regulation of master EMT transcription factors including Snail and ZEB1. Importantly, blocking MALT1 signaling, through either siRNA-mediated depletion of MALT1 protein or pharmacologic inhibition of its activity, was effective at partially reversing the molecular and phenotypic indicators of EMT. Treatment of mice with mepazine, a pharmacologic MALT1 inhibitor, reduced growth of PAR1+, MDA-MB-231 xenografts and had an even more dramatic effect in reducing the burden of metastatic disease. These findings highlight MALT1 as an attractive therapeutic target for claudin-low TNBCs harboring overexpression of one or more selected GPCRs. Implications: This study nominates a GPCR/MALT1 signaling axis as a pathway that can be pharmaceutically targeted to abrogate EMT and metastatic progression in TNBC, an aggressive form of breast cancer that currently lacks targeted therapies.
Collapse
Affiliation(s)
| | | | | | - Dong Hu
- Pathology, University of Pittsburgh
| | | | - Zongyou Cai
- Pathology, University of Pittsburgh School of Medicine
| | - Max I Myers
- Pathology, University of Pittsburgh School of Medicine
| | | | - Lidija Covic
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center
| | - Sameer Agnihotri
- Children's Hospital, Department of Neurological Surgery, University of Pittsburgh
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration - Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München
| | - Frederic Bornancin
- Autoimmunity Transplantation & Inflammation, Novartis Institutes for Biomedical Research
| | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, University of Pittsburgh
| | | | - Peter C Lucas
- Pathology and Pediatrics, University of Pittsburgh School of Medicine
| |
Collapse
|
15
|
Panza S, Malivindi R, Caruso A, Russo U, Giordano F, Győrffy B, Gelsomino L, De Amicis F, Barone I, Conforti FL, Giordano C, Bonofiglio D, Catalano S, Andò S. Novel Insights into the Antagonistic Effects of Losartan against Angiotensin II/AGTR1 Signaling in Glioblastoma Cells. Cancers (Basel) 2021; 13:cancers13184555. [PMID: 34572782 PMCID: PMC8469998 DOI: 10.3390/cancers13184555] [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: 08/09/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Patients with high-grade glioma (HGG) such as glioblastoma (GBM) who undergo surgical resection with adjuvant therapy have a mean overall survival of 14.6 months and 100% of recurrence. Thus, these disappointing outcomes in terms of glioblastoma life expectancy require seeking novel pharmacological tools, including drug repurposing. In the present study, we identify a novel molecular mechanism through which Losartan antagonizes Angiotensin II (Ang II)/Angiotensin II type I receptor (AGTR1) signaling, overexpressed in GBM cells. For instance, we demonstrate how Losartan drastically inhibits the stimulatory effects of Ang II on aromatase activity and consequently reduces local estrogen production, sustaining cancer progression. Thus, it is reasonable to repurpose Losartan as an adjuvant pharmacological tool to be implemented prospectively in the novel therapeutic strategies adopted in GBM patients. Abstract New avenues for glioblastoma therapy are required due to the limited mortality benefit of the current treatments. The renin-angiotensin system (RAS) exhibits local actions and works as a paracrine system in different tissues and tumors, including glioma. The glioblastoma cell lines U-87 MG and T98G overexpresses Angiotensin II (Ang II)/Angiotensin II type I receptor (AGTR1) signaling, which enhances in vitro and in vivo local estrogen production through a direct up-regulation of the aromatase gene promoters p I.f and p I.4. In addition, Ang II/AGTR1 signaling transactivates estrogen receptor-α in a ligand-independent manner through mitogen-activated protein kinase (MAPK) activation. The higher aromatase mRNA expression in patients with glioblastoma was associated with the worst survival prognostic, according to The Cancer Genome Atlas (TCGA). An intrinsic immunosuppressive glioblastoma tumor milieu has been previously documented. We demonstrate how Ang II treatment in glioblastoma cells increases programmed death-ligand 1 (PD-L1) expression reversed by combined exposure to Losartan (LOS) in vitro and in vivo. Our findings highlight how LOS, in addition, antagonizes the previously documented neoangiogenetic, profibrotic, and immunosuppressive effects of Ang II and drastically inhibits its stimulatory effects on local estrogen production, sustaining glioblastoma cell growth. Thus, Losartan may represent an adjuvant pharmacological tool to be repurposed prospectively for glioblastoma treatment.
Collapse
Affiliation(s)
- Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Umberto Russo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary;
- Cancer Biomarker Research Group, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy; (S.P.); (R.M.); (A.C.); (U.R.); (F.G.); (L.G.); (F.D.A.); (I.B.); (F.L.C.); (C.G.); (D.B.); (S.C.)
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
- Correspondence: ; Tel.: +39-0984-496201; Fax: +39-0984-496203
| |
Collapse
|
16
|
Qiao ZW, Jiang Y, Wang L, Wang L, Jiang J, Zhang JR, Mu P. LINC00852 promotes the proliferation and invasion of ovarian cancer cells by competitively binding with miR-140-3p to regulate AGTR1 expression. BMC Cancer 2021; 21:1004. [PMID: 34496800 PMCID: PMC8424870 DOI: 10.1186/s12885-021-08730-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 08/25/2021] [Indexed: 12/29/2022] Open
Abstract
Background Dysregulation of long non-coding RNAs (lncRNAs) has been identified in ovarian cancer. However, the expression and biological functions of LINC00852 in ovarian cancer are not understood. Methods The expressions of LINC00852, miR-140-3p and AGTR1 mRNA in ovarian cancer tissues and cells were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay. Gain- and loss-of-function assays were performed to explore the biological functions of LINC00852 and miR-140-3p in the progression of ovarian cancer in vitro. The bindings between LINC00852 and miR-140-3p were confirmed by luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay. Results We found that LINC00852 expression was significantly up-regulated in ovarian cancer tissues and cells, whereas miR-140-3p expression was significantly down-regulated in ovarian cancer tissues. Functionally, LINC00852 knockdown inhibited the viability, proliferation and invasion of ovarian cancer cells, and promoted the apoptosis of ovarian cancer cells. Further investigation showed that LINC00852 interacted with miR-140-3p, and miR-140-3p overexpression suppressed the viability, proliferation and invasion of ovarian cancer cells. In addition, miR-140-3p interacted with AGTR1 and negatively regulated its level in ovarian cancer cells. Mechanistically, we found that LINC00852 acted as a ceRNA of miR-140-3p to promote AGTR1 expression and activate MEK/ERK/STAT3 pathway. Finally, LINC00852 knockdown inhibited the growth and invasion ovarian cancer in vivo. Conclusion LINC00852/miR-140-3p/AGTR1 is an important pathway to promote the proliferation and invasion of ovarian cancer.
