1
|
Chintalaramulu N, Singh DP, Sapkota B, Raman D, Alahari S, Francis J. Caveolin-1: an ambiguous entity in breast cancer. Mol Cancer 2025; 24:109. [PMID: 40197489 PMCID: PMC11974173 DOI: 10.1186/s12943-025-02297-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer in women and the second leading cause of death from cancer among women. Metastasis is the major cause of BC-associated mortality. Accumulating evidence implicates Caveolin-1 (Cav-1), a structural protein of plasma membrane caveolae, in BC metastasis. Cav-1 exhibits a dual role, as both a tumor suppressor and promoter depending on the cellular context and BC subtype. This review highlights the role of Cav-1 in modulating glycolytic metabolism, tumor-stromal interactions, apoptosis, and senescence. Additionally, stromal Cav-1's expression is identified as a potential prognostic marker, offering insights into its contrasting roles in tumor suppression and progression. Furthermore, Cav-1's context-dependent effects are explored in BC subtypes including hormone receptor-positive, HER2-positive, and triple-negative BC (TNBC). The review further delves into the role of Cav-1 in regulating the metastatic cascade including extracellular matrix interactions, cell migration and invasion, and premetastatic niche formation. The later sections discuss the therapeutic targeting of Cav-1 by metabolic inhibitors such as betulinic acid and Cav-1 modulating compounds. While Cav-1 may be a potential biomarker and therapeutic target, its heterogeneous expression and context-specific activity necessitates further research to develop precise interventions. Future studies investigating the mechanistic role of Cav-1 in metastasis may pave the way for effective treatment of metastatic BC.
Collapse
Affiliation(s)
- Naveen Chintalaramulu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | | | - Biplov Sapkota
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
2
|
Li G, Liu R, Peng Z, Zhang S, Sun R, Wang Z, Li J, Gao Y, Xu Y, Cui J, Liu J, Yan J, Cao L, Ren S, Chu Y, Feng L, Yang L, Shen Y, Qi Z. Inhibition of CAV1 attenuates diabetic cardiomyopathy through reducing ferroptosis via activating NRF2/GCLC signaling pathway. Theranostics 2025; 15:4989-5006. [PMID: 40303344 PMCID: PMC12036865 DOI: 10.7150/thno.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Diabetic cardiomyopathy (DCM), a prevalent complication of diabetes, is a major cause of heart failure and death among patients with diabetes. However, the pathological mechanisms underlying the development of DCM remain unclear. This study aims to investigate the role and underlying mechanisms of caveolin-1 (CAV1) in DCM. Methods: DCM model was established in vivo through intraperitoneal injection of streptozotocin in mice and in vitro through high-glucose (HG) treatment in neonatal rat ventricular myocytes (NRVMs). CAV1-knockout (CAV1-KO) and overexpression (by injecting adeno-associated virus 9 (AAV9) encoding CAV1) mice were utilized to explore the role of CAV1 in DCM. Nuclear factor erythroid 2-related factor 2 (NRF2)-KO and AAV9-NRF2 mice and ML385 (an NRF2 inhibitor) were used to investigate the effect of NRF2 on DCM. Results: CAV1 expression was significantly increased in the cardiac tissues of diabetic mice and HG-treated NRVMs. CAV1 deficiency significantly alleviated diabetes-induced myocardial hypertrophy, fibrosis, abnormal mitochondria, excessive reactive oxygen species production, and ferroptosis. Conversely, cardiac-specific overexpression of CAV1 exacerbated cardiac dysfunction and myocardial histological abnormalities caused by diabetes. Mechanistically, CAV1 directly bound to NRF2 and inhibited its nuclear translocation, reducing the transcription of glutamate cysteine ligase catalytic subunit (GCLC), accumulating excess peroxide, and inducing ferroptosis and myocardial injury. Conclusion: CAV1 exacerbates the progression of DCM by suppressing the NRF2/GCLC pathway, suggesting that targeting CAV1 is a potential therapeutic approach for DCM.
Collapse
Affiliation(s)
- Guangru Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Ruiqing Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
- The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Zeyan Peng
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Runjia Sun
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Ziwei Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300000, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Jianlin Cui
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Lei Cao
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300000, China
| | - Shan Ren
- The First Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University, Shihezi, 832003, China
| | - Yushun Chu
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300000, China
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University; Department of Cardiology, Beichen Hospital, Nankai University, Tianjin, 300071, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300000, China
- The First Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University, Shihezi, 832003, China
- Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| |
Collapse
|
3
|
Kamposioras K, Dinas PC, Barriuoso J, Trachana V, Dimas K. Caveolin-1 protein expression as a prognostic biomarker of gastrointestinal tumours: A systematic review and meta-analysis. Eur J Clin Invest 2023; 53:e14065. [PMID: 37497737 DOI: 10.1111/eci.14065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Gastrointestinal (GI) cancers remain a major threat worldwide, accounting for over 30% of cancer deaths. The identification of novel prognostic biomarkers remains a challenge despite significant advances in the field. The CAV1 gene, encoding the caveolin-1 protein, remains enigmatic in cancer and carcinogenesis, as it has been proposed to act as both a tumour promoter and a tumour suppressor. METHODS To analyse the differential role of caveolin-1 expression in both tumour cells and stroma in relation to prognosis in GI tumours, we performed a systematic review and meta-analysis according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines; PROSPERO registration number: CRD42022299148. RESULTS Our analysis showed that high levels of caveolin-1 in tumour cells were associated with poor prognosis and inferior overall survival (OS) in oesophageal and pancreatic cancer and hepatocellular carcinoma (HCC), but not in gastric and colorectal cancer. Importantly, our study showed that higher stromal caveolin-1 expression was associated with significantly longer OS and disease-free survival in colorectal cancer. Analysis of stromal caveolin-1 expression in the remaining tumours showed a similar trend, although it did not reach statistical significance. CONCLUSIONS The data suggest that caveolin-1 expression in the tumour cells of oesophageal, pancreatic cancer and HCC and in the stroma of colorectal cancer may be an important novel predictive biomarker for the clinical management of these diseases in a curative setting. However, the main conclusion of our analysis is that caveolin-1 expression should always be assessed separately in stroma and tumour cells.
Collapse
Affiliation(s)
| | - Petros C Dinas
- FAME Laboratory, Department of Physical Education and Sport Science, University of Thessaly, Volos, Greece
| | - Jorge Barriuoso
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Volos, Greece
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Volos, Greece
| |
Collapse
|
4
|
Wan H, Cai Y, Xiao L, Ling Y, Ge L, Mo S, Xie Q, Peng S, Zhou B, Zeng X, Chen X. JFD, a Novel Natural Inhibitor of Keap1 Alkylation, Suppresses Intracellular Mycobacterium Tuberculosis Growth through Keap1/Nrf2/SOD2-Mediated ROS Accumulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6726654. [PMID: 36819778 PMCID: PMC9937762 DOI: 10.1155/2023/6726654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 02/12/2023]
Abstract
It is an effective strategy to treat tuberculosis by enhancing reactive oxygen species- (ROS-) mediated killing of Mycobacterium tuberculosis in macrophages, but there are no current therapeutic agents targeting this pathway. Honeysuckle has been used as the traditional medicine for tuberculosis treatment for 1500 years. Japoflavone D (JFD) is a novel biflavonoid isolated from Honeysuckle promoting ROS accumulation by Nrf2 pathway in hepatocarcinoma cells. However, its activity to kill M. tuberculosis in macrophages and molecular mechanism has not been reported. Our results showed that JFD enhances the M. tuberculosis elimination by boosting ROS levels in THP-1 cells. Moreover, the massive ROS accumulation activates p38 to induce apoptosis. Notably, the mechanism revealed that JFD suppresses the nuclear transport of Nrf2, thereby inhibiting SOD2 transcription, leading to a large ROS accumulation. Further studies showed that JFD disrupts the Keap1 alkylation at specific residues Cys14, Cys257, and Cys319, which is crucial for Nrf2 activation, thereby interrupts the nuclear transport of Nrf2. In pharmacokinetic study, JFD can stay as the prototype for 24 h in mice and can be excreted in feces without any toxicity. Our data reveal for the first time that a novel biflavonoid JFD as a potent inhibitor of Keap1 alkylation can suppress the nuclear transport of Nrf2. And it is the first research of the inhibitor of Keap1 alkylation. Furthermore, JFD robustly promotes M. tuberculosis elimination from macrophages by inhibiting Keap1/Nrf2/SOD2 pathway, resulting in the ROS accumulation. This work identified Keap1 alkylation as a new drug target for tuberculosis and provides a preliminary basis for the development of antituberculosis lead compounds based on JFD.
Collapse
Affiliation(s)
- Haoqiang Wan
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Lingyun Xiao
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Yunzhi Ling
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Lanlan Ge
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
| | - Siwei Mo
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Qiujie Xie
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Shusong Peng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Boping Zhou
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
| | - Xiaobin Zeng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong Province, China
- Department of Pathology (Longhua Branch), Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, 518020 Guangdong Province, China
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518120 Guangdong Province, China
| |
Collapse
|
5
|
Nrf2 Modulation in Breast Cancer. Biomedicines 2022; 10:biomedicines10102668. [PMID: 36289931 PMCID: PMC9599257 DOI: 10.3390/biomedicines10102668] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 12/05/2022] Open
Abstract
Reactive oxygen species (ROS) are identified to control the expression and activity of various essential signaling intermediates involved in cellular proliferation, apoptosis, and differentiation. Indeed, ROS represents a double-edged sword in supporting cell survival and death. Many common pathological processes, including various cancer types and neurodegenerative diseases, are inflammation and oxidative stress triggers, or even initiate them. Keap1-Nrf2 is a master antioxidant pathway in cytoprotective mechanisms through Nrf2 target gene expression. Activation of the Nfr2 pathway benefits cells in the early stages and reduces the level of ROS. In contrast, hyperactivation of Keap1-Nrf2 creates a context that supports the survival of both healthy and cancerous cells, defending them against oxidative stress, chemotherapeutic drugs, and radiotherapy. Considering the dual role of Nrf2 in suppressing or expanding cancer cells, determining its inhibitory/stimulatory position and targeting can represent an impressive role in cancer treatment. This review focused on Nrf2 modulators and their roles in sensitizing breast cancer cells to chemo/radiotherapy agents.
