1
|
Khan F, Elsori D, Verma M, Pandey S, Obaidur Rab S, Siddiqui S, Alabdallah NM, Saeed M, Pandey P. Unraveling the intricate relationship between lipid metabolism and oncogenic signaling pathways. Front Cell Dev Biol 2024; 12:1399065. [PMID: 38933330 PMCID: PMC11199418 DOI: 10.3389/fcell.2024.1399065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Lipids, the primary constituents of the cell membrane, play essential roles in nearly all cellular functions, such as cell-cell recognition, signaling transduction, and energy provision. Lipid metabolism is necessary for the maintenance of life since it regulates the balance between the processes of synthesis and breakdown. Increasing evidence suggests that cancer cells exhibit abnormal lipid metabolism, significantly affecting their malignant characteristics, including self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. Prominent oncogenic signaling pathways that modulate metabolic gene expression and elevate metabolic enzyme activity include phosphoinositide 3-kinase (PI3K)/AKT, MAPK, NF-kB, Wnt, Notch, and Hippo pathway. Conversely, when metabolic processes are not regulated, they can lead to malfunctions in cellular signal transduction pathways. This, in turn, enables uncontrolled cancer cell growth by providing the necessary energy, building blocks, and redox potentials. Therefore, targeting lipid metabolism-associated oncogenic signaling pathways could be an effective therapeutic approach to decrease cancer incidence and promote survival. This review sheds light on the interactions between lipid reprogramming and signaling pathways in cancer. Exploring lipid metabolism as a target could provide a promising approach for creating anticancer treatments by identifying metabolic inhibitors. Additionally, we have also provided an overview of the drugs targeting lipid metabolism in cancer in this review.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Deena Elsori
- Faculty of Resilience, Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Samra Siddiqui
- Department of Health Service Management, College of Public Health and Health Informatics, University of Hail, Haʼil, Saudi Arabia
| | - Nadiyah M. Alabdallah
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Haʼil, Saudi Arabia
| | - Pratibha Pandey
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
2
|
Sun J, Yu L, Qu X, Huang T. The role of peroxisome proliferator-activated receptors in the tumor microenvironment, tumor cell metabolism, and anticancer therapy. Front Pharmacol 2023; 14:1184794. [PMID: 37251321 PMCID: PMC10213337 DOI: 10.3389/fphar.2023.1184794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been extensively studied for over 3 decades and consist of three isotypes, including PPARα, γ, and β/δ, that were originally considered key metabolic regulators controlling energy homeostasis in the body. Cancer has become a leading cause of human mortality worldwide, and the role of peroxisome proliferator-activated receptors in cancer is increasingly being investigated, especially the deep molecular mechanisms and effective cancer therapies. Peroxisome proliferator-activated receptors are an important class of lipid sensors and are involved in the regulation of multiple metabolic pathways and cell fate. They can regulate cancer progression in different tissues by activating endogenous or synthetic compounds. This review emphasizes the significance and knowledge of peroxisome proliferator-activated receptors in the tumor microenvironment, tumor cell metabolism, and anti-cancer treatment by summarizing recent research on peroxisome proliferator-activated receptors. In general, peroxisome proliferator-activated receptors either promote or suppress cancer in different types of tumor microenvironments. The emergence of this difference depends on various factors, including peroxisome proliferator-activated receptor type, cancer type, and tumor stage. Simultaneously, the effect of anti-cancer therapy based on drug-targeted PPARs differs or even opposes among the three peroxisome proliferator-activated receptor homotypes and different cancer types. Therefore, the current status and challenges of the use of peroxisome proliferator-activated receptors agonists and antagonists in cancer treatment are further explored in this review.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Liyan Yu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Xueling Qu
- Dalian Women and Children’s Medical Center(Group), Dalian, Liaoning, China
| | - Tao Huang
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
3
|
Fan J, To KKW, Chen ZS, Fu L. ABC transporters affects tumor immune microenvironment to regulate cancer immunotherapy and multidrug resistance. Drug Resist Updat 2023; 66:100905. [PMID: 36463807 DOI: 10.1016/j.drup.2022.100905] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022]
Abstract
Multidrug resistance (MDR) is the phenomenon in which cancer cells simultaneously develop resistance to a broad spectrum of structurally and mechanistically unrelated drugs. MDR severely hinders the effective treatment of cancer and is the major cause of chemotherapy failure. ATP-binding cassette (ABC) transporters are extensively expressed in various body tissues, and actively transport endogenous and exogenous substrates through biological membranes. Overexpression of ABC transporters is frequently observed in MDR cancer cells, which promotes efflux of chemotherapeutic drugs and reduces their intracellular accumulation. Increasing evidence suggests that ABC transporters regulate tumor immune microenvironment (TIME) by transporting various cytokines, thus controlling anti-tumor immunity and sensitivity to anticancer drugs. On the other hand, the expression of various ABC transporters is regulated by cytokines and other immune signaling molecules. Targeted inhibition of ABC transporter expression or function can enhance the efficacy of immune checkpoint inhibitors by promoting anticancer immune microenvironment. This review provides an update on the recent research progress in this field.
Collapse
Affiliation(s)
- Jingyi Fan
- State Key Laboratory of Oncology in South China;Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing 100038, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States.
| | - Liwu Fu
- State Key Laboratory of Oncology in South China;Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
4
|
Wagner N, Wagner KD. Peroxisome Proliferator-Activated Receptors and the Hallmarks of Cancer. Cells 2022; 11:cells11152432. [PMID: 35954274 PMCID: PMC9368267 DOI: 10.3390/cells11152432] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) function as nuclear transcription factors upon the binding of physiological or pharmacological ligands and heterodimerization with retinoic X receptors. Physiological ligands include fatty acids and fatty-acid-derived compounds with low specificity for the different PPAR subtypes (alpha, beta/delta, and gamma). For each of the PPAR subtypes, specific pharmacological agonists and antagonists, as well as pan-agonists, are available. In agreement with their natural ligands, PPARs are mainly focused on as targets for the treatment of metabolic syndrome and its associated complications. Nevertheless, many publications are available that implicate PPARs in malignancies. In several instances, they are controversial for very similar models. Thus, to better predict the potential use of PPAR modulators for personalized medicine in therapies against malignancies, it seems necessary and timely to review the three PPARs in relation to the didactic concept of cancer hallmark capabilities. We previously described the functions of PPAR beta/delta with respect to the cancer hallmarks and reviewed the implications of all PPARs in angiogenesis. Thus, the current review updates our knowledge on PPAR beta and the hallmarks of cancer and extends the concept to PPAR alpha and PPAR gamma.