Collapse
Affiliation(s)
- Zhi-Wei Qiao
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China
| | - Ying Jiang
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China
| | - Ling Wang
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China
| | - Lei Wang
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China
| | - Jing Jiang
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China
| | - Jing-Ru Zhang
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China.
| | - Peng Mu
- Department of Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, No.44, Xiaoheyan Road, Shenyang, 110042, Liaoning Province, China.
| |
Collapse
|
17
|
Takiguchi T, Takahashi-Yanaga F, Ishikane S, Tetsuo F, Hosoda H, Arioka M, Kitazono T, Sasaguri T. Angiotensin II promotes primary tumor growth and metastasis formation of murine TNBC 4T1 cells through the fibroblasts around cancer cells. Eur J Pharmacol 2021; 909:174415. [PMID: 34375673 DOI: 10.1016/j.ejphar.2021.174415] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 10/20/2022]
Abstract
Angiotensin II (Ang II) reportedly facilitates primary tumor growth and distal hematogenous metastasis formation in various murine intravenous metastasis models. However, it is unclear whether Ang II accelerates the initial processes of metastasis formation that begins in primary tumors surrounded by tumor microenvironment. We examined the effects of Ang II on primary tumors and lung metastasis lesions using a murine spontaneous metastasis model, in which triple negative breast cancer 4T1 cells constitutively expressing luciferase (4T1-Luc cells) were injected into the mammary fat pad of BALB/c mice. Subcutaneous injection of Ang II significantly accelerated primary tumor growth and lung metastasis formation. Ang II increased the protein expression levels of c-Myc, cyclin D1, fibronectin, vimentin, αSMA and Snail, and the treatment with the Ang II type 1 receptor blocker valsartan significantly suppressed the Ang II-induced increases of fibronectin and vimentin. Valsartan also significantly reduced lung metastatic lesions. However, Ang II did not have significant effects on 4T1-Luc cells including the proliferation, migration, invasion, or the expressions of proteins related to cell proliferation and epithelial-to-mesenchymal transition. In contrast, when 4T1-Luc cells were co-cultured with dermal fibroblasts, Ang II significantly accelerated cell migration and increased the expressions of fibronectin, vimentin, αSMA and Snail in 4T1-Luc cells. And moreover, Ang II significantly increased the mRNA expression of IL-6 in fibroblasts co-cultured with 4T1-Luc cells. These results suggested that Ang II accelerates surrounding fibroblasts by soluble factors such as IL-6 to promote epithelial-to-mesenchymal transition, which result in the initiation of cancer metastasis.
Collapse
Affiliation(s)
- Tomohiro Takiguchi
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Shin Ishikane
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Fumi Tetsuo
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Masaki Arioka
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiyuki Sasaguri
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
18
|
Mehranfard D, Perez G, Rodriguez A, Ladna JM, Neagra CT, Goldstein B, Carroll T, Tran A, Trivedi M, Speth RC. Alterations in Gene Expression of Renin-Angiotensin System Components and Related Proteins in Colorectal Cancer. J Renin Angiotensin Aldosterone Syst 2021; 2021:9987115. [PMID: 34285715 PMCID: PMC8277508 DOI: 10.1155/2021/9987115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/13/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022] Open
Abstract
MATERIALS AND METHODS Quantitative expression of the RNA of these 17 genes in normal and cancerous tissues obtained using chip arrays from the public functional genomics data repository, Gene Expression Omnibus (GEO) application, was compared statistically. RESULTS Expression of four genes, AGT (angiotensinogen), ENPEP (aminopeptidase A) MME (neprilysin), and PREP (prolyl endopeptidase), was significantly upregulated in CRC specimens. Expression of REN (renin), THOP (thimet oligopeptidase), NLN (neurolysin), PRCP (prolyl carboxypeptidase), ANPEP (aminopeptidase N), and MAS1 (Mas receptor) was downregulated in CRC specimens. CONCLUSIONS Presuming gene expression parallel protein expression, these results suggest that increased production of the angiotensinogen precursor of angiotensin (ANG) peptides, with the reduction of the enzymes that metabolize it to ANG II, can lead to accumulation of angiotensinogen in CRC tissues. Downregulation of THOP, NLN, PRCP, and MAS1 gene expression, whose proteins contribute to the ACE2/ANG 1-7/Mas axis, suggests that reduced activity of this RAS branch could be permissive for oncogenicity. Components of the RAS may be potential therapeutic targets for treatment of CRC.
Collapse
Affiliation(s)
- Danial Mehranfard
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Gabriela Perez
- Department of Internal Medicine, Palmetto General Hospital, Hialeah, FL, USA
| | - Andres Rodriguez
- Department of Internal Medicine, University of Miami/Jackson Memorial Hospital, Miami, FL, USA
| | | | | | | | - Timothy Carroll
- College of Psychology, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Alice Tran
- Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Malav Trivedi
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Robert C. Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
19
|
Sanyal S, Alabraba E, Ibrahim H, Olaru A, Cameron I, Gomez D. ACE Inhibitor Therapy Does Not Influence the Survival Outcomes of Patients with Colorectal Liver Metastases Following Liver Resection. J Gastrointest Cancer 2021; 52:106-112. [PMID: 31853827 DOI: 10.1007/s12029-019-00350-6] [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: 11/28/2022]
Abstract
BACKGROUND Angiotensin-converting enzyme (ACE) inhibitors have been shown to possibly influence the survival outcomes in certain cancers. The aim of this study was to evaluate the impact of ACE inhibitors on the outcomes of patients undergoing liver resection for colorectal liver metastases (CRLM). The secondary aim was to determine whether ACE inhibitors influenced histopathological changes in CRLM. METHODS Patients treated with liver resection for CRLM over a 13-year period were identified from a prospectively maintained database. Data including demographics, primary tumour treatment, surgical data, histopathology analysis and clinical outcome were collated and analysed. RESULTS A total of 586 patients underwent primary hepatic resections for CRLM during this period including 100 patients on ACE inhibitors. The median follow-up period was 23 (range: 12-96) months, in which 267 patients developed recurrent disease and 131 patients died. Independent predictors of disease-free survival on multivariate analysis included synchronous presentation, neoadjuvant chemotherapy, major liver resection, tumour size and number, extent of hepatic steatosis, R0 resection and presence of perineural invasion. Poorer overall survival was associated with neoadjuvant treatment, major liver resection, presence of multiple metastases, perineural invasion and positive resection margins on multivariate analysis. ACE inhibitors did not influence the survival outcome or histological presentation in CRLM. CONCLUSION The use of ACE inhibitors did not affect the survival outcome or tumour biology in patients with CRLM following liver resection.
Collapse
Affiliation(s)
- Sudip Sanyal
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Derby Rd, Nottingham, NG7 2UH, UK
| | - Edward Alabraba
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Derby Rd, Nottingham, NG7 2UH, UK
| | - Hussain Ibrahim
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Derby Rd, Nottingham, NG7 2UH, UK
| | - Adina Olaru
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Derby Rd, Nottingham, NG7 2UH, UK
| | - Iain Cameron
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Derby Rd, Nottingham, NG7 2UH, UK
| | - Dhanny Gomez
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Derby Rd, Nottingham, NG7 2UH, UK. .,NIHR Nottingham Digestive Disease Biomedical Research Uni, University of Nottingham, Nottingham, UK.
| |
Collapse
|
20
|
Soheilifar MH, Masoudi-Khoram N, Madadi S, Nobari S, Maadi H, Keshmiri Neghab H, Amini R, Pishnamazi M. Angioregulatory microRNAs in breast cancer: Molecular mechanistic basis and implications for therapeutic strategies. J Adv Res 2021; 37:235-253. [PMID: 35499045 PMCID: PMC9039675 DOI: 10.1016/j.jare.2021.06.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/13/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer-associated angiogenesis is a fundamental process in tumor growth and metastasis. Angioregulatory miRNA–target gene interaction is not only involved in sprouting vessels of breast tumors but also, trans-differentiation of breast cancer cells to endothelial cells in a process termed vasculogenic mimicry. Successful targeting of tumor angiogenesis is still a missing link in the treatment of Breast cancer (BC) due to the low effectiveness of anti-angiogenic therapies in this cancer. Response to anti-angiogenic therapeutics are controlled by a miRNAs, so the identification of interaction networks of miRNAs–targets can be applicable in determining anti-angiogeneic therapy and new biomarkers in BC. Angioregulatory miRNAs in breast cancer cells and their microenvironment have therapeutic potential in cancer treatment.