Collapse
|
6
|
Arjoune A, Sirard MA. The genomic response of human granulosa cells (KGN) to melatonin and specific agonists/antagonists to the melatonin receptors. Sci Rep 2022; 12:17539. [PMID: 36266374 PMCID: PMC9584952 DOI: 10.1038/s41598-022-21162-y] [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: 08/24/2022] [Accepted: 09/23/2022] [Indexed: 01/13/2023] Open
Abstract
Melatonin is a known modulator of follicle development; it acts through several molecular cascades via binding to its two specific receptors MT1 and MT2. Even though it is believed that melatonin can modulate granulosa cell (GC) functions, there is still limited knowledge of how it can act in human GC through MT1 and MT2 and which one is more implicated in the effects of melatonin on the metabolic processes in the dominant follicle. To better characterize the roles of these receptors on the effects of melatonin on follicular development, human granulosa-like tumor cells (KGN) were treated with specific melatonin receptor agonists and antagonists, and gene expression was analyzed with RNA-seq technology. Following appropriate normalization and the application of a fold change cut-off of 1.5 (FC 1.5, p ≤ 0.05) for each treatment, lists of the principal differentially expressed genes (DEGs) are generated. Analysis of major upstream regulators suggested that the MT1 receptor may be involved in the melatonin antiproliferative effect by reprogramming the metabolism of human GC by activating the PKB signaling pathway. Our data suggest that melatonin may act complementary through both MT1 and MT2 receptors to modulate human GC steroidogenesis, proliferation, and differentiation. However, MT2 receptors may be the ones implicated in transducing the effects of melatonin on the prevention of GC luteinization and follicle atresia at the antral follicular stage through stimulating the PKA pathway.
Collapse
Affiliation(s)
- Asma Arjoune
- grid.23856.3a0000 0004 1936 8390Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des Sciences de L’agriculture et de l’alimentation, Département des Sciences animales, Université Laval, Québec, QC G1V 0A6 Canada ,grid.419508.10000 0001 2295 3249Department of Animal Production, National Agronomic Institute of Tunisia, University of Carthage, 43 Avenue Charles Nicolle, 1082 Mahrajène, Tunisia
| | - Marc-André Sirard
- grid.23856.3a0000 0004 1936 8390Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des Sciences de L’agriculture et de l’alimentation, Département des Sciences animales, Université Laval, Québec, QC G1V 0A6 Canada
| |
Collapse
|
7
|
Petsouki E, Cabrera SNS, Heiss EH. AMPK and NRF2: Interactive players in the same team for cellular homeostasis? Free Radic Biol Med 2022; 190:75-93. [PMID: 35918013 DOI: 10.1016/j.freeradbiomed.2022.07.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 11/27/2022]
Abstract
NRF2 (Nuclear factor E2 p45-related factor 2) is a stress responsive transcription factor lending cells resilience against oxidative, xenobiotic, and also nutrient or proteotoxic insults. AMPK (AMP-activated kinase), considered as prime regulator of cellular energy homeostasis, not only tunes metabolism to provide the cell at any time with sufficient ATP or building blocks, but also controls redox balance and inflammation. Due to observed overlapping cellular responses upon AMPK or NRF2 activation and common stressors impinging on both AMPK and NRF2 signaling, it is plausible to assume that AMPK and NRF2 signaling may interdepend and cooperate to readjust cellular homeostasis. After a short introduction of the two players this narrative review paints the current picture on how AMPK and NRF2 signaling might interact on the molecular level, and highlights their possible crosstalk in selected examples of pathophysiology or bioactivity of drugs and phytochemicals.
Collapse
Affiliation(s)
- Eleni Petsouki
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Faculty of Life Sciences, Althanstrasse 14, 1090 Vienna, Austria
| | - Shara Natalia Sosa Cabrera
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Faculty of Life Sciences, Althanstrasse 14, 1090 Vienna, Austria; Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Austria
| | - Elke H Heiss
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Faculty of Life Sciences, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
8
|
Kim YS, Tang PW, Welles JE, Pan W, Javed Z, Elhaw AT, Mythreye K, Kimball SR, Hempel N. HuR-dependent SOD2 protein synthesis is an early adaptation to anchorage-independence. Redox Biol 2022; 53:102329. [PMID: 35594792 PMCID: PMC9121325 DOI: 10.1016/j.redox.2022.102329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/18/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
During metastasis cancer cells must adapt to survive loss of anchorage and evade anoikis. An important pro-survival adaptation is the ability of metastatic tumor cells to increase their antioxidant capacity and restore cellular redox balance. Although much is known about the transcriptional regulation of antioxidant enzymes in response to stress, how cells acutely adapt to alter antioxidant enzyme levels is less well understood. Using ovarian cancer cells as a model, we demonstrate that an increase in mitochondrial superoxide dismutase SOD2 protein expression is a very early event initiated in response to detachment, an important step during metastasis that has been associated with increased oxidative stress. SOD2 protein synthesis is rapidly induced within 0.5-2 h of matrix detachment, and polyribosome profiling demonstrates an increase in the number of ribosomes bound to SOD2 mRNA, indicating an increase in SOD2 mRNA translation in response to anchorage-independence. Mechanistically, we find that anchorage-independence induces cytosolic accumulation of the RNA binding protein HuR/ELAVL1 and promotes HuR binding to SOD2 mRNA. Using HuR siRNA-mediated knockdown, we show that the presence of HuR is necessary for the increase in SOD2 mRNA association with the heavy polyribosome fraction and consequent nascent SOD2 protein synthesis in anchorage-independence. Cellular detachment also activates the stress-response mitogen-activated kinase p38, which is necessary for HuR-SOD2 mRNA interactions and induction of SOD2 protein output. These findings illustrate a novel translational regulatory mechanism of SOD2 by which ovarian cancer cells rapidly increase their mitochondrial antioxidant capacity as an acute stress response to anchorage-independence.
Collapse
Affiliation(s)
- Yeon Soo Kim
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Priscilla W Tang
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Jaclyn E Welles
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Weihua Pan
- Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Zaineb Javed
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Amal Taher Elhaw
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Karthikeyan Mythreye
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Nadine Hempel
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Jiang Y, Krantz S, Qin X, Li S, Gunasekara H, Kim YM, Zimnicka A, Bae M, Ma K, Toth PT, Hu Y, Shajahan-Haq AN, Patel HH, Gentile S, Bonini MG, Rehman J, Liu Y, Minshall RD. Caveolin-1 controls mitochondrial damage and ROS production by regulating fission - fusion dynamics and mitophagy. Redox Biol 2022; 52:102304. [PMID: 35413643 PMCID: PMC9018165 DOI: 10.1016/j.redox.2022.102304] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022] Open
Abstract
As essential regulators of mitochondrial quality control, mitochondrial dynamics and mitophagy play key roles in maintenance of metabolic health and cellular homeostasis. Here we show that knockdown of the membrane-inserted scaffolding and structural protein caveolin-1 (Cav-1) and expression of tyrosine 14 phospho-defective Cav-1 mutant (Y14F), as opposed to phospho-mimicking Y14D, altered mitochondrial morphology, and increased mitochondrial matrix mixing, mitochondrial fusion and fission dynamics as well as mitophagy in MDA-MB-231 triple negative breast cancer cells. Further, we found that interaction of Cav-1 with mitochondrial fusion/fission machinery Mitofusin 2 (Mfn2) and Dynamin related protein 1 (Drp1) was enhanced by Y14D mutant indicating Cav-1 Y14 phosphorylation prevented Mfn2 and Drp1 translocation to mitochondria. Moreover, limiting mitochondrial recruitment of Mfn2 diminished formation of the PINK1/Mfn2/Parkin complex required for initiation of mitophagy resulting in accumulation of damaged mitochondria and ROS (mtROS). Thus, these studies indicate that phospho-Cav-1 may be an important switch mechanism in cancer cell survival which could lead to novel strategies for complementing cancer therapies.
Collapse
Affiliation(s)
- Ying Jiang
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA; Center for Informational Biology, University of Electronic Science and Technology of China, 610054, China
| | - Sarah Krantz
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Xiang Qin
- Center for Informational Biology, University of Electronic Science and Technology of China, 610054, China
| | - Shun Li
- Center for Informational Biology, University of Electronic Science and Technology of China, 610054, China
| | | | - Young-Mee Kim
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Adriana Zimnicka
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Misuk Bae
- Anesthesiology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ke Ma
- Research Resources Center, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Peter T Toth
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA; Research Resources Center, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ying Hu
- Chemistry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Hemal H Patel
- VA San Diego Health System and Department of Anesthesiology, University of California at San Diego, San Diego, CA, 92161, USA
| | - Saverio Gentile
- Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Marcelo G Bonini
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60614, USA
| | - Jalees Rehman
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA; Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yiyao Liu
- Center for Informational Biology, University of Electronic Science and Technology of China, 610054, China
| | - Richard D Minshall
- Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA; Anesthesiology, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
SOD2, a Potential Transcriptional Target Underpinning CD44-Promoted Breast Cancer Progression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030811. [PMID: 35164076 PMCID: PMC8839817 DOI: 10.3390/molecules27030811] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022]
Abstract
CD44, a cell-adhesion molecule has a dual role in tumor growth and progression; it acts as a tumor suppressor as well as a tumor promoter. In our previous work, we developed a tetracycline-off regulated expression of CD44's gene in the breast cancer (BC) cell line MCF-7 (B5 clone). Using cDNA oligo gene expression microarray, we identified SOD2 (superoxide dismutase 2) as a potential CD44-downstream transcriptional target involved in BC metastasis. SOD2 gene belongs to the family of iron/manganese superoxide dismutase family and encodes a mitochondrial protein. SOD2 plays a role in cell proliferation and cell invasion via activation of different signaling pathways regulating angiogenic abilities of breast tumor cells. This review will focus on the findings supporting the underlying mechanisms associated with the oncogenic potential of SOD2 in the onset and progression of cancer, especially in BC and the potential clinical relevance of its various inhibitors.