Collapse
Affiliation(s)
- Nicole Wagner
- Correspondence: (N.W.); (K.-D.W.); Tel.: +33-489-153-713 (K.-D.W.)
| | | |
Collapse
|
5
|
Dadhich R, Kapoor S. Lipidomic and Membrane Mechanical Signatures in Triple-Negative Breast Cancer: Scope for Membrane-Based Theranostics. Mol Cell Biochem 2022; 477:2507-2528. [PMID: 35595957 DOI: 10.1007/s11010-022-04459-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer associated with poor prognosis, higher grade, and a high rate of metastatic occurrence. Limited therapeutic interventions and the compounding issue of drug resistance in triple-negative breast cancer warrants the discovery of novel therapeutic targets and diagnostic modules. To this view, in addition to proteins, lipids also regulate cellular functions via the formation of membranes that modulate membrane protein function, diffusion, and their localization; thus, orchestrating signaling hot spots enriched in specific lipids/proteins on cell membranes. Lipid deregulation in cancer leads to reprogramming of the membrane dynamics and functions impacting cell proliferation, metabolism, and metastasis, providing exciting starting points for developing lipid-based approaches for treating TNBC. In this review, we provide a detailed account of specific lipidic changes in breast cancer, link the altered lipidome with membrane structure and mechanical properties, and describe how these are linked to subsequent downstream functions implicit in cancer progression, metastasis, and chemoresistance. At the fundamental level, we discuss how the lipid-centric findings in TNBC are providing cues for developing lipid-inspired theranostic strategies while bridging existing gaps in our understanding of the functional involvement of lipid membranes in cancer.
Collapse
Affiliation(s)
- Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India. .,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan.
| |
Collapse
|
6
|
Han W, Wang N, Kong R, Bao W, Lu J. Ligand-activated PPARδ expression promotes hepatocellular carcinoma progression by regulating the PI3K-AKT signaling pathway. J Transl Med 2022; 20:86. [PMID: 35151320 PMCID: PMC8840031 DOI: 10.1186/s12967-022-03288-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor-beta/delta (PPARδ) was considered as the key regulator involved in the evolution of various tumors. Given that PPARδ potential role in hepatocellular carcinoma (HCC) is still obscure, we comprehensively assessed its expression pattern, prognosis, functions and correlation with tumor microenvironment in HCC using public database data and in vitro studies. Methods Transcriptional data and clinical data in the TCGA and GEO database were analyzed in R software. Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemistry were used to detect the expression level of related RNA and proteins. The malignant biological characteristics were explored by cell counting Kit-8 (CCK8), 5-Ethynyl-2ʹ-deoxyuridine (EdU) assay and wound healing assay. Results Our results illustrated that PPARδ expression was significantly higher in HCC tissues and HCC cell lines. Elevated expression of PPARδ suggested poor clinical staging and prognosis in HCC. Ligand-activated PPARδ expression promoted the proliferation and invasion of HCC cells via PDK1/AKT/GSK3β signaling pathway. The expression of PPARδ was closely related to the HCC tumor microenvironment. Conclusions PPARδ plays an important part in HCC progression, penetrating investigation of the related regulatory mechanism may shed light upon further biological and pharmacological value.
Collapse
|
7
|
Palano MT, Gallazzi M, Cucchiara M, Dehò F, Capogrosso P, Bruno A, Mortara L. The tumor innate immune microenvironment in prostate cancer: an overview of soluble factors and cellular effectors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:694-718. [PMID: 36338516 PMCID: PMC9630328 DOI: 10.37349/etat.2022.00108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 01/14/2023] Open
Abstract
Prostate cancer (PCa) accounts as the most common non-cutaneous disease affecting males, and as the first cancer, for incidence, in male. With the introduction of the concept of immunoscore, PCa has been classified as a cold tumor, thus driving the attention in the development of strategies aimed at blocking the infiltration/activation of immunosuppressive cells, while favoring the infiltration/activation of anti-tumor immune cells. Even if immunotherapy has revolutionized the approaches to cancer therapy, there is still a window failure, due to the immune cell plasticity within PCa, that can acquire pro-tumor features, subsequent to the tumor microenvironment (TME) capability to polarize them. This review discussed selected relevant soluble factors [transforming growth factor-beta (TGFβ), interleukin-6 (IL-6), IL-10, IL-23] and cellular components of the innate immunity, as drivers of tumor progression, immunosuppression, and angiogenesis within the PCa-TME.
Collapse
Affiliation(s)
- Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy
| | - Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Martina Cucchiara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Federico Dehò
- Unit of Urology, ASST-Sette Laghi, Ospedale di Circolo e Fondazione Macchi, University of Insubria, 21100 Varese, Italy
| | - Paolo Capogrosso
- Unit of Urology, ASST-Sette Laghi, Ospedale di Circolo e Fondazione Macchi, University of Insubria, 21100 Varese, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy,Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy,Correspondence: Antonino Bruno,
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy,Lorenzo Mortara, . Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
8
|
Jeong DW, Lee S, Chun YS. How cancer cells remodel lipid metabolism: strategies targeting transcription factors. Lipids Health Dis 2021; 20:163. [PMID: 34775964 PMCID: PMC8590761 DOI: 10.1186/s12944-021-01593-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Reprogramming of lipid metabolism has received increasing recognition as a hallmark of cancer cells because lipid dysregulation and the alteration of related enzyme profiles are closely correlated with oncogenic signals and malignant phenotypes, such as metastasis and therapeutic resistance. In this review, we describe recent findings that support the importance of lipids, as well as the transcription factors involved in cancer lipid metabolism. With recent advances in transcription factor analysis, including computer-modeling techniques, transcription factors are emerging as central players in cancer biology. Considering the limited number and the crucial role of transcription factors associated with lipid rewiring in cancers, transcription factor targeting is a promising potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Do-Won Jeong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Seulbee Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
9
|
Trivedi T, Pagnotti GM, Guise TA, Mohammad KS. The Role of TGF-β in Bone Metastases. Biomolecules 2021; 11:1643. [PMID: 34827641 PMCID: PMC8615596 DOI: 10.3390/biom11111643] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Complications associated with advanced cancer are a major clinical challenge and, if associated with bone metastases, worsen the prognosis and compromise the survival of the patients. Breast and prostate cancer cells exhibit a high propensity to metastasize to bone. The bone microenvironment is unique, providing fertile soil for cancer cell propagation, while mineralized bone matrices store potent growth factors and cytokines. Biologically active transforming growth factor β (TGF-β), one of the most abundant growth factors, is released following tumor-induced osteoclastic bone resorption. TGF-β promotes tumor cell secretion of factors that accelerate bone loss and fuel tumor cells to colonize. Thus, TGF-β is critical for driving the feed-forward vicious cycle of tumor growth in bone. Further, TGF-β promotes epithelial-mesenchymal transition (EMT), increasing cell invasiveness, angiogenesis, and metastatic progression. Emerging evidence shows TGF-β suppresses immune responses, enabling opportunistic cancer cells to escape immune checkpoints and promote bone metastases. Blocking TGF-β signaling pathways could disrupt the vicious cycle, revert EMT, and enhance immune response. However, TGF-β's dual role as both tumor suppressor and enhancer presents a significant challenge in developing therapeutics that target TGF-β signaling. This review presents TGF-β's role in cancer progression and bone metastases, while highlighting current perspectives on the therapeutic potential of targeting TGF-β pathways.
