1
|
Ruiz Luque J, Cevey ÁC, Pieralisi AV, Poncini C, Erra Díaz F, Azevedo Reis MV, Donato M, Mirkin GA, Goren NB, Penas FN. Fenofibrate Induces a Resolving Profile in Heart Macrophage Subsets and Attenuates Acute Chagas Myocarditis. ACS Infect Dis 2024; 10:1793-1807. [PMID: 38648355 DOI: 10.1021/acsinfecdis.4c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Chagas disease, caused by Trypanosoma cruzi, stands as the primary cause of dilated cardiomyopathy in the Americas. Macrophages play a crucial role in the heart's response to infection. Given their functional and phenotypic adaptability, manipulating specific macrophage subsets could be vital in aiding essential cardiovascular functions including tissue repair and defense against infection. PPARα are ligand-dependent transcription factors involved in lipid metabolism and inflammation regulation. However, the role of fenofibrate, a PPARα ligand, in the activation profile of cardiac macrophages as well as its effect on the early inflammatory and fibrotic response in the heart remains unexplored. The present study demonstrates that fenofibrate significantly reduces not only the serum activity of tissue damage biomarker enzymes (LDH and GOT) but also the circulating proportions of pro-inflammatory monocytes (CD11b+ LY6Chigh). Furthermore, both CD11b+ Ly6Clow F4/80high macrophages (MΦ) and recently differentiated CD11b+ Ly6Chigh F4/80high monocyte-derived macrophages (MdMΦ) shift toward a resolving phenotype (CD206high) in the hearts of fenofibrate-treated mice. This shift correlates with a reduction in fibrosis, inflammation, and restoration of ventricular function in the early stages of Chagas disease. These findings encourage the repositioning of fenofibrate as a potential ancillary immunotherapy adjunct to antiparasitic drugs, addressing inflammation to mitigate Chagas disease symptoms.
Collapse
Affiliation(s)
- Javier Ruiz Luque
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| | - Ágata Carolina Cevey
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| | - Azul Victoria Pieralisi
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| | - Carolina Poncini
- CONICET - Universidad de Buenos Aires. Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires C1121A6B, Argentina
| | - Fernando Erra Díaz
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| | - Marcus Vinicius Azevedo Reis
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| | - Martin Donato
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Fisiopatología Cardiovascular (INFICA), Buenos Aires C1121A6B, Argentina
| | - Gerardo Ariel Mirkin
- CONICET - Universidad de Buenos Aires. Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires C1121A6B, Argentina
| | - Nora Beatriz Goren
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| | - Federico Nicolás Penas
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires C1121A6B, Argentina
| |
Collapse
|
2
|
Trejo-Solís C, Castillo-Rodríguez RA, Serrano-García N, Silva-Adaya D, Vargas-Cruz S, Chávez-Cortéz EG, Gallardo-Pérez JC, Zavala-Vega S, Cruz-Salgado A, Magaña-Maldonado R. Metabolic Roles of HIF1, c-Myc, and p53 in Glioma Cells. Metabolites 2024; 14:249. [PMID: 38786726 PMCID: PMC11122955 DOI: 10.3390/metabo14050249] [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: 04/01/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/25/2024] Open
Abstract
The metabolic reprogramming that promotes tumorigenesis in glioblastoma is induced by dynamic alterations in the hypoxic tumor microenvironment, as well as in transcriptional and signaling networks, which result in changes in global genetic expression. The signaling pathways PI3K/AKT/mTOR and RAS/RAF/MEK/ERK stimulate cell metabolism, either directly or indirectly, by modulating the transcriptional factors p53, HIF1, and c-Myc. The overexpression of HIF1 and c-Myc, master regulators of cellular metabolism, is a key contributor to the synthesis of bioenergetic molecules that mediate glioma cell transformation, proliferation, survival, migration, and invasion by modifying the transcription levels of key gene groups involved in metabolism. Meanwhile, the tumor-suppressing protein p53, which negatively regulates HIF1 and c-Myc, is often lost in glioblastoma. Alterations in this triad of transcriptional factors induce a metabolic shift in glioma cells that allows them to adapt and survive changes such as mutations, hypoxia, acidosis, the presence of reactive oxygen species, and nutrient deprivation, by modulating the activity and expression of signaling molecules, enzymes, metabolites, transporters, and regulators involved in glycolysis and glutamine metabolism, the pentose phosphate cycle, the tricarboxylic acid cycle, and oxidative phosphorylation, as well as the synthesis and degradation of fatty acids and nucleic acids. This review summarizes our current knowledge on the role of HIF1, c-Myc, and p53 in the genic regulatory network for metabolism in glioma cells, as well as potential therapeutic inhibitors of these factors.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Departamento de Neurofisiología, Laboratorio Clínico y Banco de Sangre y Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (N.S.-G.); (D.S.-A.); (S.Z.-V.)
| | | | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Departamento de Neurofisiología, Laboratorio Clínico y Banco de Sangre y Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (N.S.-G.); (D.S.-A.); (S.Z.-V.)
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Departamento de Neurofisiología, Laboratorio Clínico y Banco de Sangre y Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (N.S.-G.); (D.S.-A.); (S.Z.-V.)
- Centro de Investigación Sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), Ciudad de Mexico 14330, Mexico
| | - Salvador Vargas-Cruz
- Departamento de Cirugía, Hospital Ángeles del Pedregal, Camino a Sta. Teresa, Ciudad de Mexico 10700, Mexico;
| | | | - Juan Carlos Gallardo-Pérez
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de Mexico 14080, Mexico;
| | - Sergio Zavala-Vega
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Departamento de Neurofisiología, Laboratorio Clínico y Banco de Sangre y Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (N.S.-G.); (D.S.-A.); (S.Z.-V.)
| | - Arturo Cruz-Salgado
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Roxana Magaña-Maldonado
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Departamento de Neurofisiología, Laboratorio Clínico y Banco de Sangre y Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (N.S.-G.); (D.S.-A.); (S.Z.-V.)
| |
Collapse
|
3
|
Alizadehasl A, Alavi MS, Boudagh S, Alavi MS, Mohebi S, Aliabadi L, Akbarian M, Ahmadi P, Mannarino MR, Sahebkar A. Lipid-lowering drugs and cancer: an updated perspective. Pharmacol Rep 2024; 76:1-24. [PMID: 38015371 DOI: 10.1007/s43440-023-00553-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/29/2023]
Abstract
Statins and non-statin medications used for the management of dyslipidemia have been shown to possess antitumor properties. Since the use of these drugs has steadily increased over the past decades, more knowledge is required about their relationship with cancer. Lipid-lowering agents are heterogeneous compounds; therefore, it remains to be revealed whether anticancer potential is a class effect or related to them all. Here, we reviewed the literature on the influence of lipid-lowering medications on various types of cancer during development or metastasis. We also elaborated on the underlying mechanisms associated with the anticancer effects of antihyperlipidemic agents by linking the reported in vivo and in vitro studies.
Collapse
Affiliation(s)
- Azin Alizadehasl
- Cardio-Oncology Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sadat Alavi
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Boudagh
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Somaye Mohebi
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Aliabadi
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Akbarian
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Ahmadi
- Echocardiography Research CenterRajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Jin J, Liang X, Bi W, Liu R, Zhang S, He Y, Xie Q, Liu S, Xiao JC, Zhang P. Identification of a Difluorinated Alkoxy Sulfonyl Chloride as a Novel Antitumor Agent for Hepatocellular Carcinoma through Activating Fumarate Hydratase Activity. Pharmaceuticals (Basel) 2023; 16:1705. [PMID: 38139831 PMCID: PMC10748328 DOI: 10.3390/ph16121705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Fenofibrate is known as a lipid-lowering drug. Although previous studies have reported that fenofibrate exhibits potential antitumor activities, IC50 values of fenofibrate could be as high as 200 μM. Therefore, we investigated the antitumor activities of six synthesized fenofibrate derivatives. We discovered that one compound, SIOC-XJC-SF02, showed significant antiproliferative activity on human hepatocellular carcinoma (HCC) HCCLM3 cells and HepG2 cells (the IC50 values were 4.011 μM and 10.908 μM, respectively). We also found this compound could inhibit the migration of human HCC cells. Transmission electron microscope and flow cytometry assays demonstrated that this compound could induce apoptosis of human HCC cells. The potential binding sites of this compound acting on human HCC cells were identified by mass spectrometry-cellular thermal shift assay (MS-CETSA). Molecular docking, Western blot, and enzyme activity assay-validated binding sites in human HCC cells. The results showed that fumarate hydratase may be a potential binding site of this compound, exerting antitumor effects. A xenograft model in nude mice demonstrated the anti-liver cancer activity and the mechanism of action of this compound. These findings indicated that the antitumor effect of this compound may act via activating fumarate hydratase, and this compound may be a promising antitumor candidate for further investigation.