Collapse
|
11
|
Al Haq AT, Tseng HY, Chen LM, Wang CC, Hsu HL. Targeting prooxidant MnSOD effect inhibits triple-negative breast cancer (TNBC) progression and M2 macrophage functions under the oncogenic stress. Cell Death Dis 2022; 13:49. [PMID: 35017469 PMCID: PMC8752602 DOI: 10.1038/s41419-021-04486-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/23/2021] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
Abstract
Triple-negative breast cancer (TNBC) has been shown with high mitochondrial oxidative phosphorylation and production of reactive oxygen species (ROS). MnSOD (SOD2) is a mitochondrial antioxidant defense that has been implicated in inhibition of human malignancies. However, the impact of MnSOD on immunosuppressive macrophage functions and TNBC aggressiveness has never been explored. We found here that SOD2high is primarily observed in the aggressive subtypes of HER2(+) breast cancers and TNBCs patients. Further analyses demonstrated that the oncoprotein multiple copies in T-cell malignancy-1 (MCT-1 or MCTS1) induces mitochondrial superoxide dismutase (MnSOD) in TNBC cells by stabilizing the transcription factor Nrf2. SOD2high/MCTS1high expression correlates with a poor prognosis in breast cancer patients. MnSOD in TNBC cells functions as a prooxidant peroxidase that increases mitochondrial ROS (mROS) and adaptation to oxidative stress under the oncogenic effect. Interleukin-6 (IL-6) in the MCT-1 pathway elevates Nrf2/MnSOD and mROS levels. Knockdown of MnSOD inhibits TNBC cell invasion, breast cancer stem cells (BCSCs), mROS, and IL-6 excretion promoted by MCT-1. TNBC cells deficient in MnSOD prevent the polarization and chemotaxis of M2 macrophages but improve the ability of M1 macrophages to engulf cancer cells. Quenching mROS with MitoQ, a mitochondria-targeted non-metal-based antioxidant MnSOD mimics, effectively suppresses BCSCs and M2 macrophage invasion exacerbated by MnSOD and MCT-1. Consistently, silencing MnSOD impedes TNBC progression and intratumoral M2 macrophage infiltration. We revealed a novel stratagem for TNBC management involving targeting the MCT-1 oncogene-induced mitochondrial prooxidant MnSOD pathway, which prevents the development of an immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Aushia Tanzih Al Haq
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan.,Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Hong-Yu Tseng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Mei Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Chien-Chia Wang
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Hsin-Ling Hsu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan.
| |
Collapse
|
12
|
Zhu Y, Qiu Y, Zhang X. TKTL1 participated in malignant progression of cervical cancer cells via regulating AKT signal mediated PFKFB3 and thus regulating glycolysis. Cancer Cell Int 2021; 21:678. [PMID: 34922556 PMCID: PMC8684167 DOI: 10.1186/s12935-021-02383-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cervical cancer (CC) is the second most common cancer among women with high morbidity and mortality. TKTL1 is a key protein in glucose metabolism in cancer cells and controls the pentose phosphate pathway (PPP). In this paper, we aim to explore whether TKTL1 can participate in the malignant process of CC cells through glucose metabolism. METHODS The expression and activity of TKTL1 in CC cell lines were detected by RT-qPCR and Western blot. Cell transfection was conducted to interfere the expression of TKTL1 in SiHa cells, with efficiency detected by RT-qPCR and Western blot. Cell proliferation was then measured by CCK-8 kits. Wound Healing and Transwell experiments were performed to respectively detect the levels of cell migration and invasion, and western blot was used to detect the expressions of migration-related proteins. Tunel and Western blot were used to detect the apoptosis and apoptosis-related proteins. Glucose uptake, lactate production, and ATP production were measured by corresponding commercial kits. Next, the expression of p-Akt, AKT, p-MTOR, mTOR, HK2 and PFKFB3 was detected by Western blot. The mechanism was further investigated by interfering the expression of HK2 and PFKFB3 and adding AKT agonist SC79. At the animal level, the tumor bearing mouse model of CC was constructed, and the weight, volume and pathological morphology of the tumor tissue were detected to verify the cell experiment. RESULTS TKTL1 expression was increased in CC cells. Interference of TKTL1 expression can inhibit TKTL1 enzyme activity, proliferation, invasion and migration of CC cells, and simultaneously suppress the generation of glycolysis. In addition, the results showed that TKTL1 activated PFKFB3 through AKT rather than HK2 signaling and is involved in glycolysis, cell invasion, migration, and apoptosis of CC cells. In animal level, inhibition of TKTL1 also contributed to decreased tumor volume of CC tumor bearing mice and improved histopathological status. CONCLUSION TKTL1 participated in malignant progression of CC cells via regulating AKT signal-mediated HK2 and PFKFB3 and thus regulating glucose metabolism.
Collapse
Affiliation(s)
- Yingping Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310006, Zhejiang, China
| | - Yu Qiu
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, NO.10 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China.
| | - Xueqin Zhang
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, NO.10 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China.
| |
Collapse
|
13
|
Mathew R. Critical Role of Caveolin-1 Loss/Dysfunction in Pulmonary Hypertension. Med Sci (Basel) 2021; 9:medsci9040058. [PMID: 34698188 PMCID: PMC8544475 DOI: 10.3390/medsci9040058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/17/2021] [Accepted: 09/16/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a rare disease with a high morbidity and mortality rate. A number of systemic diseases and genetic mutations are known to lead to PH. The main features of PH are altered vascular relaxation responses and the activation of proliferative and anti-apoptotic pathways, resulting in pulmonary vascular remodeling, elevated pulmonary artery pressure, and right ventricular hypertrophy, ultimately leading to right heart failure and premature death. Important advances have been made in the field of pulmonary pathobiology, and several deregulated signaling pathways have been shown to be associated with PH. Clinical and experimental studies suggest that, irrespective of the underlying disease, endothelial cell disruption and/or dysfunction play a key role in the pathogenesis of PH. Endothelial caveolin-1, a cell membrane protein, interacts with and regulates several transcription factors and maintains homeostasis. Disruption of endothelial cells leads to the loss or dysfunction of endothelial caveolin-1, resulting in reciprocal activation of proliferative and inflammatory pathways, leading to cell proliferation, medial hypertrophy, and PH, which initiates PH and facilitates its progression. The disruption of endothelial cells, accompanied by the loss of endothelial caveolin-1, is accompanied by enhanced expression of caveolin-1 in smooth muscle cells (SMCs) that leads to pro-proliferative and pro-migratory responses, subsequently leading to neointima formation. The neointimal cells have low caveolin-1 and normal eNOS expression that may be responsible for promoting nitrosative and oxidative stress, furthering cell proliferation and metabolic alterations. These changes have been observed in human PH lungs and in experimental models of PH. In hypoxia-induced PH, there is no endothelial disruption, loss of endothelial caveolin-1, or enhanced expression of caveolin-1 in SMCs. Hypoxia induces alterations in membrane composition without caveolin-1 or any other membrane protein loss. However, caveolin-1 is dysfunctional, resulting in cell proliferation, medial hypertrophy, and PH. These alterations are reversible upon removal of hypoxia, provided there is no associated EC disruption. This review examined the role of caveolin-1 disruption and dysfunction in PH.
Collapse
Affiliation(s)
- Rajamma Mathew
- Section of Pediatric Cardiology, Departments of Pediatrics and Physiology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
14
|
Mustafa MF, Saliluddin SM, Fakurazi S, Tizen Laim NMS, Md Pauzi SH, Nik Yahya NH, S Raja Gopal N, Abdullah MA, Maniam S. Expression of Autophagy and Mitophagy Markers in Breast Cancer Tissues. Front Oncol 2021; 11:612009. [PMID: 34490076 PMCID: PMC8416475 DOI: 10.3389/fonc.2021.612009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/21/2021] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play important roles in regulating cell bioenergetics status and reactive oxygen species (ROS) generation. ROS-induced mitochondrial damage is among the main intracellular signal inducers of autophagy. Autophagy is a cellular catabolic process that regulates protein and organelle turnover, while a selective form of autophagy, mitophagy, specifically targets dysfunctional mitochondrial degradation. This study aims to measure the levels of autophagy, mitophagy, oxidative stress, and apoptosis in invasive breast carcinoma tissues using immunohistochemistry (IHC). Tissue microarrays of 76 patients with breast cancer were stained with six IHC markers (MnSOD, Beclin-1, LC3, BNIP3, Parkin, and cleaved caspase 3). The expression intensity was determined for each tumor tissue and the adjacent tumor-matched control tissues. Intermediate and strong staining scores of MnSOD, Beclin-1, LC-3, BNIP-3, and Parkin were significantly higher in tumor tissues compared to the adjacent matched control. The scoring intensity was further classified into tissues with negative staining and positive staining, which showed that positive scores of Beclin-1 and Parkin were significantly high in tumor tissues compared to other markers. Positive association was also noted between BNIP-3 and Beclin-1 as well as LC-3 and cleaved caspase-3 immunostaining. To our knowledge, this is one of the first studies that measure both mitophagy and autophagy in the same breast cancer tissues and the adjacent matched control. The findings from this study will be of great potential in identifying new cancer biomarkers and inspire significant interest in applying anti-autophagy therapies as a possible treatment for breast cancer.