Background Cancer-associated angiogenesis is a fundamental process in tumor growth and metastasis. A variety of signaling regulators and pathways contribute to establish neovascularization, among them as small endogenous non-coding RNAs, microRNAs (miRNAs) play prominent dual regulatory function in breast cancer (BC) angiogenesis. Aim of Review This review aims at describing the current state-of-the-art in BC angiogenesis-mediated by angioregulatory miRNAs, and an overview of miRNAs dysregulation association with the anti-angiogenic response in addition to potential clinical application of miRNAs-based therapeutics. Key Scientific Concepts of Review Angioregulatory miRNA–target gene interaction is not only involved in sprouting vessels of breast tumors but also, trans-differentiation of BC cells to endothelial cells (ECs) in a process termed vasculogenic mimicry. Using canonical and non-canonical angiogenesis pathways, the tumor cell employs the oncogenic characteristics such as miRNAs dysregulation to increase survival, proliferation, oxygen and nutrient supply, and treatment resistance. Angioregulatory miRNAs in BC cells and their microenvironment have therapeutic potential in cancer treatment. Although, miRNAs dysregulation can serve as tumor biomarker nevertheless, due to the association of miRNAs dysregulation with anti-angiogenic resistant phenotype, clinical benefits of anti-angiogenic therapy might be challenging in BC. Hence, unveiling the molecular mechanism underlying angioregulatory miRNAs sparked a booming interest in finding new treatment strategies such as miRNA-based therapies in BC.
Collapse
Affiliation(s)
- Mohammad Hasan Soheilifar
- Department of Medical Laser, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Corresponding authorsat: Yara Institute, Academic Center for Education, Culture and Research (ACECR), Enghelab St, Tehran 1315795613, Iran (Mohammad Hasan Soheilifar). University of Limerick, Limerick V94 T9PX, Ireland (Mahboubeh Pishnamazi).
| | - Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Soheil Madadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sima Nobari
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Maadi
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahboubeh Pishnamazi
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
- Corresponding authorsat: Yara Institute, Academic Center for Education, Culture and Research (ACECR), Enghelab St, Tehran 1315795613, Iran (Mohammad Hasan Soheilifar). University of Limerick, Limerick V94 T9PX, Ireland (Mahboubeh Pishnamazi).
| |
Collapse
|
21
|
Stoyell-Conti FF, Itty S, Abraham C, Rigatto K, West CA, Speth RC. 125I-Angiotensin 1-7 binds to a different site than angiotensin 1-7 in tissue membrane preparations. Endocrine 2021; 72:529-538. [PMID: 33415576 DOI: 10.1007/s12020-020-02572-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE To study the receptor for Angiotensin (Ang) 1-7 using a radioligand (125I-Ang 1-7)-binding assay. For more than a decade, Mas has been viewed as the receptor for Ang 1-7; however, Ang 1-7 binding has not been pharmacologically characterized in tissue membrane preparations. METHODS Radioligand-binding assays were carried out using tissue membrane preparations using radioiodinated Angiotensin 1-7 (125I-Ang 1-7) to characterize its binding site. Non-radioactive 127I-Ang 1-7 was used to test if the addition of an iodine to the tyrosine4 moiety of Ang 1-7 changes the ability of Ang 1-7 to competitively inhibit 125I-Ang 1-7 binding. RESULTS 125I-Ang 1-7 binds saturably, with moderately high affinity (10-20 nM) to a binding site in rat liver membranes that is displaceable by 127I-Ang 1-7 at nanomolar concentrations (IC50 = 62 nM) while Ang 1-7 displaces at micromolar concentrations (IC50 = 80 µM) at ~22 °C. This binding was also displaceable by inhibitors of metalloproteases at room temperature. This suggests that 125I-Ang 1-7 binds to MMPs and/or ADAMs as well as other liver membrane elements at ~ 22 °C. However, when 125I-Ang 1-7-binding assays were run at 0-4 °C, the same MMP inhibitors did not effectively compete for 125I-Ang 1-7. CONCLUSIONS The addition of an iodine molecule to the tyrosine in position 4 of Ang 1-7 drastically changes the binding characteristics of this peptide making it unsuitable for characterization of Ang 1-7 receptors.
Collapse
Affiliation(s)
- Filipe F Stoyell-Conti
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Sarin Itty
- Halmos College of Natural Science & Oceanography, Nova Southeastern University, Fort Lauderdale, FL, USA
- Kiran P. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Christy Abraham
- Halmos College of Natural Science & Oceanography, Nova Southeastern University, Fort Lauderdale, FL, USA
- Kiran P. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Katya Rigatto
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Laboratório de Fisiologia Translacional, Universidade Federal de Ciências da Saúde de Porto, Alegre, RS, Brazil
| | - Crystal A West
- Department of Biology, Appalachian State University, North Carolina Research Campus, Kannapolis, NC, USA
| | - Robert C Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, USA.
| |
Collapse
|
22
|
The Angiotensin II Receptor Blocker Losartan Sensitizes Human Liver Cancer Cells to Lenvatinib-Mediated Cytostatic and Angiostatic Effects. Cells 2021; 10:cells10030575. [PMID: 33807929 PMCID: PMC8001516 DOI: 10.3390/cells10030575] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
Molecular targeted therapy with lenvatinib is commonly offered to advanced hepatocellular carcinoma (HCC) patients, although it is often interrupted by adverse effects which require a reduction in the initial dose. Thus, an alternative lenvatinib-based therapy to compensate for dose reduction is anticipated. This study aimed to assess the effect of combination of low-dose of lenvatinib and the angiotensin-II (AT-II) receptor blocker losartan on human HCC cell growth. In vitro studies found that losartan suppressed the proliferation by inducing G1 arrest and caused apoptosis as indicated by the cleavage of caspase-3 in AT-II-stimulated HCC cell lines (Huh-7, HLE, and JHH-6). Losartan attenuated the AT-II-stimulated production of vascular endothelial growth factor-A (VEGF-A) and interleukin-8 and suppressed lenvatinib-mediated autocrine VEGF-A production in HCC cells. Moreover, it directly inhibited VEGF-mediated endothelial cell growth. Notably, the combination of lenvatinib and losartan augmented the cytostatic and angiostatic effects of the former at a low-dose, reaching those achieved with a conventional dose. Correspondingly, a HCC tumor xenograft assay showed that the oral administration of losartan combined with lenvatinib reduced the subcutaneous tumor burden and intratumor vascularization in BALB/c nude mice. These findings support that this regimen could be a viable option for patients intolerant to standard lenvatinib dosage.