Collapse
Affiliation(s)
- Jin Jin
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xujun Liang
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wu Bi
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ruijie Liu
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Sai Zhang
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yi He
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qingming Xie
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shilei Liu
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ji-Chang Xiao
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Pengfei Zhang
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
5
|
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
|
6
|
Guan B, Li H, Yao J, Guo J, Yu F, Li G, Wan B, Ma J, Huang D, Sun L, Chen Y. CCL3-CCR5 axis promotes cell migration and invasion of colon adenocarcinoma via Akt signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:172-184. [PMID: 36346222 DOI: 10.1002/tox.23675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/19/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Infiltration of tumor-associated macrophages (TAMs) can promote tumorigenesis and development. C-C motif chemokine ligand 3 (CCL3) was reported to be derived from TAMs and tumor cells and facilitate the progression of several cancers. Nevertheless, whether CCL3 can be derived from TAMs and tumor cells of colon adenocarcinoma (COAD) is unclarified. METHODS Peripheral blood monocytes-derived macrophages were polarized by the conditioned medium from COAD cells to establish TAM-like macrophages (TAM1/2). RT-qPCR and western blotting were used for detection of expression levels of CCL3 and its receptors C-C motif chemokine receptor 1 (CCR1) and CCR5 in TAM1/2 and COAD cells. Immunofluorescence staining was utilized for evaluating CCL3, CD163 and CCR5 expression. The Akt signaling pathway-associated protein levels were measured by western blotting. Transwell assays were used for assessing cell migration and invasiveness. RESULTS CCL3 displayed a high level in TAMs and cancer cells of COAD. CCL3 activated the Akt signaling pathway by binding to CCR5. CCL3-CCR5 axis facilitated COAD cell migration and invasiveness by activating the Akt signaling. CCL3 derived from both TAMs and cancer cells contributed to the malignant behaviors of COAD cells. High expression of CCL3/CCR5 was closely associated with poor prognoses of COAD patients. CONCLUSION CCL3-CCR5 interaction promotes cell migration and invasiveness, and functions as a prognostic biomarker for COAD.
Collapse
Affiliation(s)
- Bugao Guan
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Hongbo Li
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jian Yao
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jinbao Guo
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Fei Yu
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Guangrun Li
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Benhai Wan
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jun Ma
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Desong Huang
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Lu Sun
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Yan Chen
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| |
Collapse
|
7
|
Su CC, Lin JW, Chang KY, Wu CT, Liu SH, Chang KC, Liu JM, Lee KI, Fang KM, Chen YW. Involvement of AMPKα and MAPK-ERK/-JNK Signals in Docetaxel-Induced Human Tongue Squamous Cell Carcinoma Cell Apoptosis. Int J Mol Sci 2022; 23:ijms232213857. [PMID: 36430348 PMCID: PMC9696237 DOI: 10.3390/ijms232213857] [Citation(s) in RCA: 4] [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: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Cancers of the oral cavity can develop in the anatomic area extending from the lip, gum, tongue, mouth, and to the palate. Histologically, about 85-90% of oral cavity cancers are of the type squamous cells carcinomas (SCCs). The incidence of oral tongue SCC is higher in the tongue than any other anatomic area of the oral cavity. Here, we investigated the therapeutic effects and molecular mechanisms of docetaxel, which is a paclitaxel antitumor agent, on the cell growth of a human tongue SCC-derived SAS cell line. The results showed that docetaxel (10-300 nM) induced cytotoxicity and caspase-3 activity in SAS cells. Moreover, docetaxel (100 nM) promoted the expression of apoptosis-related signaling molecules, including the cleavages of caspase-3, caspase-7, and poly (ADP-ribose) polymerase (PARP). In mitochondria, docetaxel (100 nM) decreased the mitochondrial membrane potential (MMP) and Bcl-2 mRNA and protein expression and increased cytosolic cytochrome c protein expression and Bax mRNA and protein expression. In terms of mitogen-activated protein kinase (MAPK) and adenosine monophosphate-activated protein kinase (AMPK) signaling, docetaxel increased the expression of phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-c-Jun N-terminal kinase (JNK), and p-AMPKα protein expression but not p-p38 protein expression. Moreover, the increase in caspase-3/-7 activity and Bax protein expression and decreased Bcl-2 protein expression and MMP depolarization observed in docetaxel-treated SAS cells could be reversed by treatment with either SP600125 (a JNK inhibitor), PD98059 (an MEK1/2 (mitogen-activated protein kinase kinase 1/2) inhibitor), or compound c (an AMPK inhibitor). The docetaxel-induced increases in p-JNK, p-ERK, and p-AMPKα protein expression could also be reversed by treatment with either SP600125, PD98059, or compound c. These results indicate that docetaxel induces human tongue SCC cell apoptosis via interdependent MAPK-JNK, MAPK-ERK1/2, and AMPKα signaling pathways. Our results show that docetaxel could possibly exert a potent pharmacological effect on human oral tongue SCC cell growth.