Collapse
Affiliation(s)
- Trupti Trivedi
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Gabriel M. Pagnotti
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Theresa A. Guise
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Khalid S. Mohammad
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
10
|
Cheng HS, Yip YS, Lim EKY, Wahli W, Tan NS. PPARs and Tumor Microenvironment: The Emerging Roles of the Metabolic Master Regulators in Tumor Stromal-Epithelial Crosstalk and Carcinogenesis. Cancers (Basel) 2021; 13:2153. [PMID: 33946986 PMCID: PMC8125182 DOI: 10.3390/cancers13092153] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been extensively studied for more than three decades. Consisting of three isotypes, PPARα, γ, and β/δ, these nuclear receptors are regarded as the master metabolic regulators which govern many aspects of the body energy homeostasis and cell fate. Their roles in malignancy are also increasingly recognized. With the growing interest in crosstalk between tumor stroma and epithelium, this review aims to highlight the current knowledge on the implications of PPARs in the tumor microenvironment. PPARγ plays a crucial role in the metabolic reprogramming of cancer-associated fibroblasts and adipocytes, coercing the two stromal cells to become substrate donors for cancer growth. Fibroblast PPARβ/δ can modify the risk of tumor initiation and cancer susceptibility. In endothelial cells, PPARβ/δ and PPARα are pro- and anti-angiogenic, respectively. Although the angiogenic role of PPARγ remains ambiguous, it is a crucial regulator in autocrine and paracrine signaling of cancer-associated fibroblasts and tumor-associated macrophages/immune cells. Of note, angiopoietin-like 4 (ANGPTL4), a secretory protein encoded by a target gene of PPARs, triggers critical oncogenic processes such as inflammatory signaling, extracellular matrix derangement, anoikis resistance and metastasis, making it a potential drug target for cancer treatment. To conclude, PPARs in the tumor microenvironment exhibit oncogenic activities which are highly controversial and dependent on many factors such as stromal cell types, cancer types, and oncogenesis stages. Thus, the success of PPAR-based anticancer treatment potentially relies on innovative strategies to modulate PPAR activity in a cell type-specific manner.
Collapse
Affiliation(s)
- Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
| | - Yun Sheng Yip
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
| | - Eldeen Kai Yi Lim
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR 1331, UPS, Université de Toulouse, 31300 Toulouse, France
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
11
|
O'Sullivan SE, Kaczocha M. FABP5 as a novel molecular target in prostate cancer. Drug Discov Today 2020; 25:S1359-6446(20)30375-5. [PMID: 32966866 PMCID: PMC8059105 DOI: 10.1016/j.drudis.2020.09.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/07/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Emerging evidence suggests that dysregulated lipid signaling is a key factor in prostate cancer (PC), through fatty acid activation of the nuclear receptors peroxisome proliferator-activated receptors (PPARs), leading to the upregulation of protumoral genes. Fatty acid-binding proteins (FABPs) are intracellular lipid-binding proteins that transport fatty acid to PPARs, facilitating their activation. FABP5 is overexpressed in PC, and correlates with poor patient prognosis and survival. Genetic knockdown or silencing of FABP5 decreases the proliferation and invasiveness of PC cells in vitro, and reduces tumor growth and metastasis in vivo. Pharmacological FABP5-specific inhibitors also reduce tumor growth and metastases, and produce synergistic effects with taxanes. In this review, we present current data supporting FABP5 as a novel molecular target for PC.
Collapse
Affiliation(s)
| | - Martin Kaczocha
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NYH, USA
| |
Collapse
|
12
|
Her NG, Kesari S, Nurmemmedov E. Thrombospondin-1 counteracts the p97 inhibitor CB-5083 in colon carcinoma cells. Cell Cycle 2020; 19:1590-1601. [PMID: 32423265 DOI: 10.1080/15384101.2020.1754584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
p97 has recently emerged as a therapeutic target for cancer due to its essential functions in protein homeostasis. CB-5083 is a first-in-class, potent and selective ATP-competitive p97 inhibitor that induces proteotoxic stress in cancer cells. Potential mechanisms regulating the sensitivity of cells to p97 inhibition remain poorly studied. Here, we demonstrate that Thrombospondin-1 (THBS1) is a CB-5083-upregulated gene that helps confer resistance of HCT116 cells to CB-5083. Our immunoblotting and immunofluorescence data showed that CB-5083 significantly increases the steady-state abundance of THBS1. Blockade of THBS1 induction sensitized cells to CB-5083-mediated growth inhibition. Suppression of THBS1 caused an increase of CB-5083-induced sub-G1 population and caspase 3/7 activity suggesting that its function is linked to the survival of cancer cells in response to p97 inhibition. Altogether our data provide new evidence that THBS1 is important for the susceptibility of cells to p97 inhibition.