Collapse
|
23
|
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: 3.3] [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
|
24
|
Lee HY, Son SW, Moeng S, Choi SY, Park JK. The Role of Noncoding RNAs in the Regulation of Anoikis and Anchorage-Independent Growth in Cancer. Int J Mol Sci 2021; 22:ijms22020627. [PMID: 33435156 PMCID: PMC7827914 DOI: 10.3390/ijms22020627] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is a global health concern, and the prognosis of patients with cancer is associated with metastasis. Multistep processes are involved in cancer metastasis. Accumulating evidence has shown that cancer cells acquire the capacity of anoikis resistance and anchorage-independent cell growth, which are critical prerequisite features of metastatic cancer cells. Multiple cellular factors and events, such as apoptosis, survival factors, cell cycle, EMT, stemness, autophagy, and integrins influence the anoikis resistance and anchorage-independent cell growth in cancer. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are dysregulated in cancer. They regulate cellular signaling pathways and events, eventually contributing to cancer aggressiveness. This review presents the role of miRNAs and lncRNAs in modulating anoikis resistance and anchorage-independent cell growth. We also discuss the feasibility of ncRNA-based therapy and the natural features of ncRNAs that need to be contemplated for more beneficial therapeutic strategies against cancer.
Collapse
|
25
|
Almutlaq M, Alamro AA, Alamri HS, Alghamdi AA, Barhoumi T. The Effect of Local Renin Angiotensin System in the Common Types of Cancer. Front Endocrinol (Lausanne) 2021; 12:736361. [PMID: 34539580 PMCID: PMC8446618 DOI: 10.3389/fendo.2021.736361] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
The Renin Angiotensin System (RAS) is a hormonal system that is responsible for blood pressure hemostasis and electrolyte balance. It is implicated in cancer hallmarks because it is expressed locally in almost all of the body's tissues. In this review, current knowledge on the effect of local RAS in the common types of cancer such as breast, lung, liver, prostate and skin cancer is summarised. The mechanisms by which RAS components could increase or decrease cancer activity are also discussed. In addition to the former, this review explores how the administration of AT1R blockers and ACE inhibitors drugs intervene with cancer therapy and contribute to the outcomes of cancer.
Collapse
Affiliation(s)
- Moudhi Almutlaq
- King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
- *Correspondence: Moudhi Almutlaq, ; Tlili Barhoumi,
| | - Abir Abdullah Alamro
- Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hassan S. Alamri
- King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Medical Research Core Facility and Platforms, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Amani Ahmed Alghamdi
- Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tlili Barhoumi
- King Abdullah International Medical Research Centre, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Medical Research Core Facility and Platforms, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- *Correspondence: Moudhi Almutlaq, ; Tlili Barhoumi,
| |
Collapse
|
26
|
Ma Y, Xia Z, Ye C, Lu C, Zhou S, Pan J, Liu C, Zhang J, Liu T, Hu T, Xie L, Wu G, Zhao Y. AGTR1 promotes lymph node metastasis in breast cancer by upregulating CXCR4/SDF-1α and inducing cell migration and invasion. Aging (Albany NY) 2020; 11:3969-3992. [PMID: 31219799 PMCID: PMC6628987 DOI: 10.18632/aging.102032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 06/13/2019] [Indexed: 12/21/2022]
Abstract
The angiotensin II type I receptor (AGTR1) has a strong influence on tumor growth, angiogenesis, inflammation and immunity. However, the role of AGTR1 on lymph node metastasis (LNM) in breast cancer, which correlates with tumor progression and patient survival, has not been examined. AGTR1 was highly expressed in lymph node-positive tumor tissues, which was confirmed by the Oncomine database. Next, inhibition of AGTR1 reduced tumor growth and LNM in orthotopic xenografts by bioluminescence imaging (BLI). Losartan, an AGTR1-specific inhibitor, decreased the chemokine pair CXCR4/SDF-1α levels in vivo and inhibited AGTR1-induced cell migration and invasion in vitro. Finally, the molecular mechanism of AGTR1-induced cell migration and LNM was assessed by knocking down AGTR1 in normal cells or CXCR4 in AGTR1high cells. AGTR1-silenced cells treated with losartan showed lower CXCR4 expression. AGTR1 overexpression caused the upregulation of FAK/RhoA signaling molecules, while knocking down CXCR4 in AGTR1high cells downregulated these molecules. Collectively, AGTR1 promotes LNM by increasing the chemokine pair CXCR4/SDF-1α and tumor cell migration and invasion. The potential mechanism of AGTR1-mediated cell movement relies on activating the FAK/RhoA pathway. Our study indicated that inhibiting AGTR1 may be a potential therapeutic target for LNM in early-stage breast cancer.
Collapse
Affiliation(s)
- Yuxi Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zihan Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunmei Ye
- Department of Breast Surgery, Wuhan Women and Children's Health Care Center, Wuhan 430022, China
| | - Chong Lu
- Department of Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Zhou
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Juan Pan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cuiwei Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jieying Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Linka Xie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanxia Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
27
|
Acconcia F. The Network of Angiotensin Receptors in Breast Cancer. Cells 2020; 9:cells9061336. [PMID: 32471115 PMCID: PMC7349848 DOI: 10.3390/cells9061336] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
The renin-angiotensin system (RAS) is a network of proteins regulating many aspects of human physiology, including cardiovascular, pulmonary, and immune system physiology. The RAS is a complicated network of G-protein coupled receptors (GPCRs) (i.e., AT1R, AT2R, MASR, and MRGD) orchestrating the effects of several hormones (i.e., angiotensin II, angiotensin (1-7), and alamandine) produced by protease-based transmembrane receptors (ACE1 and ACE2). Two signaling axes have been identified in the RAS endocrine system that mediate the proliferative actions of angiotensin II (i.e., the AT1R-based pathway) or the anti-proliferative effects of RAS hormones (i.e., the AT2R-, MAS-, and MRGD-based pathways). Disruption of the balance between these two axes can cause different diseases (e.g., cardiovascular pathologies and the severe acute respiratory syndrome coronavirus 2- (SARS-CoV-2)-based COVID-19 disease). It is now accepted that all the components of the RAS endocrine system are expressed in cancer, including cancer of the breast. Breast cancer (BC) is a multifactorial pathology for which there is a continuous need to identify novel drugs. Here, I reviewed the possible roles of both axes of the RAS endocrine network as potential druggable pathways in BC. Remarkably, the analysis of the current knowledge of the different GPCRs of the RAS molecular system not only confirms that AT1R could be considered a drug target and that its inhibition by losartan and candesartan could be useful in the treatment of BC, but also identifies Mas-related GPCR member D (MRGD) as a druggable protein. Overall, the RAS of GPCRs offers multifaceted opportunities for the development of additional compounds for the treatment of BC.