Collapse
Affiliation(s)
- Mohd Fazirul Mustafa
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| | - Suhainizam Muhamad Saliluddin
- Department of Community Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| | | | - Suria Hayati Md Pauzi
- Department of Pathology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | | | | | - Maizaton Atmadini Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| | - Sandra Maniam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
15
|
Johar D, Elmehrath AO, Khalil RM, Elberry MH, Zaky S, Shalabi SA, Bernstein LH. Protein networks linking Warburg and reverse Warburg effects to cancer cell metabolism. Biofactors 2021; 47:713-728. [PMID: 34453457 DOI: 10.1002/biof.1768] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
It was 80 years after the Otto Warburg discovery of aerobic glycolysis, a major hallmark in the understanding of cancer. The Warburg effect is the preference of cancer cell for glycolysis that produces lactate even when sufficient oxygen is provided. "reverse Warburg effect" refers to the interstitial tissue communications with adjacent epithelium, that in the process of carcinogenesis, is needed to be explored. Among these cell-cell communications, the contact between epithelial cells; between epithelial cells and matrix; and between fibroblasts and inflammatory cells in the underlying matrix. Cancer involves dysregulation of Warburg and reverse Warburg cellular metabolic pathways. How these gene and protein-based regulatory mechanisms have functioned has been the basis for this review. The importance of the Warburg in oxidative phosphorylation suppression, with increased glycolysis in cancer growth and proliferation is emphasized. Studies that are directed at pathways that would be expected to shift cell metabolism to an increased oxidation and to a decrease in glycolysis are emphasized. Key enzymes required for oxidative phosphorylation, and affect the inhibition of fatty acid metabolism and glutamine dependence are conferred. The findings are of special interest to cancer pharmacotherapy. Studies described in this review are concerned with the effects of therapeutic modalities that are intimately related to the Warburg effect. These interactions described may be helpful as adjuvant therapy in controlling the process of proliferation and metastasis.
Collapse
Affiliation(s)
- Dina Johar
- Department of Biochemistry and Nutrition, Faculty of Women for Arts, Sciences and Education, Ain Shams University, Heliopolis, Cairo, Egypt
| | | | - Rania M Khalil
- Department of Biochemistry, Pharmacy College, Delta University for Science and Technology, Gamasa, Egypt
| | - Mostafa H Elberry
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Samy Zaky
- Hepatogastroenterology and Infectious Diseases, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Samy A Shalabi
- Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Consultant Pathologist, Kuwait, Kuwait
| | - Larry H Bernstein
- Emeritus Prof. Department of Pathology, Yale University, Connecticut, USA
- Triplex Consulting Pharmaceuticals, 54 Firethorn Lane Northampton, MA 01060, USA
| |
Collapse
|
16
|
Involvement of NRF2 in Breast Cancer and Possible Therapeutical Role of Polyphenols and Melatonin. Molecules 2021; 26:molecules26071853. [PMID: 33805996 PMCID: PMC8038098 DOI: 10.3390/molecules26071853] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is defined as a disturbance in the prooxidant/antioxidant balance in favor of the former and a loss of control over redox signaling processes, leading to potential biomolecular damage. It is involved in the etiology of many diseases, varying from diabetes to neurodegenerative diseases and cancer. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor and reported as one of the most important oxidative stress regulators. Due to its regulatory role in the expression of numerous cytoprotective genes involved in the antioxidant and anti-inflammatory responses, the modulation of NRF2 seems to be a promising approach in the prevention and treatment of cancer. Breast cancer is the prevalent type of tumor in women and is the leading cause of death among female cancers. Oxidative stress-related mechanisms are known to be involved in breast cancer, and therefore, NRF2 is considered to be beneficial in its prevention. However, its overactivation may lead to a negative clinical impact on breast cancer therapy by causing chemoresistance. Some known “oxidative stress modulators”, such as melatonin and polyphenols, are suggested to play an important role in the prevention and treatment of cancer, where the activation of NRF2 is reported as a possible underlying mechanism. In the present review, the potential involvement of oxidative stress and NRF2 in breast cancer will be reviewed, and the role of the NRF2 modulators—namely, polyphenols and melatonin—in the treatment of breast cancer will be discussed.
Collapse
|
17
|
Abstract
Cellular senescence is a feature of most somatic cells. It is characterized by an irreversible cell cycle arrest and by the ability to secrete a plethora of mediators of inflammation and growth factors, which can alter the senescent cell's microenvironment. Senescent cells accumulate in tissues over time and contribute to both aging and the development of age-associated diseases. Senescent cells have antagonistic pleiotropic roles in cancer. Given the inability of senescent cells to proliferate, cellular senescence is a powerful tumor suppressor mechanism in young individuals. However, accumulation of senescent stromal cells during aging can fuel cancer cell growth in virtue of their capacity to release factors that stimulate cell proliferation. Caveolin-1 is a structural protein component of caveolae, invaginations of the plasma membrane involved in a variety of cellular processes, including signal transduction. Mounting evidence over the last 10-15 years has demonstrated a central role of caveolin-1 in the development of a senescent phenotype and the regulation of both the anti-tumorigenic and pro-tumorigenic properties of cellular senescence. In this review, we discuss the cellular mechanisms and functions of caveolin-1 in the context of cellular senescence and their relevance to the biology of cancer.
Collapse
|
18
|
Lolo FN, Jiménez-Jiménez V, Sánchez-Álvarez M, Del Pozo MÁ. Tumor-stroma biomechanical crosstalk: a perspective on the role of caveolin-1 in tumor progression. Cancer Metastasis Rev 2021; 39:485-503. [PMID: 32514892 DOI: 10.1007/s10555-020-09900-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.
Collapse
Affiliation(s)
- Fidel Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
19
|
Jaganjac M, Milkovic L, Sunjic SB, Zarkovic N. The NRF2, Thioredoxin, and Glutathione System in Tumorigenesis and Anticancer Therapies. Antioxidants (Basel) 2020; 9:E1151. [PMID: 33228209 PMCID: PMC7699519 DOI: 10.3390/antiox9111151] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer remains an elusive, highly complex disease and a global burden. Constant change by acquired mutations and metabolic reprogramming contribute to the high inter- and intratumor heterogeneity of malignant cells, their selective growth advantage, and their resistance to anticancer therapies. In the modern era of integrative biomedicine, realizing that a personalized approach could benefit therapy treatments and patients' prognosis, we should focus on cancer-driving advantageous modifications. Namely, reactive oxygen species (ROS), known to act as regulators of cellular metabolism and growth, exhibit both negative and positive activities, as do antioxidants with potential anticancer effects. Such complexity of oxidative homeostasis is sometimes overseen in the case of studies evaluating the effects of potential anticancer antioxidants. While cancer cells often produce more ROS due to their increased growth-favoring demands, numerous conventional anticancer therapies exploit this feature to ensure selective cancer cell death triggered by excessive ROS levels, also causing serious side effects. The activation of the cellular NRF2 (nuclear factor erythroid 2 like 2) pathway and induction of cytoprotective genes accompanies an increase in ROS levels. A plethora of specific targets, including those involved in thioredoxin (TRX) and glutathione (GSH) systems, are activated by NRF2. In this paper, we briefly review preclinical research findings on the interrelated roles of the NRF2 pathway and TRX and GSH systems, with focus given to clinical findings and their relevance in carcinogenesis and anticancer treatments.
Collapse
Affiliation(s)
| | | | | | - Neven Zarkovic
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia; (M.J.); (L.M.); (S.B.S.)
| |
Collapse
|
20
|
Smolková K, Mikó E, Kovács T, Leguina-Ruzzi A, Sipos A, Bai P. Nuclear Factor Erythroid 2-Related Factor 2 in Regulating Cancer Metabolism. Antioxid Redox Signal 2020; 33:966-997. [PMID: 31989830 PMCID: PMC7533893 DOI: 10.1089/ars.2020.8024] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Nuclear factor erythroid 2 (NFE2)-related factor 2 (NFE2L2, or NRF2) is a transcription factor predominantly affecting the expression of antioxidant genes. NRF2 plays a significant role in the control of redox balance, which is crucial in cancer cells. NRF2 activation regulates numerous cancer hallmarks, including metabolism, cancer stem cell characteristics, tumor aggressiveness, invasion, and metastasis formation. We review the molecular characteristics of the NRF2 pathway and discuss its interactions with the cancer hallmarks previously listed. Recent Advances: The noncanonical activation of NRF2 was recently discovered, and members of this pathway are involved in carcinogenesis. Further, cancer-related changes (e.g., metabolic flexibility) that support cancer progression were found to be redox- and NRF2 dependent. Critical Issues: NRF2 undergoes Janus-faced behavior in cancers. The pro- or antineoplastic effects of NRF2 are context dependent and essentially based on the specific molecular characteristics of the cancer in question. Therefore, systematic investigation of NRF2 signaling is necessary to clarify its role in cancer etiology. The biggest challenge in the NRF2 field is to determine which cancers can be targeted for better clinical outcomes. Further, large-scale genomic and transcriptomic studies are missing to correlate the clinical outcome with the activity of the NRF2 system. Future Directions: To exploit NRF2 in a clinical setting in the future, the druggable members of the NRF2 pathway should be identified. In addition, it will be important to study how the modulation of the NRF2 system interferes with cytostatic drugs and their combinations.
Collapse
Affiliation(s)
- Katarína Smolková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alberto Leguina-Ruzzi
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary.,Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
21
|
Danes JM, de Abreu ALP, Kerketta R, Huang Y, Palma FR, Gantner BN, Mathison AJ, Urrutia RA, Bonini MG. Inorganic arsenic promotes luminal to basal transition and metastasis of breast cancer. FASEB J 2020; 34:16034-16048. [PMID: 33047385 DOI: 10.1096/fj.202001192r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022]
Abstract
Inorganic arsenic (iAs/As2 O3 2- ) is an environmental toxicant found in watersheds around the world including in densely populated areas. iAs is a class I carcinogen known to target the skin, lungs, bladder, and digestive organs, but its role as a primary breast carcinogen remains controversial. Here, we examined a different possibility: that exposure to iAs promotes the transition of well-differentiated epithelial breast cancer cells characterized by estrogen and progesterone receptor expression (ER+/PR+), to more basal phenotypes characterized by active proliferation, and propensity to metastasis in vivo. Our results indicate two clear phenotypic responses to low-level iAs that depend on the duration of the exposure. Short-term pulses of iAs activate ER signaling, consistent with its reported pseudo-estrogen activity, but longer-term, chronic treatments for over 6 months suppresses both ER and PR expression and signaling. In fact, washout of these chronically exposed cells for up to 1 month failed to fully reverse the transcriptional and phenotypic effects of prolonged treatments, indicating durable changes in cellular physiologic identity. RNA-seq studies found that chronic iAs drives the transition toward more basal phenotypes characterized by impaired hormone receptor signaling despite the conservation of estrogen receptor expression. Because treatments for breast cancer patients are largely designed based on the detection of hormone receptor expression, our results suggest greater scrutiny of ER+ cancers in patients exposed to iAs, because these tumors may spawn more aggressive phenotypes than unexposed ER+ tumors, in particular, basal subtypes that tend to develop therapy resistance and metastasis.