Collapse
Affiliation(s)
- Nam-Gu Her
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences , Seoul, Republic of Korea.,Department of Neuro-sciences and Neuro-therapeutics, John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center , Santa Monica, CA, USA
| | - Santosh Kesari
- Institute for Refractory Cancer Research, Current Address: Samsung Medical Center , Seoul, Republic of Korea
| | - Elmar Nurmemmedov
- Institute for Refractory Cancer Research, Current Address: Samsung Medical Center , Seoul, Republic of Korea
| |
Collapse
|
13
|
Wagner N, Wagner KD. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020; 9:cells9051133. [PMID: 32375405 PMCID: PMC7291220 DOI: 10.3390/cells9051133] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family. Three different isoforms, PPAR alpha, PPAR beta/delta and PPAR gamma have been identified. They all form heterodimers with retinoic X receptors to activate or repress downstream target genes dependent on the presence/absence of ligands and coactivators or corepressors. PPARs differ in their tissue expression profile, ligands and specific agonists and antagonists. PPARs attract attention as potential therapeutic targets for a variety of diseases. PPAR alpha and gamma agonists are in clinical use for the treatment of dyslipidemias and diabetes. For both receptors, several clinical trials as potential therapeutic targets for cancer are ongoing. In contrast, PPAR beta/delta has been suggested as a therapeutic target for metabolic syndrome. However, potential risks in the settings of cancer are less clear. A variety of studies have investigated PPAR beta/delta expression or activation/inhibition in different cancer cell models in vitro, but the relevance for cancer growth in vivo is less well documented and controversial. In this review, we summarize critically the knowledge of PPAR beta/delta functions for the different hallmarks of cancer biological capabilities, which interplay to determine cancer growth.
Collapse
|
14
|
Nobili S, Lapucci A, Landini I, Coronnello M, Roviello G, Mini E. Role of ATP-binding cassette transporters in cancer initiation and progression. Semin Cancer Biol 2020; 60:72-95. [PMID: 31412294 DOI: 10.1016/j.semcancer.2019.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022]
Abstract
ATP Binding Cassette (ABC) transporters, widely studied in cancer for their role in drug resistance, have been more recently also considered for their contribution to cancer cell biology. To date, many data provide evidences for their potential role in all the phases of cancer development from cancer susceptibility, tumor initiation, tumor progression and metastasis. Although many evidences are based on correlative analyses, data describing a direct or indirect role of ABC transporters in cancer biology are increasing. Overall, current available information suggests a relevant molecular effector role of some ABC transporters in cancer invasion and metastasis as reported in experimental tumor models. From a therapeutic point of view, due to the physiological relevant roles that ABC transporters play in the organism, the capability to selectively inhibit the function or the expression of ABC transporters in cancer stem cells or other tumor cells, represents the main challenge for researcher scientists. A detailed and updated description of the current knowledge on the role of ABC transporters in cancer biology is provided.
Collapse
Affiliation(s)
- Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy.
| | - Andrea Lapucci
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | | | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
15
|
Carbonetti G, Wilpshaar T, Kroonen J, Studholme K, Converso C, d'Oelsnitz S, Kaczocha M. FABP5 coordinates lipid signaling that promotes prostate cancer metastasis. Sci Rep 2019; 9:18944. [PMID: 31831821 PMCID: PMC6908725 DOI: 10.1038/s41598-019-55418-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/15/2019] [Indexed: 01/11/2023] Open
Abstract
Prostate cancer (PCa) is defined by dysregulated lipid signaling and is characterized by upregulation of lipid metabolism-related genes including fatty acid binding protein 5 (FABP5), fatty acid synthase (FASN), and monoacylglycerol lipase (MAGL). FASN and MAGL are enzymes that generate cellular fatty acid pools while FABP5 is an intracellular chaperone that delivers fatty acids to nuclear receptors to enhance PCa metastasis. Since FABP5, FASN, and MAGL have been independently implicated in PCa progression, we hypothesized that FABP5 represents a central mechanism linking cytosolic lipid metabolism to pro-metastatic nuclear receptor signaling. Here, we show that the abilities of FASN and MAGL to promote nuclear receptor activation and PCa metastasis are critically dependent upon co-expression of FABP5 in vitro and in vivo. Our findings position FABP5 as a key driver of lipid-mediated metastasis and suggest that disruption of lipid signaling via FABP5 inhibition may constitute a new avenue to treat metastatic PCa.
Collapse
Affiliation(s)
- Gregory Carbonetti
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tessa Wilpshaar
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jessie Kroonen
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Keith Studholme
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Cynthia Converso
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Simon d'Oelsnitz
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Martin Kaczocha
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA. .,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA. .,Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
16
|
Farràs M, Arranz S, Carrión S, Subirana I, Muñoz-Aguayo D, Blanchart G, Kool M, Solà R, Motilva MJ, Escolà-Gil JC, Rubió L, Fernández-Castillejo S, Pedret A, Estruch R, Covas MI, Fitó M, Hernáez Á, Castañer O. A Functional Virgin Olive Oil Enriched with Olive Oil and Thyme Phenolic Compounds Improves the Expression of Cholesterol Efflux-Related Genes: A Randomized, Crossover, Controlled Trial. Nutrients 2019; 11:nu11081732. [PMID: 31357534 PMCID: PMC6723782 DOI: 10.3390/nu11081732] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022] Open
Abstract
The consumption of antioxidant-rich foods such as virgin olive oil (VOO) promotes high-density lipoprotein (HDL) anti-atherogenic capacities. Intake of functional VOOs (enriched with olive/thyme phenolic compounds (PCs)) also improves HDL functions, but the gene expression changes behind these benefits are not fully understood. Our aim was to determine whether these functional VOOs could enhance the expression of cholesterol efflux-related genes. In a randomized, double-blind, crossover, controlled trial, 22 hypercholesterolemic subjects ingested for three weeks 25 mL/day of: (1) a functional VOO enriched with olive oil PCs (500 mg/kg); (2) a functional VOO enriched with olive oil (250 mg/kg) and thyme PCs (250 mg/kg; FVOOT), and; (3) a natural VOO (olive oil PCs: 80 mg/kg, control intervention). We assessed whether these interventions improved the expression of cholesterol efflux-related genes in peripheral blood mononuclear cells by quantitative reverse-transcription polymerase chain reactions. The FVOOT intervention upregulated the expression of CYP27A1 (p = 0.041 and p = 0.053, versus baseline and the control intervention, respectively), CAV1 (p = 0.070, versus the control intervention), and LXRβ, RXRα, and PPARβ/δ (p = 0.005, p = 0.005, and p = 0.038, respectively, relative to the baseline). The consumption of a functional VOO enriched with olive oil and thyme PCs enhanced the expression of key cholesterol efflux regulators, such as CYP27A1 and nuclear receptor-related genes.