Collapse
Affiliation(s)
- Filippo Acconcia
- Department of Sciences, Biomedical Sciences and Technology Section, University Roma TRE, Viale Guglielmo Marconi 446, I-00146 Rome, Italy
| |
Collapse
|
28
|
PBX3 Promotes Tumor Growth and Angiogenesis via Activation of AT1R/VEGFR2 Pathway in Papillary Thyroid Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8954513. [PMID: 32047817 PMCID: PMC7007751 DOI: 10.1155/2020/8954513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/20/2019] [Accepted: 07/29/2019] [Indexed: 11/17/2022]
Abstract
PBX3 (Pre-B-cell leukemia homeobox 3) had been considered to be a multifunctional oncogene which involved in tumor growth, invasion, and metastasis in leukemia and some solid tumors. However, the contribution of PBX3 to papillary thyroid carcinoma (PTC) remains unclear. In this study, we found that PBX3 expression was significantly upregulated in PTC tissues compared to adjacent normal tissues, and high levels of PBX3 were correlated with tumor size, lymphatic metastasis, TMN stage, and poor prognosis of PTC patients. Overexpression of PBX3 in PTC cell lines promoted cell proliferation. Consistently, knockdown of PBX3 by shRNA induced cell cycle arrest at G0/G1 phase, and inhibited angiogenesis and tumor growth in vitro and in vivo. Furthermore, PBX3 promoted PTC cell proliferation and angiogenesis through activation of AT1R/VEGFR2 pathway while overexpression of AT1R and treatment with VEGFA reversed PBX3-shRNA-induced decreased phosphorylation of VEGFR2 and its downstream (ERK1/2, AKT and Src). It demonstrated that PBX3 could be used as a potential prognostic biomarker and therapeutic target for PTC.
Collapse
|
29
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
30
|
Liu Z, Li M, Hua Q, Li Y, Wang G. Identification of an eight-lncRNA prognostic model for breast cancer using WGCNA network analysis and a Cox‑proportional hazards model based on L1-penalized estimation. Int J Mol Med 2019; 44:1333-1343. [PMID: 31432096 PMCID: PMC6713414 DOI: 10.3892/ijmm.2019.4303] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
An ever‑increasing number of long noncoding (lnc)RNAs has been identified in breast cancer. The present study aimed to establish an lncRNA signature for predicting survival in breast cancer. RNA expression profiling was performed using microarray gene expression data from the National Center for Biotechnology Information Gene Expression Omnibus, followed by the identification of breast cancer‑related preserved modules using weighted gene co‑expression network (WGCNA) network analysis. From the lncRNAs identified in these preserved modules, prognostic lncRNAs were selected using univariate Cox regression analysis in combination with the L1‑penalized (LASSO) Cox‑proportional Hazards (Cox‑PH) model. A risk score based on these prognostic lncRNAs was calculated and used for risk stratification. Differentially expressed RNAs (DERs) in breast cancer were identified using MetaDE. Gene Set Enrichment Analysis pathway enrichment analysis was conducted for these prognostic lncRNAs and the DERs related to the lncRNAs in the preserved modules. A total of five preserved modules comprising 73 lncRNAs were mined. An eight‑lncRNA signature (IGHA1, IGHGP, IGKV2‑28, IGLL3P, IGLV3‑10, AZGP1P1, LINC00472 and SLC16A6P1) was identified using the LASSO Cox‑PH model. Risk score based on these eight lncRNAs could classify breast cancer patients into two groups with significantly different survival times. The eight‑lncRNA signature was validated using three independent cohorts. These prognostic lncRNAs were significantly associated with the cell adhesion molecules pathway, JAK‑signal transducer and activator of transcription 5A pathway, and erbb pathway and are potentially involved in regulating angiotensin II receptor type 1, neuropeptide Y receptor Y1, KISS1 receptor, and C‑C motif chemokine ligand 5. The developed eight‑lncRNA signature may have clinical implications for predicting prognosis in breast cancer. Overall, this study provided possible molecular targets for the development of novel therapies against breast cancer.
Collapse
Affiliation(s)
- Zhenbin Liu
- Department of Ulcer and Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Menghu Li
- Department of Ulcer and Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Qi Hua
- Department of Ulcer and Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yanfang Li
- Department of Ulcer and Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Gang Wang
- Department of Ulcer and Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| |
Collapse
|
31
|
Feng YL, Chen DQ, Vaziri ND, Guo Y, Zhao YY. Small molecule inhibitors of epithelial-mesenchymal transition for the treatment of cancer and fibrosis. Med Res Rev 2019; 40:54-78. [PMID: 31131921 DOI: 10.1002/med.21596] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/20/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
Tissue fibrosis and cancer both lead to high morbidity and mortality worldwide; thus, effective therapeutic strategies are urgently needed. Because drug resistance has been widely reported in fibrotic tissue and cancer, developing a strategy to discover novel targets for targeted drug intervention is necessary for the effective treatment of fibrosis and cancer. Although many factors lead to fibrosis and cancer, pathophysiological analysis has demonstrated that tissue fibrosis and cancer share a common process of epithelial-mesenchymal transition (EMT). EMT is associated with many mediators, including transcription factors (Snail, zinc-finger E-box-binding protein and signal transducer and activator of transcription 3), signaling pathways (transforming growth factor-β1, RAC-α serine/threonine-protein kinase, Wnt, nuclear factor-kappa B, peroxisome proliferator-activated receptor, Notch, and RAS), RNA-binding proteins (ESRP1 and ESRP2) and microRNAs. Therefore, drugs targeting EMT may be a promising therapy against both fibrosis and tumors. A large number of compounds that are synthesized or derived from natural products and their derivatives suppress the EMT by targeting these mediators in fibrosis and cancer. By targeting EMT, these compounds exhibited anticancer effects in multiple cancer types, and some of them also showed antifibrotic effects. Therefore, drugs targeting EMT not only have both antifibrotic and anticancer effects but also exert effective therapeutic effects on multiorgan fibrosis and cancer, which provides effective therapy against fibrosis and cancer. Taken together, the results highlighted in this review provide new concepts for discovering new antifibrotic and antitumor drugs.