Collapse
Affiliation(s)
- Jeanne M Danes
- Division of Hematology Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andre L P de Abreu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Romica Kerketta
- Genomic Sciences and Precision Medicine Center (GSPMC), Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yunping Huang
- Division of Hematology Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Flavio R Palma
- Division of Hematology Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin N Gantner
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Angela J Mathison
- Genomic Sciences and Precision Medicine Center (GSPMC), Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Raul A Urrutia
- Genomic Sciences and Precision Medicine Center (GSPMC), Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marcelo G Bonini
- Division of Hematology Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
22
|
Wang Y, Lin Z, Song J, Wei S, Ye Z, Chen S, Zeng Y, Lin Z, Chen X, Chen L. MicroRNA-451a targets caveolin-1 in stomach cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2524-2533. [PMID: 33165443 PMCID: PMC7642723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/29/2020] [Indexed: 06/11/2023]
Abstract
MicroRNA-451 (miR-451) is lowly expressed in stomach cancer cells and improves their metastatic ability by down-regulating extracellular signal-regulated kinase 2 (ERK2). Many studies have found that caveolin-1 (CAV1) plays an important role in cancer progression. Additionally, miR-451 has been reported to regulate the expression of CAV1 in chronic obstructive pulmonary disease. Therefore, this study aims to determine if miR-451 regulates the biological functions of stomach cancer cells by regulating CAV1 expression. Through a bioinformatics analysis, we found that miR-451a regulates CAV1 expression, and miR-451a expression is relatively low in stomach cancer cells. Next, we confirmed that miR-451a negatively regulates CAV1 expression using a dual-luciferase reporter assay. Then MTT, 5-ethynyl-2'-deoxyuridine (EdU), propidium iodide (PI), an Annexin V-FITC/PI apoptosis kit, and transwell assays were used to measure the changes in cell proliferation, the cell cycle, apoptosis, cell migration, and invasiveness in stomach cancer cells overexpressing miR-451a or both miR-451a and CAV1. It was found that increasing the miR-451a expression in stomach cancer cells inhibits cell growth, migration, and invasiveness, and promotes apoptosis. After restoring the CAV1 expression, these biological processes resumed. In summary, in stomach cancer cells, the overexpression of miR-451a can restrain cell growth and promote apoptosis, so it is a potential treatment for stomach cancer.
Collapse
Affiliation(s)
- Yi Wang
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Zhenmeng Lin
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Jintian Song
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Shenghong Wei
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Zaisheng Ye
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Shu Chen
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Yi Zeng
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Zhitao Lin
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Xiaoling Chen
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Luchuan Chen
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| |
Collapse
|
23
|
De Nuccio C, Bernardo A, Troiano C, Brignone MS, Falchi M, Greco A, Rosini M, Basagni F, Lanni C, Serafini MM, Minghetti L, Visentin S. NRF2 and PPAR-γ Pathways in Oligodendrocyte Progenitors: Focus on ROS Protection, Mitochondrial Biogenesis and Promotion of Cell Differentiation. Int J Mol Sci 2020; 21:E7216. [PMID: 33003644 PMCID: PMC7583077 DOI: 10.3390/ijms21197216] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
An adequate protection from oxidative and inflammatory reactions, together with the promotion of oligodendrocyte progenitor (OP) differentiation, is needed to recover from myelin damage in demyelinating diseases. Mitochondria are targets of inflammatory and oxidative insults and are essential in oligodendrocyte differentiation. It is known that nuclear factor-erythroid 2-related factor/antioxidant responsive element (NRF2/ARE) and peroxisome proliferator-activated receptor gamma/PPAR-γ response element (PPAR-γ/PPRE) pathways control inflammation and overcome mitochondrial impairment. In this study, we analyzed the effects of activators of these pathways on mitochondrial features, protection from inflammatory/mitochondrial insults and cell differentiation in OP cultures, to depict the specificities and similarities of their actions. We used dimethyl-fumarate (DMF) and pioglitazone (pio) as agents activating NRF2 and PPAR-γ, respectively, and two synthetic hybrids acting differently on the NRF2/ARE pathway. Only DMF and compound 1 caused early effects on the mitochondria. Both DMF and pio induced mitochondrial biogenesis but different antioxidant repertoires. Moreover, pio induced OP differentiation more efficiently than DMF. Finally, DMF, pio and compound 1 protected from tumor necrosis factor-alpha (TNF-α) insult, with pio showing faster kinetics of action and compound 1 a higher activity than DMF. In conclusion, NRF2 and PPAR-γ by inducing partially overlapping pathways accomplish complementary functions aimed at the preservation of mitochondrial function, the defense against oxidative stress and the promotion of OP differentiation.
Collapse
Affiliation(s)
- Chiara De Nuccio
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Antonietta Bernardo
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| | - Carmen Troiano
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | | | - Mario Falchi
- National Research Center on HIV/AIDS, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anita Greco
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Filippo Basagni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Cristina Lanni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (C.L.); (M.M.S.)
| | | | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| |
Collapse
|
24
|
Dees S, Pontiggia L, Jasmin JF, Sotgia F, Lisanti MP, Mercier I. Essential role of STAT5a in DCIS formation and invasion following estrogen treatment. Aging (Albany NY) 2020; 12:15104-15120. [PMID: 32633727 PMCID: PMC7425506 DOI: 10.18632/aging.103586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/10/2020] [Indexed: 11/25/2022]
Abstract
Ductal carcinoma in situ (DCIS) is one of the earliest stages of breast cancer (BCa). The mechanisms by which DCIS lesions progress to an invasive state while others remain indolent are yet to be fully characterized and both diagnosis and treatment of this pre-invasive disease could benefit from better understanding the pathways involved. While a decreased expression of Caveolin-1 (Cav-1) in the tumor microenvironment of patients with DCIS breast cancer was linked to progression to invasive breast cancer (IBC), the downstream effector(s) contributing to this process remain elusive. The current report shows elevated expression of Signal Transducer and Activator of Transcription 5a (STAT5a) within the DCIS-like lesions in Cav-1 KO mice following estrogen treatment and inhibition of STAT5a expression prevented the formation of these mammary lesions. In addition, STAT5a overexpression in a human DCIS cell line (MCF10DCIS.com) promoted their invasion, a process accelerated by estrogen treatment and associated with increased levels of the matrix metalloproteinase-9 (MMP-9) precursor. In sum, our results demonstrate a novel regulatory axis (Cav-1♦STAT5a♦MMP-9) in DCIS that is fully activated by the presence of estrogen. Our sudies suggest to further study phosphorylated STAT5a (Y694) as a potential biomarker to guide and predict outcome of DCIS patient population.
Collapse
Affiliation(s)
- Sundee Dees
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Laura Pontiggia
- Department of Mathematics, Physics and Statistics, Misher College of Arts and Sciences, University of the Sciences, Philadelphia, PA, USA
| | - Jean-Francois Jasmin
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Federica Sotgia
- Translational Medicine, School of Science, Engineering and Environment (SEE), Biomedical Research Centre (BRC), University of Salford, Greater Manchester, United Kingdom
| | - Michael P. Lisanti
- Translational Medicine, School of Science, Engineering and Environment (SEE), Biomedical Research Centre (BRC), University of Salford, Greater Manchester, United Kingdom
| | - Isabelle Mercier
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
- Program in Personalized Medicine and Targeted Therapeutics, University of the Sciences, Philadelphia, PA, USA
| |
Collapse
|
25
|
Caveolin-1 inhibits breast cancer stem cells via c-Myc-mediated metabolic reprogramming. Cell Death Dis 2020; 11:450. [PMID: 32528105 PMCID: PMC7290025 DOI: 10.1038/s41419-020-2667-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 12/31/2022]
Abstract
Breast cancer stem cells (BCSCs) are considered to be the root of breast cancer occurrence and progression. However, the characteristics and regulatory mechanisms of BCSCs metabolism have been poorly revealed, which hinders the development of metabolism-targeted treatment strategies for BCSCs elimination. Herein, we demonstrated that the downregulation of Caveolin-1 (Cav-1) usually occurred in BCSCs and was associated with a metabolic switch from mitochondrial respiration to aerobic glycolysis. Meanwhile, Cav-1 could inhibit the self-renewal capacity and aerobic glycolysis activity of BCSCs. Furthermore, Cav-1 loss was associated with accelerated mammary-ductal hyperplasia and mammary-tumor formation in transgenic mice, which was accompanied by enrichment and enhanced aerobic glycolysis activity of BCSCs. Mechanistically, Cav-1 could promote Von Hippel-Lindau (VHL)-mediated ubiquitination and degradation of c-Myc in BCSCs through the proteasome pathway. Notably, epithelial Cav-1 expression significantly correlated with a better overall survival and delayed onset age of breast cancer patients. Together, our work uncovers the characteristics and regulatory mechanisms of BCSCs metabolism and highlights Cav-1-targeted treatments as a promising strategy for BCSCs elimination.
Collapse
|
26
|
Mathew R, Huang J, Iacobas S, Iacobas DA. Pulmonary Hypertension Remodels the Genomic Fabrics of Major Functional Pathways. Genes (Basel) 2020; 11:genes11020126. [PMID: 31979420 PMCID: PMC7074533 DOI: 10.3390/genes11020126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary hypertension (PH) is a serious disorder with high morbidity and mortality rate. We analyzed the right-ventricular systolic pressure (RVSP), right-ventricular hypertrophy (RVH), lung histology, and transcriptomes of six-week-old male rats with PH induced by (1) hypoxia (HO), (2) administration of monocrotaline (CM), or (3) administration of monocrotaline and exposure to hypoxia (HM). The results in PH rats were compared to those in control rats (CO). After four weeks exposure, increased RVSP and RVH, pulmonary arterial wall thickening, and alteration of the lung transcriptome were observed in all PH groups. The HM group exhibited the largest alterations, as well as neointimal lesions and obliteration of the lumen in small arteries. We found that PH increased the expression of caveolin1, matrix metallopeptidase 2, and numerous inflammatory and cell proliferation genes. The cell cycle, vascular smooth muscle contraction, and oxidative phosphorylation pathways, as well as their interplay, were largely perturbed. Our results also suggest that the upregulated Rhoa (Ras homolog family member A) mediates its action through expression coordination with several ATPases. The upregulation of antioxidant genes and the extensive mitochondrial damage observed, especially in the HM group, indicate metabolic shift toward aerobic glycolysis.