Collapse
Affiliation(s)
- Marta Farràs
- Molecular Bases of Cardiovascular Risk Group, IIB-Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Sara Arranz
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Sílvia Carrión
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Isaac Subirana
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Cardiovascular Genetics and Epidemiology Research Group, IMIM, 08003 Barcelona, Spain
| | - Daniel Muñoz-Aguayo
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
| | - Gemma Blanchart
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Marjon Kool
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Rosa Solà
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - María José Motilva
- Instituto de Ciencias de la Vid y el Vino, CSIC-Universidad de la Rioja, 26007 Logroño, Spain
| | - Joan Carles Escolà-Gil
- Molecular Bases of Cardiovascular Risk Group, IIB-Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Laura Rubió
- Food Technology Department, Agrotecnio Center, University of Lleida, 25198 Lleida, Spain
| | - Sara Fernández-Castillejo
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
| | - Anna Pedret
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
| | - Ramón Estruch
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
- Internal Medicine Service, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - María Isabel Covas
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
- NUPROAS Handelsbolag, Nackă, Sweden
| | - Montserrat Fitó
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
| | - Álvaro Hernáez
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain.
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.
| | - Olga Castañer
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain.
| |
Collapse
|
17
|
Yang X, Wei C, Liu N, Wu F, Chen J, Wang C, Sun Z, Wang Y, Liu L, Zhang X, Wang B, Zhang Y, Zhong H, Han Y, He X. GP73, a novel TGF-β target gene, provides selective regulation on Smad and non-Smad signaling pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:588-597. [PMID: 30615900 DOI: 10.1016/j.bbamcr.2019.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 12/26/2022]
Abstract
Increased GP73 expression in hepatocytes from patients with acute hepatitis, through disease progression to cirrhosis and chronic liver disease suggests that progressive tissue remodeling and fibrogenesis are driving forces for GP73 upregulation. Nevertheless, details about regulation of GP73 expression and its biological functions remain elusive and await further characterization. In this study, we demonstrate that GP73 is a direct target of TGF-β1 transcriptional regulation. Its induced expression inhibits TGF-β-Smad mediated growth suppression. On the other hand, elevated GP73 results in upregulation of ERK/Akt signaling induced by TGF-β1. Mechanistically, upregulation of lipid raft and caveolae-1 induced by GP73 overexpression mediates its regulatory effect on TGF-β1 signaling. Notably, lipid raft expression is elevated in HCC tumors and tissues with higher GP73 expression yield more intensive Flotillin staining. Our results establish the linkage between GP73 and TGF-β signaling, indicating that GP73 may promote HCC tumorigenesis by selectively regulating TGF-β signaling through lipid raft modulation.
Collapse
Affiliation(s)
- Xiaoli Yang
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Ning Liu
- Jinzhou Medical University, Jinzhou 121001, China
| | - Feixiang Wu
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiankang Chen
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Cui Wang
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Zhenyu Sun
- Harbin Medical University, Harbin 150001, China
| | - Yufei Wang
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Liping Liu
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Xiaoli Zhang
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Beihan Wang
- Department of Clinical Laboratory, the Third Medical Centre, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100039, China
| | - Yanhong Zhang
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing 100850, China.
| | - Yue Han
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Xiang He
- Beijing Institute of Biotechnology, Beijing 100850, China.
| |
Collapse
|
18
|
Liu Y, Colby JK, Zuo X, Jaoude J, Wei D, Shureiqi I. The Role of PPAR-δ in Metabolism, Inflammation, and Cancer: Many Characters of a Critical Transcription Factor. Int J Mol Sci 2018; 19:3339. [PMID: 30373124 PMCID: PMC6275063 DOI: 10.3390/ijms19113339] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptor-delta (PPAR-δ), one of three members of the PPAR group in the nuclear receptor superfamily, is a ligand-activated transcription factor. PPAR-δ regulates important cellular metabolic functions that contribute to maintaining energy balance. PPAR-δ is especially important in regulating fatty acid uptake, transport, and β-oxidation as well as insulin secretion and sensitivity. These salutary PPAR-δ functions in normal cells are thought to protect against metabolic-syndrome-related diseases, such as obesity, dyslipidemia, insulin resistance/type 2 diabetes, hepatosteatosis, and atherosclerosis. Given the high clinical burden these diseases pose, highly selective synthetic activating ligands of PPAR-δ were developed as potential preventive/therapeutic agents. Some of these compounds showed some efficacy in clinical trials focused on metabolic-syndrome-related conditions. However, the clinical development of PPAR-δ agonists was halted because various lines of evidence demonstrated that cancer cells upregulated PPAR-δ expression/activity as a defense mechanism against nutritional deprivation and energy stresses, improving their survival and promoting cancer progression. This review discusses the complex relationship between PPAR-δ in health and disease and highlights our current knowledge regarding the different roles that PPAR-δ plays in metabolism, inflammation, and cancer.