Collapse
Affiliation(s)
- Ya-Long Feng
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Dan-Qian Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Nosratola D Vaziri
- Department of Medicine, University of California Irvine, Irvine, California
| | - Yan Guo
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China.,Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Ying-Yong Zhao
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
32
|
Cho TM, Kim JY, Kim YJ, Sung D, Oh E, Jang S, Farrand L, Hoang VH, Nguyen CT, Ann J, Lee J, Seo JH. C-terminal HSP90 inhibitor L80 elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Cancer Lett 2019; 447:141-153. [DOI: 10.1016/j.canlet.2019.01.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/28/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
|
33
|
Xu K, Han B, Bai Y, Ma XY, Ji ZN, Xiong Y, Miao SK, Zhang YY, Zhou LM. MiR-451a suppressing BAP31 can inhibit proliferation and increase apoptosis through inducing ER stress in colorectal cancer. Cell Death Dis 2019; 10:152. [PMID: 30770794 PMCID: PMC6377610 DOI: 10.1038/s41419-019-1403-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 12/23/2018] [Accepted: 01/18/2019] [Indexed: 02/05/2023]
Abstract
The global morbidity and mortality of colorectal cancer (CRC) are ranked the third among gastrointestinal tumors in the world. MiR-451a is associated with several types of cancer, including CRC. However, the roles and mechanisms of miR-451a in CRC have not been elucidated. BAP31 is a predicted target gene of miR-451a in our suppression subtractive hybridization library. Its relationship with miR-451a and function in CRC are unclear. We hypothesized that miR-451a could induce apoptosis through suppressing BAP31 in CRC. Immunohistochemistry and real-time PCR were used to measure BAP31 expressions in CRC tissues and pericarcinous tissues from 57 CRC patients and CRC cell lines. Dual-luciferase reporter assay was used to detect the binding of miR-451a to BAP31. The expression of BAP31 protein in CRC tissues was significantly higher than that in pericarcinous tissues, which was correlated with distant metastasis and advanced clinical stages of CRC patients. The expression of BAP31 was higher in HCT116, HT29, SW620, and DLD cells than that in the normal colonic epithelial cell line NCM460. The expression of BAP31 was absolutely down-regulated when over-expressing miR-451a in HCT116 and SW620 cells compared with control cells. Mir-451a inhibited the expression of BAP31 by binding to its 5'-UTR. Over-expressing miR-451a or silencing BAP31 suppressed the proliferation and apoptosis of CRC cells by increasing the expressions of endoplasmic reticulum stress (ERS)-associated proteins, including GRP78/BIP, BAX, and PERK/elF2α/ATF4/CHOP, which resulted in increased ERS, cytoplasmic calcium ion flowing, and apoptosis of CRC cells. These changes resulting from over-expressing miR-451a were reversed by over-expressing BAP31 with mutated miR-451a-binding sites. Over-expressing miR-451a or silencing BAP31 inhibited tumor growth by inducing ERS. The present study demonstrated that miR-451a can inhibit proliferation and increase apoptosis through inducing ERS by binding to the 5'-UTR of BAP31 in CRC.
Collapse
Affiliation(s)
- Ke Xu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Bin Han
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yang Bai
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiu-Ying Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Zhen-Ni Ji
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yao Xiong
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shi-Kun Miao
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuan-Yuan Zhang
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China.
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Li-Ming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China.
- 985 Science and Technology Platform for Innovative Drugs, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
34
|
Oh E, Kim YJ, An H, Sung D, Cho TM, Farrand L, Jang S, Seo JH, Kim JY. Flubendazole elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Int J Cancer 2018; 143:1978-1993. [PMID: 29744876 DOI: 10.1002/ijc.31585] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/23/2018] [Accepted: 04/12/2018] [Indexed: 12/16/2022]
Abstract
Tumor metastasis remains the cause of 90% of cancer-related deaths. Cancer stem cells (CSC) are thought to be responsible for the aggressive and metastatic nature of triple-negative breast cancers (TNBC), and new therapeutic strategies are being devised to target them. Flubendazole (FLU) is a widely used anthelmintic agent that also exhibits anticancer activity in several cancer types. The aim of this study was to characterize the mechanism of action of FLU on breast cancer stem cell (BCSC)-like properties and metastasis in TNBC. FLU treatment caused a significant induction of apoptosis, accompanied by G2/M phase accumulation, caspase-3/-7 activation and the dysregulation of STAT3 activation in TNBC cells. The latter phenomenon was associated with impairment of cancer stem-like traits, concomitant with a reduction in the CD24low /CD44high , CD24high /CD49fhigh subpopulation, ALDH1 activity and mammosphere formation. The BCSC-enriched populations exhibited enhanced metastasis with higher STAT3 activation, while FLU administration inhibited tumor growth, angiogenesis and lung and liver metastasis, coinciding with decreased MMP-2 and MMP-9 levels in circulating blood. FLU kills not only rapid proliferating tumor cells but also effectively eradicates BCSC-like cells in vitro and in vivo. Our findings warrant further investigation of FLU as a treatment for metastatic TNBC.
Collapse
Affiliation(s)
- Eunhye Oh
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 152-703, Republic of Korea
| | - Hyunsook An
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| | - Daeil Sung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| | - Tae-Min Cho
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| | - Lee Farrand
- Medvet Science, 65 Hardys Rd, Underdale, Adelaide, Australia
| | - Seojin Jang
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 152-703, Republic of Korea
| |
Collapse
|
35
|
Chaudhary M, Chaudhary S. Unravelling the Lesser Known Facets of Angiotensin II Type 1 Receptor. Curr Hypertens Rep 2018; 19:1. [PMID: 28083801 DOI: 10.1007/s11906-017-0699-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Hypertension is an important risk factor in various pathologies. Despite enormous advancements in health sciences, the number of hypertensive individuals is increasing worldwide. The complex interplay between genetic and epigenetic factors seems to be a promising pathway to exploring the pathophysiology of hypertension. RECENT FINDINGS Various single gene and genome wide association studies have generated huge but non-reproducible data that highlights the role of some additional but as yet unidentified factor(s) in disease outcome. Dietary pattern and epigenetic mechanism (mainly DNA methylation) have shown a profound effect on blood pressure regulation. Angiotensin II and its receptors are known to play an important role in maintaining blood pressure; hence, a larger section of antihypertensive drugs targets the renin-angiotensin system (RAS). Angiotensin II type 1 receptor (AT1R), besides maintaining blood pressure, also has a role in cancer progression. Besides other pathways, RAS still remains the main player in blood pressure regulation. Additionally, AT1R has recently emerged as a molecule with diverse roles ranging from physiologic to cancer progression.
Collapse
Affiliation(s)
- Mayank Chaudhary
- Department cum National Centre for Human Genome Studies and Research (NCHGSR), Panjab University, Chandigarh, 160 014, India
| | - Shashi Chaudhary
- Department cum National Centre for Human Genome Studies and Research (NCHGSR), Panjab University, Chandigarh, 160 014, India.
| |
Collapse
|
36
|
Ishikane S, Takahashi-Yanaga F. The role of angiotensin II in cancer metastasis: Potential of renin-angiotensin system blockade as a treatment for cancer metastasis. Biochem Pharmacol 2018. [PMID: 29534876 DOI: 10.1016/j.bcp.2018.03.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hypertension, which often exists as a comorbid condition in cancer patients, is considered as a factor affecting cancer progression. The renin-angiotensin system (RAS) plays an important role in the regulation of blood pressure, and angiotensin II (Ang II) is a well-known pressor peptide in RAS. There is also accumulated evidence indicating that Ang II plays a critical role in the metastasis of various cancers by modulating adhesion, migration invasion, proliferation, and angiogenesis. Consistent with this, large epidemiological studies have reported the potential beneficial effects of angiotensin-converting enzyme (ACE) inhibitors and Ang II type 1 receptor blockers (ARBs) against cancer metastasis; however, some of the results remain controversial. Although the precise Ang II-related mechanisms involved in cancer metastasis are not completely clear yet, a number of basic and meta-analytic studies have shown that ACE inhibitors and ARBs reduce the metastatic potential of tumors. In this review, we summarize the relationships among hypertension, RAS, and metastasis as demonstrated in basic and clinical studies. Finally, we discuss the possibility of using RAS inhibitors as anti-metastatic drugs.
Collapse
Affiliation(s)
- Shin Ishikane
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Japan.