Collapse
Affiliation(s)
- Rajamma Mathew
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA; (R.M.); (J.H.)
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| | - Jing Huang
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA; (R.M.); (J.H.)
| | - Sanda Iacobas
- Department of Pathology, New York Medical College, Valhalla, NY 10595, USA
| | - Dumitru A. Iacobas
- Personalized Genomics Laboratory, Center for Computational Systems Biology, Roy G Perry College of Engineering, Prairie View A&M University, Prairie View, TX 77446, USA
- Correspondence: ; Tel.: +1-936-261-9926
| |
Collapse
|
27
|
Zheng N, Wang T, Wei A, Chen W, Zhao C, Li H, Wang L. High-content analysis boosts identification of the initial cause of triptolide-induced hepatotoxicity. J Appl Toxicol 2019; 39:1337-1347. [PMID: 31218727 DOI: 10.1002/jat.3821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/21/2022]
Abstract
Triptolide (TP) has been widely used in China for more than 40 years as an immunosuppressive agent. Recently, serious concerns have been raised over TP-induced liver injury, though the real hepatotoxic mechanism is still unclear, particularly in terms of the initial cause. To our knowledge, this study is the first to screen systematically the mechanism of TP-induced toxicity through a global cytotoxicity profile high-content analysis using three independent cytotoxic assay panels with multiple endpoints of cytotoxicity, including cell loss, mitochondrial membrane potential, nuclear membrane permeability, manganese superoxide dismutase, phosphorylated gamma-H2AX, light chain 3B, lysosome, reactive oxygen species and glutathione. We assessed nine parameters and four stress response pathway models by labeling nuclear factor erythroid 2-related factor 2, activating transcription factor 6, hypoxia inducible factor 1α and nuclear factor κB and found that all testing parameters except glutathione and manganese superoxide dismutase showed concentration- and time-dependent changes, as well as increased cell loss after TP treatment. Considering that RNA polymerase II is the molecular target of TP, we quantified transcription from inducible genes, bromodeoxyuridine incorporation, and expression from transiently transfected green fluorescence protein plasmids in HepG2 cells. The results show that inhibition of global transcription by TP took place at earlier times and at lower concentrations than those observed for cell death. Therefore, global transcriptional suppression and the cell dysfunction it drives play a central role in TP-induced hepatotoxicity. This provides valuable information for the safe use of TP in the clinic.
Collapse
Affiliation(s)
- Nan Zheng
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Tiantian Wang
- Institute of Pharmacology and Toxicology, Beijing, China.,Key Laboratory of Cell Proliferation and Regulation Biology, Beijing Normal University, Beijing, China
| | - Aili Wei
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Wei Chen
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Changqi Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Beijing Normal University, Beijing, China
| | - Hua Li
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Lili Wang
- Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| |
Collapse
|
28
|
Ansaryan S, Khayamian MA, Saghafi M, Shalileh S, Nikshoar MS, Abbasvandi F, Mahmoudi M, Bahrami F, Abdolahad M. Stretch Induces Invasive Phenotypes in Breast Cells Due to Activation of Aerobic-Glycolysis-Related Pathways. ACTA ACUST UNITED AC 2019; 3:e1800294. [PMID: 32648669 DOI: 10.1002/adbi.201800294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 03/22/2019] [Indexed: 12/19/2022]
Abstract
It is increasingly being accepted that cells' physiological functions are substantially dependent on the mechanical characteristics of their surrounding tissue. This is mainly due to the key role of biomechanical forces on cells and their nucleus' shapes, which have the capacity to regulate chromatin conformation and thus gene regulations. Therefore, it is reasonable to postulate that altering the biomechanical properties of tissue may have the capacity to change cell functions. Here, the role of cell stretching (as a model of biomechanical variations) is probed in cell migration and invasion capacity using human normal and cancerous breast cells. By several analyses (i.e., scratch assay, invasion to endothelial barrier, real-time RNA sequencing, confocal imaging, patch clamp, etc.), it is revealed that the cell-stretching process could increase the migration and invasion capabilities of normal and cancerous cells, respectively. More specifically, it is found that poststretched breast cancer cells are found in low grades of invasion; they substantially upregulate the expression of manganese-dependent superoxide dismutase (MnSOD) through activation of H-Ras proteins, which subsequently induce aerobic glycolysis followed by an overproduction of matrix metalloproteinases (MMP)-reinforced filopodias. Presence of such invadopodias facilitates targeting of the endothelial layer, and increased invasive behaviors in breast cells are observed.
Collapse
Affiliation(s)
- Saeid Ansaryan
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Mohammad Ali Khayamian
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,School of Mechanical Engineering, College of Engineering, University of Tehran, 11155-4563, Tehran, Iran
| | - Mohammad Saghafi
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Shahriar Shalileh
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Mohammad Saied Nikshoar
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Fereshteh Abbasvandi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX 15179/64311, Tehran, Iran
| | - Morteza Mahmoudi
- Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, 13169-43551, Tehran, Iran
| | - Farideh Bahrami
- Neuroscience Research Center and Dept. of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O.Box: 19839-63113, Tehran, Iran
| | - Mohammad Abdolahad
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| |
Collapse
|
29
|
Chai F, Li Y, Liu K, Li Q, Sun H. Caveolin enhances hepatocellular carcinoma cell metabolism, migration, and invasion in vitro via a hexokinase 2‐dependent mechanism. J Cell Physiol 2018; 234:1937-1946. [PMID: 30144070 DOI: 10.1002/jcp.27074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Fang Chai
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| | - Yan Li
- Department of General Surgery The Fourth Affiliated Hospital of China Medical University Shenyang China
| | - Keyi Liu
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| | - Qiang Li
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| | - Hongzhi Sun
- Department of General Surgery Affiliated Hospital of Jinzhou Medical University Jinzhou China
| |
Collapse
|
30
|
Cuadrado A, Manda G, Hassan A, Alcaraz MJ, Barbas C, Daiber A, Ghezzi P, León R, López MG, Oliva B, Pajares M, Rojo AI, Robledinos-Antón N, Valverde AM, Guney E, Schmidt HHHW. Transcription Factor NRF2 as a Therapeutic Target for Chronic Diseases: A Systems Medicine Approach. Pharmacol Rev 2018; 70:348-383. [PMID: 29507103 DOI: 10.1124/pr.117.014753] [Citation(s) in RCA: 457] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Systems medicine has a mechanism-based rather than a symptom- or organ-based approach to disease and identifies therapeutic targets in a nonhypothesis-driven manner. In this work, we apply this to transcription factor nuclear factor (erythroid-derived 2)-like 2 (NRF2) by cross-validating its position in a protein-protein interaction network (the NRF2 interactome) functionally linked to cytoprotection in low-grade stress, chronic inflammation, metabolic alterations, and reactive oxygen species formation. Multiscale network analysis of these molecular profiles suggests alterations of NRF2 expression and activity as a common mechanism in a subnetwork of diseases (the NRF2 diseasome). This network joins apparently heterogeneous phenotypes such as autoimmune, respiratory, digestive, cardiovascular, metabolic, and neurodegenerative diseases, along with cancer. Importantly, this approach matches and confirms in silico several applications for NRF2-modulating drugs validated in vivo at different phases of clinical development. Pharmacologically, their profile is as diverse as electrophilic dimethyl fumarate, synthetic triterpenoids like bardoxolone methyl and sulforaphane, protein-protein or DNA-protein interaction inhibitors, and even registered drugs such as metformin and statins, which activate NRF2 and may be repurposed for indications within the NRF2 cluster of disease phenotypes. Thus, NRF2 represents one of the first targets fully embraced by classic and systems medicine approaches to facilitate both drug development and drug repurposing by focusing on a set of disease phenotypes that appear to be mechanistically linked. The resulting NRF2 drugome may therefore rapidly advance several surprising clinical options for this subset of chronic diseases.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Gina Manda
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Ahmed Hassan
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - María José Alcaraz
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Coral Barbas
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Andreas Daiber
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Pietro Ghezzi
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Rafael León
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Manuela G López
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Baldo Oliva
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Marta Pajares
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Ana I Rojo
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Natalia Robledinos-Antón
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Angela M Valverde
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Emre Guney
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| | - Harald H H W Schmidt
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM (Autonomous University of Madrid)-CSIC (Centro Superior de Investigaciones Biomédicas), Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., M.P., A.I.R., N.R.-A.); Victor Babes National Institute of Pathology, Bucharest, Romania (A.C., G.M.); Department Pharmacology and Personalized Medicine, School for Cardiovascular Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastrich, The Netherlands (A.H., H.H.H.W.S.); Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain (M.J.A.); Centre for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad CEU (Centro de Estudios Universitarios)-San Pablo, Madrid, Spain (C.B.); Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Brighton and Sussex Medical School, Brighton, United Kingdom (P.G.); Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (R.L., M.G.L.); Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, Madrid, Spain (R.L., M.G.L.); GRIB (Unidad de Investigación en Informática Biomédica), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (B.O., E.G.); Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC and Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain (A.M.V.); and Structural Bioinformatics Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain (E.G.)
| |
Collapse
|
31
|
Abstract
Resistance of solid tumors to chemo- and radiotherapy remains a major obstacle in anti-cancer treatment. Herein, the membrane protein caveolin-1 (CAV1) came into focus as it is highly expressed in many tumors and high CAV1 levels were correlated with tumor progression, invasion and metastasis, and thus a worse clinical outcome. Increasing evidence further indicates that the heterogeneous tumor microenvironment, also known as the tumor stroma, contributes to therapy resistance resulting in poor clinical outcome. Again, CAV1 seems to play an important role in modulating tumor host interactions by promoting tumor growth, metastasis, therapy resistance and cell survival. However, the mechanisms driving stroma-mediated tumor growth and radiation resistance remain to be clarified. Understanding these interactions and thus, targeting CAV1 may offer a novel strategy for preventing cancer therapy resistance and improving clinical outcomes. In this review, we will summarize the resistance-promoting effects of CAV1 in tumors, and emphasize its role in the tumor-stroma communication as well as the resulting malignant phenotype of epithelial tumors.