Collapse
Affiliation(s)
- Yi Liu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Jennifer K Colby
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Xiangsheng Zuo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Jonathan Jaoude
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| |
Collapse
|
19
|
Aguirre-Portolés C, Feliu J, Reglero G, Ramírez de Molina A. ABCA1 overexpression worsens colorectal cancer prognosis by facilitating tumour growth and caveolin-1-dependent invasiveness, and these effects can be ameliorated using the BET inhibitor apabetalone. Mol Oncol 2018; 12:1735-1752. [PMID: 30098223 PMCID: PMC6166002 DOI: 10.1002/1878-0261.12367] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/26/2018] [Accepted: 07/29/2018] [Indexed: 02/05/2023] Open
Abstract
At the time of diagnosis, 20% of patients with colorectal cancer present metastasis. Among individuals with primary lesions, 50% of them will develop distant tumours with time. Therefore, early diagnosis and prediction of aggressiveness is crucial for therapy design and disease prognosis. Tumoral cells must undergo significant changes in energy metabolism to meet increased structural and energetic demands for cell proliferation, and metabolic alterations are considered to be a hallmark of cancer. Here, we present the ATP‐binding cassette transporter (ABCA1), a regulator of cholesterol transport, as a new marker for invasion and colorectal cancer survival. ABCA1 is significantly overexpressed in patients at advanced stages of colorectal cancer, and its overexpression confers proliferative advantages together with caveolin‐1 dependent‐increased migratory and invasive capacities. Thus, intracellular cholesterol imbalances mediated by ABCA1 overexpression may contribute to primary tumour growth and dissemination to distant locations. Furthermore, we demonstrate here that increased levels of apolipoprotein A1 (APOA1), a protein involved in cholesterol efflux and high‐density lipoprotein constitution, in the extracellular compartment modulates expression of ABCA1 by regulating COX‐2, and compensate for ABCA1‐dependent excessive export of cholesterol. APOA1 emerges as a new therapeutic option to inhibit the promotion of colorectal cancer to metastasis by modulating intracellular cholesterol metabolism. Furthermore, we propose apabetalone, an orally available small molecule that is currently being evaluated in clinical trials for the treatment of atherosclerosis, as a new putative therapeutic option to prevent colorectal cancer progression by increasing APOA1 expression and regulating reverse transport of cholesterol.
Collapse
Affiliation(s)
| | - Jaime Feliu
- Medical Oncology, La Paz University Hospital (IdiPAZ), CIBERONC, cátedra UAM-AMGEN, Madrid, Spain
| | - Guillermo Reglero
- Molecular Oncology, IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | | |
Collapse
|
20
|
ITGB1-dependent upregulation of Caveolin-1 switches TGFβ signalling from tumour-suppressive to oncogenic in prostate cancer. Sci Rep 2018; 8:2338. [PMID: 29402961 PMCID: PMC5799174 DOI: 10.1038/s41598-018-20161-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/15/2018] [Indexed: 01/10/2023] Open
Abstract
Caveolin-1 (CAV1) is over-expressed in prostate cancer (PCa) and is associated with adverse prognosis, but the molecular mechanisms linking CAV1 expression to disease progression are poorly understood. Extensive gene expression correlation analysis, quantitative multiplex imaging of clinical samples, and analysis of the CAV1-dependent transcriptome, supported that CAV1 re-programmes TGFβ signalling from tumour suppressive to oncogenic (i.e. induction of SLUG, PAI-1 and suppression of CDH1, DSP, CDKN1A). Supporting such a role, CAV1 knockdown led to growth arrest and inhibition of cell invasion in prostate cancer cell lines. Rationalized RNAi screening and high-content microscopy in search for CAV1 upstream regulators revealed integrin beta1 (ITGB1) and integrin associated proteins as CAV1 regulators. Our work suggests TGFβ signalling and beta1 integrins as potential therapeutic targets in PCa over-expressing CAV1, and contributes to better understand the paradoxical dual role of TGFβ in tumour biology.
Collapse
|
21
|
Martín-Martín N, Zabala-Letona A, Fernández-Ruiz S, Arreal L, Camacho L, Castillo-Martin M, Cortazar AR, Torrano V, Astobiza I, Zúñiga-García P, Ugalde-Olano A, Loizaga-Iriarte A, Unda M, Valcárcel-Jiménez L, Arruabarrena-Aristorena A, Piva M, Sánchez-Mosquera P, Aransay AM, Gomez-Muñoz A, Barrio R, Sutherland JD, Carracedo A. PPARδ Elicits Ligand-Independent Repression of Trefoil Factor Family to Limit Prostate Cancer Growth. Cancer Res 2017; 78:399-409. [DOI: 10.1158/0008-5472.can-17-0908] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 09/18/2017] [Accepted: 11/14/2017] [Indexed: 11/16/2022]
|
22
|
Xiong T, Xu G, Huang XL, Lu KQ, Xie WQ, Yin K, Tu J. ATP-binding cassette transporter A1: A promising therapy target for prostate cancer. Mol Clin Oncol 2017; 8:9-14. [PMID: 29399345 DOI: 10.3892/mco.2017.1506] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 11/10/2017] [Indexed: 12/21/2022] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) has been found to mediate the transfer of cellular cholesterol across the plasma membrane to apolipoprotein A-I (apoA-I), and is essential for the synthesis of high-density lipoprotein. Mutations of the ABCA1 gene may induce Tangier disease and familial hypoalphalipoproteinemia; they may also lead to loss of cellular cholesterol homeostasis in prostate cancer, and increased intracellular cholesterol levels are frequently found in prostate cancer cells. Recent studies have demonstrated that ABCA1 may exert anticancer effects through cellular cholesterol efflux, which has been attracting increasing attention in association with prostate cancer. The aim of the present review was to focus on the current views on prostate cancer progression and the various functions of ABCA1, in order to provide new therapeutic targets for prostate cancer.
Collapse
Affiliation(s)
- Ting Xiong
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Gang Xu
- Department of Diagnostics Teaching and Research, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xue-Long Huang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Kai-Qiang Lu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei-Quan Xie
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Kai Yin
- Department of Diagnostics Teaching and Research, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Tu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
23
|
PPAR-delta modulates membrane cholesterol and cytokine signaling in malignant B cells. Leukemia 2017; 32:184-193. [PMID: 28555083 DOI: 10.1038/leu.2017.162] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
A deeper understanding of the mechanisms that underlie aberrant signal transduction in B-cell cancers such as chronic lymphocytic leukemia (CLL) may reveal new treatment strategies. The lipid-activated nuclear receptor peroxisome proliferator-activated receptor delta (PPARδ) accounts for a number of properties of aggressive cancers and was found to enhance Janus kinase (JAK)-mediated phosphorylation of signal transducer and activator of transcription (STAT) proteins in B lymphoma cell lines and primary CLL cells. Autocrine production of cytokines such as IL10 and interferon-beta was not increased by PPARδ but signaling responses to these cytokines were amplified and associated with increased cholesterol biosynthesis and plasma membrane levels. Plasmalemmal cholesterol and STAT phosphorylation from type 1 interferons (IFNs) were increased by PPARδ agonists, transgenes and exogenous cholesterol, and decreased by cyclodextrin, PPARD deletion and chemical PPARδ inhibitors. Functional consequences of PPARδ-mediated perturbation of IFN signaling included impaired upregulation of co-stimulatory molecules. These observations suggest PPARδ modulates signaling processes in malignant B cells in part by altering cholesterol metabolism and changes the outcomes of signaling from cytokines such as IFNs. PPARδ antagonists may have therapeutic activity as anti-leukemic signal transduction modulators.