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
37
|
Aberrant expression of lncRNAs and mRNAs in patients with intracranial aneurysm. Oncotarget 2018; 8:2477-2484. [PMID: 27965470 PMCID: PMC5356817 DOI: 10.18632/oncotarget.13908] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/01/2016] [Indexed: 01/14/2023] Open
Abstract
Intracranial aneurysm (IA) is pathological dilatations of the cerebral artery and rupture of IAs can cause subarachnoid hemorrhage, which has a high ratio of fatality and morbidity. However, the pathogenesis of IAs remains unknown. We performed long noncoding RNA (lncRNA) and messenger RNA (mRNA) expression profiles in IA tissues and superficial temporal arteries (STAs). A total of 4129 differentially expressed lncRNAs and 2926 differentially expressed mRNAs were obtained from the microarrays (P < 0.05). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses showed that up-regulated mRNAs were enriched in immune response, inflammatory response, regulation of immune response and lysosome, et al; while the down-regulated mRNAs were enriched in muscle contraction, smooth muscle contraction, cGMP-PKG signaling pathway and vascular smooth muscle contraction, et al. The lncRNA-mRNA co-expression networks were represented in immune response, inflammatory response, muscle contraction and vascular smooth muscle contraction. These findings may gain insight in the pathogenesis of IAs and provide clues to find key roles for IA patients.
Collapse
|
38
|
Kim YJ, Sung D, Oh E, Cho Y, Cho TM, Farrand L, Seo JH, Kim JY. Flubendazole overcomes trastuzumab resistance by targeting cancer stem-like properties and HER2 signaling in HER2-positive breast cancer. Cancer Lett 2018; 412:118-130. [DOI: 10.1016/j.canlet.2017.10.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 12/15/2022]
|
39
|
Ekambaram P, Lee JYL, Hubel NE, Hu D, Yerneni S, Campbell PG, Pollock N, Klei LR, Concel VJ, Delekta PC, Chinnaiyan AM, Tomlins SA, Rhodes DR, Priedigkeit N, Lee AV, Oesterreich S, McAllister-Lucas LM, Lucas PC. The CARMA3-Bcl10-MALT1 Signalosome Drives NFκB Activation and Promotes Aggressiveness in Angiotensin II Receptor-Positive Breast Cancer. Cancer Res 2017; 78:1225-1240. [PMID: 29259013 DOI: 10.1158/0008-5472.can-17-1089] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 11/09/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023]
Abstract
The angiotensin II receptor AGTR1, which mediates vasoconstrictive and inflammatory signaling in vascular disease, is overexpressed aberrantly in some breast cancers. In this study, we established the significance of an AGTR1-responsive NFκB signaling pathway in this breast cancer subset. We documented that AGTR1 overexpression occurred in the luminal A and B subtypes of breast cancer, was mutually exclusive of HER2 expression, and correlated with aggressive features that include increased lymph node metastasis, reduced responsiveness to neoadjuvant therapy, and reduced overall survival. Mechanistically, AGTR1 overexpression directed both ligand-independent and ligand-dependent activation of NFκB, mediated by a signaling pathway that requires the triad of CARMA3, Bcl10, and MALT1 (CBM signalosome). Activation of this pathway drove cancer cell-intrinsic responses that include proliferation, migration, and invasion. In addition, CBM-dependent activation of NFκB elicited cancer cell-extrinsic effects, impacting endothelial cells of the tumor microenvironment to promote tumor angiogenesis. CBM/NFκB signaling in AGTR1+ breast cancer therefore conspires to promote aggressive behavior through pleiotropic effects. Overall, our results point to the prognostic and therapeutic value of identifying AGTR1 overexpression in a subset of HER2-negative breast cancers, and they provide a mechanistic rationale to explore the repurposing of drugs that target angiotensin II-dependent NFκB signaling pathways to improve the treatment of this breast cancer subset.Significance: These findings offer a mechanistic rationale to explore the repurposing of drugs that target angiotensin action to improve the treatment of AGTR1-expressing breast cancers. Cancer Res; 78(5); 1225-40. ©2017 AACR.
Collapse
Affiliation(s)
- Prasanna Ekambaram
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jia-Ying Lloyd Lee
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nathaniel E Hubel
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dong Hu
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Phil G Campbell
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Netanya Pollock
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Linda R Klei
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Vincent J Concel
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Phillip C Delekta
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Scott A Tomlins
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel R Rhodes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Nolan Priedigkeit
- Women's Cancer Research Center, Magee-Womens Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Adrian V Lee
- Women's Cancer Research Center, Magee-Womens Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Women's Cancer Research Center, Magee-Womens Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Linda M McAllister-Lucas
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Peter C Lucas
- Departments of Pathology and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
40
|
De Francesco EM, Sotgia F, Clarke RB, Lisanti MP, Maggiolini M. G Protein-Coupled Receptors at the Crossroad between Physiologic and Pathologic Angiogenesis: Old Paradigms and Emerging Concepts. Int J Mol Sci 2017; 18:ijms18122713. [PMID: 29240722 PMCID: PMC5751314 DOI: 10.3390/ijms18122713] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have been implicated in transmitting signals across the extra- and intra-cellular compartments, thus allowing environmental stimuli to elicit critical biological responses. As GPCRs can be activated by an extensive range of factors including hormones, neurotransmitters, phospholipids and other stimuli, their involvement in a plethora of physiological functions is not surprising. Aberrant GPCR signaling has been regarded as a major contributor to diverse pathologic conditions, such as inflammatory, cardiovascular and neoplastic diseases. In this regard, solid tumors have been demonstrated to activate an angiogenic program that relies on GPCR action to support cancer growth and metastatic dissemination. Therefore, the manipulation of aberrant GPCR signaling could represent a promising target in anticancer therapy. Here, we highlight the GPCR-mediated angiogenic function focusing on the molecular mechanisms and transduction effectors driving the patho-physiological vasculogenesis. Specifically, we describe evidence for the role of heptahelic receptors and associated G proteins in promoting angiogenic responses in pathologic conditions, especially tumor angiogenesis and progression. Likewise, we discuss opportunities to manipulate aberrant GPCR-mediated angiogenic signaling for therapeutic benefit using innovative GPCR-targeted and patient-tailored pharmacological strategies.
Collapse
Affiliation(s)
- Ernestina M De Francesco
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy.
- Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Federica Sotgia
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK.
| | - Robert B Clarke
- Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Michael P Lisanti
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy.
| |
Collapse
|
41
|
Coulson R, Liew SH, Connelly AA, Yee NS, Deb S, Kumar B, Vargas AC, O'Toole SA, Parslow AC, Poh A, Putoczki T, Morrow RJ, Alorro M, Lazarus KA, Yeap EFW, Walton KL, Harrison CA, Hannan NJ, George AJ, Clyne CD, Ernst M, Allen AM, Chand AL. The angiotensin receptor blocker, Losartan, inhibits mammary tumor development and progression to invasive carcinoma. Oncotarget 2017; 8:18640-18656. [PMID: 28416734 PMCID: PMC5386636 DOI: 10.18632/oncotarget.15553] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
Drugs that target the Renin-Angiotensin System (RAS) have recently come into focus for their potential utility as cancer treatments. The use of Angiotensin Receptor Blockers (ARBs) and Angiotensin-Converting Enzyme (ACE) Inhibitors (ACEIs) to manage hypertension in cancer patients is correlated with improved survival outcomes for renal, prostate, breast and small cell lung cancer. Previous studies demonstrate that the Angiotensin Receptor Type I (AT1R) is linked to breast cancer pathogenesis, with unbiased analysis of gene-expression studies identifying significant up-regulation of AGTR1, the gene encoding AT1R in ER+ve/HER2−ve tumors correlating with poor prognosis. However, there is no evidence, so far, of the functional contribution of AT1R to breast tumorigenesis. We explored the potential therapeutic benefit of ARB in a carcinogen-induced mouse model of breast cancer and clarified the mechanisms associated with its success. Mammary tumors were induced with 7,12-dimethylbenz[α]antracene (DMBA) and medroxyprogesterone acetate (MPA) in female wild type mice and the effects of the ARB, Losartan treatment assessed in a preventative setting (n = 15 per group). Tumor histopathology was characterised by immunohistochemistry, real-time qPCR to detect gene expression signatures, and tumor cytokine levels measured with quantitative bioplex assays. AT1R was detected with radiolabelled ligand binding assays in fresh frozen tumor samples. We showed that therapeutic inhibition of AT1R, with Losartan, resulted in a significant reduction in tumor burden; and no mammary tumor incidence in 20% of animals. We observed a significant reduction in tumor progression from DCIS to invasive cancer with Losartan treatment. This was associated with reduced tumor cell proliferation and a significant reduction in IL-6, pSTAT3 and TNFα levels. Analysis of tumor immune cell infiltrates, however, demonstrated no significant differences in the recruitment of lymphocytes or tumour-associated macrophages in Losartan or vehicle-treated mammary tumors. Analysis of AT1R expression with radiolabelled ligand binding assays in human breast cancer biopsies showed high AT1R levels in 30% of invasive ductal carcinomas analysed. Furthermore, analysis of the TCGA database identified that high AT1R expression to be associated with luminal breast cancer subtype. Our in vivo data and analysis of human invasive ductal carcinoma samples identify the AT1R is a potential therapeutic target in breast cancer, with the availability of a range of well-tolerated inhibitors currently used in clinics. We describe a novel signalling pathway critical in breast tumorigenesis, that may provide new therapeutic avenues to complement current treatments.
Collapse
Affiliation(s)
- Rhiannon Coulson
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia.,Translational Breast Cancer Research, Garvan Institute, Darlinghurst, Sydney, NSW, Australia
| | - Seng H Liew
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | | | - Nicholas S Yee
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Siddhartha Deb
- Anatomical Pathology, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Beena Kumar
- Anatomical Pathology, Monash Health, Clayton, VIC, Australia
| | - Ana C Vargas
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia
| | - Sandra A O'Toole
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia.,Translational Breast Cancer Research, Garvan Institute, Darlinghurst, Sydney, NSW, Australia.,Sydney Medical School, Sydney University, NSW, Australia
| | - Adam C Parslow
- Tumor Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Ashleigh Poh
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, VIC, Australia
| | - Tracy Putoczki
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, VIC, Australia
| | - Riley J Morrow
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Mariah Alorro
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Kyren A Lazarus
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia.,Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Evie F W Yeap
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia
| | - Kelly L Walton
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Craig A Harrison
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Natalie J Hannan
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital, Heidelberg, VIC, Australia
| | - Amee J George
- The ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Colin D Clyne
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia
| | - Matthias Ernst
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Andrew M Allen
- Department of Physiology, University of Melbourne, VIC, Australia
| | - Ashwini L Chand
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia.,Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| |
Collapse
|
42
|
Oh E, Kim JY, Sung D, Cho Y, Lee N, An H, Kim YJ, Cho TM, Seo JH. Inhibition of ubiquitin-specific protease 34 (USP34) induces epithelial-mesenchymal transition and promotes stemness in mammary epithelial cells. Cell Signal 2017; 36:230-239. [DOI: 10.1016/j.cellsig.2017.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022]
|
43
|
Cambados N, Walther T, Nahmod K, Tocci JM, Rubinstein N, Böhme I, Simian M, Sampayo R, Del Valle Suberbordes M, Kordon EC, Schere-Levy C. Angiotensin-(1-7) counteracts the transforming effects triggered by angiotensin II in breast cancer cells. Oncotarget 2017; 8:88475-88487. [PMID: 29179450 PMCID: PMC5687620 DOI: 10.18632/oncotarget.19290] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 06/02/2017] [Indexed: 12/26/2022] Open
Abstract
Angiotensin (Ang) II, the main effector peptide of the renin-angiotensin system, has been implicated in multiple aspects of cancer progression such as proliferation, migration, invasion, angiogenesis and metastasis. Ang-(1-7), is a biologically active heptapeptide, generated predominantly from AngII by the enzymatic activity of angiotensin converting enzyme 2. Previous studies have shown that Ang-(1-7) counterbalances AngII actions in different pathophysiological settings. In this study, we have analysed the impact of Ang-(1-7) on AngII-induced pro-tumorigenic features on normal murine mammary epithelial cells NMuMG and breast cancer cells MDA-MB-231. AngII stimulated the activation of the survival factor AKT in NMuMG cells mainly through the AT1 receptor. This PI3K/AKT pathway activation also promoted epithelial–mesenchymal transition (EMT). Concomitant treatment of NMuMG cells with AngII and Ang-(1-7) completely abolished EMT features induced by AngII. Furthermore, Ang-(1-7) abrogated AngII induced migration and invasion of the MDA-MB-231 cells as well as pro-angiogenic events such as the stimulation of MMP-9 activity and VEGF expression. Together, these results demonstrate for the first time that Ang-(1-7) counteracts tumor aggressive signals stimulated by AngII in breast cancer cells emerging the peptide as a potential therapy to prevent breast cancer progression.
Collapse
Affiliation(s)
- Nadia Cambados
- Instituto de Fisiología, Biología Molecular y Neurociencias, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Thomas Walther
- Department of Obstetrics, University of Leipzig, Leipzig, Germany.,Department Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork, Ireland.,Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Karen Nahmod
- Department of Pediatrics, Immunology, Allergy and Rheumatology, Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas, USA
| | - Johanna M Tocci
- Instituto de Fisiología, Biología Molecular y Neurociencias, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Rubinstein
- Instituto de Fisiología, Biología Molecular y Neurociencias, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ilka Böhme
- Department of Obstetrics, University of Leipzig, Leipzig, Germany.,Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Marina Simian
- Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Rocío Sampayo
- Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Melisa Del Valle Suberbordes
- Instituto de Fisiología, Biología Molecular y Neurociencias, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Edith C Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departmento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Schere-Levy
- Instituto de Fisiología, Biología Molecular y Neurociencias, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
44
|
Kim JY, Lee N, Kim YJ, Cho Y, An H, Oh E, Cho TM, Sung D, Seo JH. Disulfiram induces anoikis and suppresses lung colonization in triple-negative breast cancer via calpain activation. Cancer Lett 2017; 386:151-160. [DOI: 10.1016/j.canlet.2016.11.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/12/2016] [Accepted: 11/17/2016] [Indexed: 01/12/2023]
|