Collapse
Affiliation(s)
- Julia Ketteler
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
32
|
Nrf2-Mediated Metabolic Reprogramming in Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9304091. [PMID: 29670683 PMCID: PMC5833252 DOI: 10.1155/2018/9304091] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/16/2017] [Accepted: 12/31/2017] [Indexed: 02/07/2023]
Abstract
Metabolic reprogramming is one of the hallmarks of cancer. Nrf2 pathway is one of the critical signaling cascades involved in cell defense and survival against oxidative stress. The significance of Nrf2 in cancer metabolism begins to be recognized. In this minireview, we focus on the Nrf2-mediated cancer metabolic reprogramming and intend to highlight the role of Nrf2 in the regulation of malignant transformation, cancer proliferation, and the development of treatment resistance via metabolic adaptations. We hope for the development of noninvasive biomarkers and novel therapeutic approaches for cancer based on Nrf2-directed cancer metabolic reprogramming in the near future.
Collapse
|
33
|
Volonte D, Liu Z, Shiva S, Galbiati F. Caveolin-1 controls mitochondrial function through regulation of m-AAA mitochondrial protease. Aging (Albany NY) 2017; 8:2355-2369. [PMID: 27705926 PMCID: PMC5115893 DOI: 10.18632/aging.101051] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/19/2016] [Indexed: 11/25/2022]
Abstract
Mitochondrial proteases ensure mitochondrial integrity and function after oxidative stress by providing mitochondrial protein quality control. However, the molecular mechanisms that regulate this basic biological function in eukaryotic cells remain largely unknown. Caveolin-1 is a scaffolding protein involved in signal transduction. We find that AFG3L2, a m-AAA type of mitochondrial protease, is a novel caveolin-1-interacting protein in vitro. We show that oxidative stress promotes the translocation of both caveolin-1 and AFG3L2 to mitochondria, enhances the interaction of caveolin-1 with AFG3L2 in mitochondria and stimulates mitochondrial protease activity in wild-type fibroblasts. Localization of AFG3L2 to mitochondria after oxidative stress is inhibited in fibroblasts lacking caveolin-1, which results in impaired mitochondrial protein quality control, an oxidative phosphorylation to aerobic glycolysis switch and reduced ATP production. Mechanistically, we demonstrate that a lack of caveolin-1 does not alter either mitochondrial number or morphology but leads to the cytoplasmic and proteasome-dependent degradation of complexes I, III, IV and V upon oxidant stimulation. Restoration of mitochondrial respiratory chain complexes in caveolin-1 null fibroblasts reverts the enhanced glycolysis observed in these cells. Expression of a mutant form of AFG3L2, which has reduced affinity for caveolin-1, fails to localize to mitochondria and promotes degradation of complex IV after oxidative stress. Thus, caveolin-1 maintains mitochondrial integrity and function when cells are challenged with free radicals by promoting the mitochondrial localization of m-AAA protease and its quality control functions.
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Zhongmin Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Vascular Medicine Institute and Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ferruccio Galbiati
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
34
|
Insights into the Dichotomous Regulation of SOD2 in Cancer. Antioxidants (Basel) 2017; 6:antiox6040086. [PMID: 29099803 PMCID: PMC5745496 DOI: 10.3390/antiox6040086] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
While loss of antioxidant expression and the resultant oxidant-dependent damage to cellular macromolecules is key to tumorigenesis, it has become evident that effective oxidant scavenging is conversely necessary for successful metastatic spread. This dichotomous role of antioxidant enzymes in cancer highlights their context-dependent regulation during different stages of tumor development. A prominent example of an antioxidant enzyme with such a dichotomous role and regulation is the mitochondria-localized manganese superoxide dismutase SOD2 (MnSOD). SOD2 has both tumor suppressive and promoting functions, which are primarily related to its role as a mitochondrial superoxide scavenger and H₂O₂ regulator. However, unlike true tumor suppressor- or onco-genes, the SOD2 gene is not frequently lost, or rarely mutated or amplified in cancer. This allows SOD2 to be either repressed or activated contingent on context-dependent stimuli, leading to its dichotomous function in cancer. Here, we describe some of the mechanisms that underlie SOD2 regulation in tumor cells. While much is known about the transcriptional regulation of the SOD2 gene, including downregulation by epigenetics and activation by stress response transcription factors, further research is required to understand the post-translational modifications that regulate SOD2 activity in cancer cells. Moreover, future work examining the spatio-temporal nature of SOD2 regulation in the context of changing tumor microenvironments is necessary to allows us to better design oxidant- or antioxidant-based therapeutic strategies that target the adaptable antioxidant repertoire of tumor cells.
Collapse
|
35
|
Chatterjee M, Ben-Josef E, Robb R, Vedaie M, Seum S, Thirumoorthy K, Palanichamy K, Harbrecht M, Chakravarti A, Williams TM. Caveolae-Mediated Endocytosis Is Critical for Albumin Cellular Uptake and Response to Albumin-Bound Chemotherapy. Cancer Res 2017; 77:5925-5937. [PMID: 28923854 DOI: 10.1158/0008-5472.can-17-0604] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/03/2017] [Accepted: 09/06/2017] [Indexed: 01/04/2023]
Abstract
Nab-paclitaxel, a nanoparticle conjugate of paclitaxel to human albumin, exhibits efficacy in pancreatic cancer, non-small cell lung cancer and breast cancer. However, there is a lack of predictive biomarkers to identify patients who might benefit most from its administration. This study addresses this gap in knowledge by identifying that caveolin-1 (Cav-1) is a candidate mechanism-based biomarker. Caveolae are small membrane invaginations important for transendothelial albumin uptake. Cav-1, the principal structural component of caveolae, is overexpressed in the cancers noted above that respond to nab-paclitaxel. Thus, we hypothesized that Cav-1 may be critical for albumin uptake in tumors and perhaps determine their response to this drug. Cav-1 protein levels correlated positively with nab-paclitaxel sensitivity. RNAi-mediated attenuation of Cav-1 expression reduced uptake of albumin and nab-paclitaxel in cancer cells and rendered them resistant to nab-paclitaxel-induced apoptosis. Conversely, Cav-1 overexpression enhanced sensitivity to nab-paclitaxel. Selection for cellular resistance to nab-paclitaxel in cell culture correlated with a loss of Cav-1 expression. In mouse xenograft models, cancer cells, where Cav-1 was attenuated, exhibited resistance to the antitumor effects of nab-paclitaxel therapy. Overall, our findings suggest Cav-1 as a predictive biomarker for the response to nab-paclitaxel and other albumin-based cancer therapeutic drugs. Cancer Res; 77(21); 5925-37. ©2017 AACR.
Collapse
Affiliation(s)
- Moumita Chatterjee
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Edgar Ben-Josef
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ryan Robb
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Marall Vedaie
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Star Seum
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Krishnan Thirumoorthy
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Kamalakannan Palanichamy
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Matthew Harbrecht
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Arnab Chakravarti
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Terence M Williams
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
| |
Collapse
|
36
|
Blajszczak C, Bonini MG. Mitochondria targeting by environmental stressors: Implications for redox cellular signaling. Toxicology 2017; 391:84-89. [PMID: 28750850 DOI: 10.1016/j.tox.2017.07.013] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Mitochondria are cellular powerhouses as well as metabolic and signaling hubs regulating diverse cellular functions, from basic physiology to phenotypic fate determination. It is widely accepted that reactive oxygen species (ROS) generated in mitochondria participate in the regulation of cellular signaling, and that some mitochondria chronically operate at a high ROS baseline. However, it is not completely understood how mitochondria adapt to persistently high ROS states and to environmental stressors that disturb the redox balance. Here we will review some of the current concepts regarding how mitochondria resist oxidative damage, how they are replaced when excessive oxidative damage compromises function, and the effect of environmental toxicants (i.e. heavy metals) on the regulation of mitochondrial ROS (mtROS) production and subsequent impact.