Collapse
|
24
|
Feng J, Zhang J, Jackson AO, Zhu X, Chen H, Chen W, Gui Q, Yin K. Apolipoprotein A1 Inhibits the TGF-β1-Induced Endothelial-to-Mesenchymal Transition of Human Coronary Artery Endothelial Cells. Cardiology 2017; 137:179-187. [PMID: 28434000 DOI: 10.1159/000464321] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/20/2017] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Transforming growth factor β1 (TGF-β1) is the major cytokine for stimulating endothelial cells (ECs) to transdifferentiate to mesenchymal cells (MCs) in the process known as endothelial-to-mesenchymal transition (EndMT). Recently, TGF-β1-induced EndMT has been implicated in the pathogenesis of atherosclerosis (AS). It has been identified that apolipoprotein A1 (ApoA-I) obstructs TGF-β1-induced endothelial dysfunction, providing a protective effect for ECs and also anti-AS activity. However, the exact role of ApoA-I in TGF-β1-induced EndMT is not clear. In this study, we aimed to investigate whether ApoA-I can modulate TGF-β1-induced EndMT in human coronary artery ECs (HCAECs). METHODS AND RESULTS The HCAECs were treated with TGF-β1 with or without ApoA-I. Morphological changes in HCAECs and the expression of EndMT-related markers were evaluated. HCAECs treated with TGF-β1 were found to transform to MC morphology, with inconspicuous expression of EC markers such as vascular endothelial cadherin and CD31, and conspicuous expression of fibroblast-specific protein 1 (FSP-1) and α-smooth muscle actin. The treatment of HCAECs with ApoA-I inhibited the TGF-β1-induced EndMT, and elevated expression of EC markers was observed but reduced expression of MC markers. Moreover, ApoA-I impeded the expression level of Slug and Snail, crucial transcriptional factors of EndMT, and it inhibited the TGF-β1-induced phosphorylation of Smad2 and Smad3 which affected the EC morphology. In addition, the knockdown of ABCA1 by RNA interference eliminated the inhibition effect of ApoA-I on TGF-β1-induced EndMT. CONCLUSIONS Our findings revealed a novel mechanism for the ApoA-I protective effect on endothelium function via the inhibition of TGF-β1-induced EndMT. This might provide new insights for developing strategies for modulating AS and vascular remodeling.
Collapse
Affiliation(s)
- Juling Feng
- Research Lab of Translational Medicine, Medical School, University of South China, Hengyang, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Zuo X, Xu W, Xu M, Tian R, Moussalli MJ, Mao F, Zheng X, Wang J, Morris JS, Gagea M, Eng C, Kopetz S, Maru DM, Rashid A, Broaddus R, Wei D, Hung MC, Sood AK, Shureiqi I. Metastasis regulation by PPARD expression in cancer cells. JCI Insight 2017; 2:e91419. [PMID: 28097239 DOI: 10.1172/jci.insight.91419] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptor-δ (PPARD) is upregulated in many major human cancers, but the role that its expression in cancer cells has in metastasis remains poorly understood. Here, we show that specific PPARD downregulation or genetic deletion of PPARD in cancer cells significantly repressed metastasis in various cancer models in vivo. Mechanistically, PPARD promoted angiogenesis via interleukin 8 in vivo and in vitro. Analysis of transcriptome profiling of HCT116 colon cancer cells with or without genetic deletion of PPARD and gene expression patterns in The Cancer Genome Atlas colorectal adenocarcinoma database identified novel pro-metastatic genes (GJA1, VIM, SPARC, STC1, SNCG) as PPARD targets. PPARD expression in cancer cells drastically affected epithelial-mesenchymal transition, migration, and invasion, further underscoring its necessity for metastasis. Clinically, high PPARD expression in various major human cancers (e.g., colorectal, lung, breast) was associated with significantly reduced metastasis-free survival. Our results demonstrate that PPARD, a druggable protein, is an important molecular target in metastatic cancer.
Collapse
Affiliation(s)
- Xiangsheng Zuo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Weiguo Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Surgical Oncology, Affiliated Hospital of Hebei United University, Tangshan, China
| | - Min Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rui Tian
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Fei Mao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Jing Wang
- Department of Bioinformatics and Computational Biology
| | | | - Mihai Gagea
- Department of Veterinary Medicine and Surgery
| | - Cathy Eng
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, and.,Department of Cancer Biology and.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
26
|
Sun N, Taguchi A, Hanash S. Switching Roles of TGF-β in Cancer Development: Implications for Therapeutic Target and Biomarker Studies. J Clin Med 2016; 5:jcm5120109. [PMID: 27916872 PMCID: PMC5184782 DOI: 10.3390/jcm5120109] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 12/15/2022] Open
Abstract
TGF-β induces complicated and even opposite responses in numerous biological processes, e.g., tumor suppression in pre-malignant cells and metastasis promotion in cancer cells. However, the cellular contextual determinants of these different TGF-β roles remain elusive, and the driver genes triggering the determinants’ changes have not been identified. Recently, however, several findings have provided new insights on the contextual determinants of Smads in TGF-β’s biological processes. These novel switches and their effectors may serve as prognostic biomarkers and therapeutic targets of TGF-β-mediated cancer progression.
Collapse
Affiliation(s)
- Nan Sun
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Ayumu Taguchi
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Samir Hanash
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Ke T, Dorajoo R, Han Y, Khor CC, van Dam RM, Yuan JM, Koh WP, Liu J, Teo YY, Goh DYT, Tai ES, Wong TY, Cheng CY, Friedlander Y, Heng CK. Interaction Between Peroxisome Proliferator Activated Receptor δ and Epithelial Membrane Protein 2 Polymorphisms Influences HDL-C Levels in the Chinese Population. Ann Hum Genet 2016; 80:282-93. [PMID: 27530449 DOI: 10.1111/ahg.12164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/09/2016] [Accepted: 06/13/2016] [Indexed: 11/30/2022]
Abstract
Peroxisome proliferator activated receptors (PPARs) are transcription factors involved in the regulation of key metabolic pathways. Numerous in vivo and in vitro studies have established their important roles in lipid metabolism. A few SNPs in PPAR genes have been reported to be associated with lipid levels. In this study, we aimed to investigate the interactive effects between single nucleotide polymorphisms (SNPs) in three PPAR isoforms α/δ/γ and other genetic variants across the genome on plasma high-density lipoprotein-cholesterol (HDL-C) levels. Study subjects (N = 2003) were genotyped using Illumina HumanOmniZhongHua-8 Beadchip. Fifty-three tag SNPs ± 100 kb of PPAR α, δ, and γ (r(2) < 0.2) were selected. The effect of interactions between PPAR SNPs and those across the genome on HDL-C was tested using linear regression models. One statistically significant interaction influencing HDL-C was detected between PPARδ SNP rs2267668 and epithelial membrane protein 2 (EMP2) downstream SNP rs7191411 (N = 1993, β = 0.74, adjusted P = 0.022). This interaction was successfully replicated in the meta-analysis of two additional Chinese cohorts (N = 3948, P = 0.01). The present study showed a novel SNP × SNP interaction between rs2267668 in PPARδ and rs7191411 in EMP2 that has significant impact on circulating HDL-C levels in the Singaporean Chinese population.
Collapse
Affiliation(s)
- Tingjing Ke
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore and Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Yi Han
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore and Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore
| | - Chiea-Chuen Khor
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore.,Duke-NUS Graduate Medical School, Singapore
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore.,Department of Statistics and Applied Probability, National University of Singapore, Singapore
| | - Daniel Y T Goh
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore and Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Tien Yin Wong
- Singapore National Eye Centre, Singapore Eye Research Institute, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore National Eye Centre, Singapore Eye Research Institute, Singapore.,Academic Medicine Research Institute, Duke-NUS Graduate Medical School, National University of Singapore, Singapore
| | - Yechiel Friedlander
- School of Public Health and Community Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore and Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore
| |
Collapse
|
28
|
Fletcher JI, Williams RT, Henderson MJ, Norris MD, Haber M. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist Updat 2016; 26:1-9. [PMID: 27180306 DOI: 10.1016/j.drup.2016.03.001] [Citation(s) in RCA: 307] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 03/04/2016] [Accepted: 03/12/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Rebekka T Williams
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Michelle J Henderson
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia.
| |
Collapse
|
29
|
Sica GS, Fiorani C, Stolfi C, Monteleone G, Candi E, Amelio I, Catani V, Sibio S, Divizia A, Tema G, Iaculli E, Gaspari AL. Peritoneal expression of Matrilysin helps identify early post-operative recurrence of colorectal cancer. Oncotarget 2015; 6:13402-13415. [PMID: 25596746 PMCID: PMC4537023 DOI: 10.18632/oncotarget.2830] [Citation(s) in RCA: 18] [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: 11/12/2014] [Accepted: 11/27/2014] [Indexed: 12/20/2022] Open
Abstract
Recurrence of colorectal cancer (CRC) following a potentially curative resection is a challenging clinical problem. Matrix metalloproteinase-7 (MMP-7) is over-expressed by CRC cells and supposed to play a major role in CRC cell diffusion and metastasis. MMP-7 RNA expression was assessed by real-time PCR using specific primers in peritoneal washing fluid obtained during surgical procedure. After surgery, patients underwent a regular follow up for assessing recurrence. transcripts for MMP-7 were detected in 31/57 samples (54%). Patients were followed-up (range 20-48 months) for recurrence prevention. Recurrence was diagnosed in 6 out of 55 patients (11%) and two patients eventually died because of this. Notably, all the six patients who had relapsed were positive for MMP-7. Sensitivity and specificity of the test were 100% and 49% respectively. Data from patients have also been corroborated by computational approaches. Public available coloncarcinoma datasets have been employed to confirm MMP7 clinical impact on the disease. Interestingly, MMP-7 expression appeared correlated to Tgfb-1, and correlation of the two factors represented a poor prognostic factor. This study proposes positivity of MMP-7 in peritoneal cavity as a novel biomarker for predicting disease recurrence in patients with CRC.
Collapse
Affiliation(s)
- Giuseppe S. Sica
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
- European Society Degenerative Disease (ESDD). www.esdd.it
| | - Cristina Fiorani
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Carmine Stolfi
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Giovanni Monteleone
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, Leicester, UK
| | - Valeria Catani
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Simone Sibio
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Andrea Divizia
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Giorgia Tema
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Edoardo Iaculli
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Achille L. Gaspari
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| |
Collapse
|
30
|
Ryter SW, Choi AMK, Kim HP. Profibrogenic phenotype in caveolin-1 deficiency via differential regulation of STAT-1/3 proteins. Biochem Cell Biol 2014; 92:370-8. [PMID: 25263949 DOI: 10.1139/bcb-2014-0075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Fibrosis underlies the pathogenesis of several human diseases, which can lead to severe injury of vital organs. We previously demonstrated that caveolin-1 expression is reduced in experimental fibrosis and that caveolin-1 exerts antiproliferative and antifibrotic effects in lung fibrosis models. The signal transducers and activators of transcription (STAT) proteins, STAT1 and STAT3, can be activated simultaneously. STAT1 can inhibit cell growth and promote apoptosis while STAT3 inhibits apoptosis. Here, we show that caveolin-1-deficient (cav-1(-/-)) lung fibroblasts display dramatically upregulated STAT3 activation in response to platelet-derived growth factor-BB and transforming growth factor-β stimuli, whereas STAT1 activation is undetectable. Downregulation of protein tyrosine phosphatase-1B played a role in the preferential activation of STAT3 in cav-1(-/-) fibroblasts. Genetic deletion of STAT3 by siRNA modulated the expression of genes involved in cell proliferation and fibrogenesis. Basal expression of α-smooth muscle actin was prominent in cav-1(-/-) liver and kidney, consistent with deposition of collagen in these organs. Collectively, we demonstrate that the antiproliferative and antifibrogenic properties of caveolin-1 in vitro are mediated by the balance between STAT1 and STAT3 activation. Deregulated STAT signaling associated with caveolin-1 deficiency may be relevant to proliferative disorders such as tissue fibrosis.
Collapse
Affiliation(s)
- Stefan W Ryter
- a Division of Pulmonary and Critical Care Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|