Collapse
Affiliation(s)
- Chuck Blajszczak
- Departments of Medicine and Pathology, University of Illinois College of Medicine at Chicago, IL, USA
| | - Marcelo G Bonini
- Departments of Medicine and Pathology, University of Illinois College of Medicine at Chicago, IL, USA.
| |
Collapse
|
37
|
Wu J, Zhou SL, Pi LH, Shi XJ, Ma LR, Chen Z, Qu ML, Li X, Nie SD, Liao DF, Pei JJ, Wang S. High glucose induces formation of tau hyperphosphorylation via Cav-1-mTOR pathway: A potential molecular mechanism for diabetes-induced cognitive dysfunction. Oncotarget 2017; 8:40843-40856. [PMID: 28489581 PMCID: PMC5522306 DOI: 10.18632/oncotarget.17257] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/07/2017] [Indexed: 12/31/2022] Open
Abstract
The abnormally hyperphosphorylated tau is thought to be implicated in diabetes-associated cognitive deficits. The role of mammalian target of rapamycin (mTOR) / S6 kinase (S6K) signalling in the formation of tau hyperphosphorylation has been previously studied. Caveolin-1 (Cav-1), the essential structure protein of caveolae, promotes neuronal survival and growth, and inhibits glucose metabolism. In this study, we aimed to investigate the role of Cav-1 in the formation of tau hyperphosphorylation under chronic hyperglycemic condition (HGC). Diabetic rats were induced by streptozotocin (STZ). Primary hippocampal neurons with or without molecular intervention such as the transient over-expression or knock-down were subjected to HGC. The obtained experimental samples were analyzed by real time quantitative RT-PCR, Western blot, immunofluorescence or immunohistochemisty. We found: 1) that a chronic HGC directly decreases Cav-1 expression, increases tau phosphorylation and activates mTOR/S6K signalling in the brain neurons of diabetic rats, 2) that overexpression of Cav-1 attenuates tau hyperphosphorylation induced by chronic HGC in primary hippocampal neurons, whereas down-regulation of Cav-1 using Cav-1 siRNA dramatically worsens tau hyperphosphorylation via mTOR/S6K signalling pathway, and 3) that the down-regulation of Cav-1 induced by HGC is independent of mTOR signalling. Our results suggest that tau hyperphosphorylation and the sustained over-activated mTOR signalling under hyperglycemia may be due to the suppression of Cav-1. Therefore, Cav-1 is a potential therapeutic target for diabetes-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Jing Wu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Shan-Lei Zhou
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Lin-Hua Pi
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Xia-Jie Shi
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Ling-Ran Ma
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, China
| | - Zi Chen
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Min-Li Qu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Li
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, China
| | - Sheng-Dan Nie
- Institute of Clinical Medicine, People's Hospital of Hunan Province, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Jin-Jing Pei
- KI-Alzheimer's Disease Research Center, Karolinska Institutet, Novum, Stockholm, Sweden
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Xicheng, Beijing, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Shan Wang
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Caveolin-1: An Oxidative Stress-Related Target for Cancer Prevention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7454031. [PMID: 28546853 PMCID: PMC5436035 DOI: 10.1155/2017/7454031] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/23/2017] [Accepted: 03/07/2017] [Indexed: 01/19/2023]
Abstract
Aberrant oxidative metabolism is one of the hallmarks of cancer. Reactive species overproduction could promote carcinogenesis via inducing genetic mutations and activating oncogenic pathways, and thus, antioxidant therapy was considered as an important strategy for cancer prevention and treatment. Caveolin-1 (Cav-1), a constituent protein of caveolae, has been shown to mediate tumorigenesis and progression through oxidative stress modulation recently. Reactive species could modulate the expression, degradation, posttranslational modifications, and membrane trafficking of Cav-1, while Cav-1-targeted treatments could scavenge the reactive species. More importantly, emerging evidences have indicated that multiple antioxidants could exert antitumor activities in cancer cells and protective activities in normal cells by modulating the Cav-1 pathway. Altogether, these findings indicate that Cav-1 may be a promising oxidative stress-related target for cancer antioxidant prevention. Elucidating the underlying interaction mechanisms between oxidative stress and Cav-1 is helpful for enhancing the preventive effects of antioxidants on cancer, for improving clinical outcomes of antioxidant-related therapeutics in cancer patients, and for developing Cav-1 targeted drugs. Herein, we summarize the available evidence of the roles of Cav-1 and oxidative stress in tumorigenesis and development and shed novel light on designing strategies for cancer prevention or treatment by utilizing the interaction mode between Cav-1 and oxidative stress.
Collapse
|
39
|
Fu P, Chen F, Pan Q, Zhao X, Zhao C, Cho WCS, Chen H. The different functions and clinical significances of caveolin-1 in human adenocarcinoma and squamous cell carcinoma. Onco Targets Ther 2017; 10:819-835. [PMID: 28243118 PMCID: PMC5317307 DOI: 10.2147/ott.s123912] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Caveolin-1 (Cav-1), a major structural protein of caveolae, is an integral membrane protein which plays an important role in the progression of carcinoma. However, whether Cav-1 acts as a tumor promoter or a tumor suppressor still remains controversial. For example, the tumor-promoting function of Cav-1 has been found in renal cancer, prostate cancer, tongue squamous cell carcinoma (SCC), lung SCC and bladder SCC. In contrast, Cav-1 also plays an inhibitory role in esophagus adenocarcinoma, lung adenocarcinoma and cutaneous SCC. The role of Cav-1 is still controversial in thyroid cancer, hepatocellular carcinoma, gastric adenocarcinoma, colon adenocarcinoma, breast cancer, pancreas cancer, oral SCC, laryngeal SCC, head and neck SCC, esophageal SCC and cervical SCC. Besides, it has been reported that the loss of stromal Cav-1 might predict poor prognosis in breast cancer, gastric cancer, pancreas cancer, prostate cancer, oral SCC and esophageal SCC. However, the accumulation of stromal Cav-1 has been found to be promoted by the progression of tongue SCC. Taken together, Cav-1 seems playing a different role in different cancer subtypes even of the same organ, as well as acting differently in the same cancer subtype of different organs. Thus, we hereby explore the functions of Cav-1 in human adenocarcinoma and SCC from the perspective of clinical significances and pathogenesis. We envision that novel targets may come with the further investigation of Cav-1 in carcinogenesis.
Collapse
Affiliation(s)
- Pin Fu
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | - Fuchun Chen
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang
| | - Qi Pan
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang
| | - Xianda Zhao
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | - Chen Zhao
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | | | - Honglei Chen
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan; Department of Pathology, Maternal and Child Health Hospital of Hubei, Wuhan, People's Republic of China
| |
Collapse
|
40
|
Endo A, Ly T, Pippa R, Bensaddek D, Nicolas A, Lamond AI. The Chromatin Assembly Factor Complex 1 (CAF1) and 5-Azacytidine (5-AzaC) Affect Cell Motility in Src-transformed Human Epithelial Cells. J Biol Chem 2016; 292:172-184. [PMID: 27872192 PMCID: PMC5217677 DOI: 10.1074/jbc.m116.751024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/16/2016] [Indexed: 12/29/2022] Open
Abstract
Tumor invasion into surrounding stromal tissue is a hallmark of high grade, metastatic cancers. Oncogenic transformation of human epithelial cells in culture can be triggered by activation of v-Src kinase, resulting in increased cell motility, invasiveness, and tumorigenicity and provides a valuable model for studying how changes in gene expression cause cancer phenotypes. Here, we show that epithelial cells transformed by activated Src show increased levels of DNA methylation and that the methylation inhibitor 5-azacytidine (5-AzaC) potently blocks the increased cell motility and invasiveness induced by Src activation. A proteomic screen for chromatin regulators acting downstream of activated Src identified the replication-dependent histone chaperone CAF1 as an important factor for Src-mediated increased cell motility and invasion. We show that Src causes a 5-AzaC-sensitive decrease in both mRNA and protein levels of the p150 (CHAF1A) and p60 (CHAF1B), subunits of CAF1. Depletion of CAF1 in untransformed epithelial cells using siRNA was sufficient to recapitulate the increased motility and invasive phenotypes characteristic of transformed cells without activation of Src. Maintaining high levels of CAF1 by exogenous expression suppressed the increased cell motility and invasiveness phenotypes when Src was activated. These data identify a critical role of CAF1 in the dysregulation of cell invasion and motility phenotypes seen in transformed cells and also highlight an important role for epigenetic remodeling through DNA methylation for Src-mediated induction of cancer phenotypes.
Collapse
Affiliation(s)
- Akinori Endo
- From the Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Tony Ly
- From the Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Raffaella Pippa
- From the Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Dalila Bensaddek
- From the Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Armel Nicolas
- From the Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Angus I Lamond
- From the Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| |
Collapse
|
41
|
Nwosu ZC, Ebert MP, Dooley S, Meyer C. Caveolin-1 in the regulation of cell metabolism: a cancer perspective. Mol Cancer 2016; 15:71. [PMID: 27852311 PMCID: PMC5112640 DOI: 10.1186/s12943-016-0558-7] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/03/2016] [Indexed: 12/16/2022] Open
Abstract
Caveolin-1 (CAV1) is an oncogenic membrane protein associated with endocytosis, extracellular matrix organisation, cholesterol distribution, cell migration and signaling. Recent studies reveal that CAV1 is involved in metabolic alterations – a critical strategy adopted by cancer cells to their survival advantage. Consequently, research findings suggest that CAV1, which is altered in several cancer types, influences tumour development or progression by controlling metabolism. Understanding the molecular interplay between CAV1 and metabolism could help uncover druggable metabolic targets or pathways of clinical relevance in cancer therapy. Here we review from a cancer perspective, the findings that CAV1 modulates cell metabolism with a focus on glycolysis, mitochondrial bioenergetics, glutaminolysis, fatty acid metabolism, and autophagy.
Collapse
Affiliation(s)
- Zeribe Chike Nwosu
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Matthias Philip Ebert
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Christoph Meyer
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany. .,Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany.
| |
Collapse
|
42
|
He C, Hart PC, Germain D, Bonini MG. SOD2 and the Mitochondrial UPR: Partners Regulating Cellular Phenotypic Transitions. Trends Biochem Sci 2016; 41:568-577. [PMID: 27180143 DOI: 10.1016/j.tibs.2016.04.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/05/2016] [Accepted: 04/15/2016] [Indexed: 12/13/2022]
Abstract
ATP and reactive oxygen species (ROS) are signaling molecules that control cellular function and phenotype. Mitochondria produce both ATP and ROS. Since the electrons needed to generate either ATP or ROS originate from NADH/FADH2, the mechanism through which electrons flow towards oxygen determines yields and whether ATP or ROS prevails. Alterations in the electron flow impact cells dramatically, such as by supporting specialization (which requires high ATP) or imposing dedifferentiation. High ROS, facilitated by enzymes such as superoxide dismutase 2 (SOD2) that enhance mitochondrial hydrogen peroxide (mtH2O2), are normally linked to dedifferentiation of somatic cells. Here we propose that combined high mtH2O2 and mitochondrial unfolded protein response (UPR(mt)) activation are essential for somatic dedifferentiation programs and the acquisition of stem-like properties in reparative processes and disease.
Collapse
Affiliation(s)
- Chenxia He
- Department of Medicine, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pathology, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Peter C Hart
- Department of Medicine, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pathology, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Doris Germain
- Department of Medicine (Hematology and Medical Oncology), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marcelo G Bonini
- Department of Medicine, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pathology, College of Medicine of the University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|