Collapse
Affiliation(s)
- Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County, Changhua County 500, Taiwan
| | - Jhe-Wei Lin
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Kai-Yao Chang
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Cheng-Tien Wu
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kai-Chih Chang
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Jui-Ming Liu
- Department of Urology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Kai-Min Fang
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Correspondence: (K.-M.F.); (Y.-W.C.)
| | - Ya-Wen Chen
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: (K.-M.F.); (Y.-W.C.)
| |
Collapse
|
8
|
Renu K, Veeraraghavan VP, Raj A T, Patil S, Valsala Gopalakrishnan A. The peroxisome proliferator-activated receptor-alpha (PPAR-α): A new therapeutic target for oral cancer. Oral Oncol 2022; 132:106007. [PMID: 35809505 DOI: 10.1016/j.oraloncology.2022.106007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600 077, India.
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600 077, India.
| | - Thirumal Raj A
- Department of Oral Pathology and Microbiology, Sri Venkateswara Dental College and Hospital, Chennai 600 130, India
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Jazan 45142, Saudi Arabia
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
9
|
Compound C Inhibits Renca Renal Epithelial Carcinoma Growth in Syngeneic Mouse Models by Blocking Cell Cycle Progression, Adhesion and Invasion. Int J Mol Sci 2022; 23:ijms23179675. [PMID: 36077072 PMCID: PMC9456023 DOI: 10.3390/ijms23179675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Compound C (CompC), an inhibitor of AMP-activated protein kinase, reduces the viability of various renal carcinoma cells. The molecular mechanism underlying anti-proliferative effect was investigated by flow cytometry and western blot analysis in Renca cells. Its effect on the growth of Renca xenografts was also examined in a syngeneic BALB/c mouse model. Subsequent results demonstrated that CompC reduced platelet-derived growth factor receptor signaling pathways and increased ERK1/2 activation as well as reactive oxygen species (ROS) production. CompC also increased the level of active Wee1 tyrosine kinase (P-Ser642-Wee1) and the inactive form of Cdk1 (P-Tyr15-Cdk1) while reducing the level of active histone H3 (P-Ser10-H3). ROS-dependent ERK1/2 activation and sequential alterations in Wee1, Cdk1, and histone H3 might be responsible for the CompC-induced G2/M cell cycle arrest and cell viability reduction. In addition, CompC reduced the adhesion, migration, and invasion of Renca cells in the in vitro cell systems, and growth of Renca xenografts in the BALB/c mouse model. Taken together, the inhibition of in vivo tumor growth by CompC may be attributed to the blockage of cell cycle progression, adhesion, migration, and invasion of tumor cells. These findings suggest the therapeutic potential of CompC against tumor development and progression.
Collapse
|
10
|
Gangwar SK, Kumar A, Jose S, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Nuclear receptors in oral cancer-emerging players in tumorigenesis. Cancer Lett 2022; 536:215666. [DOI: 10.1016/j.canlet.2022.215666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 12/24/2022]
|
11
|
Su TR, Yu CC, Chao SC, Huang CC, Liao YW, Hsieh PL, Yu CH, Lin SS. Fenofibrate diminishes the self-renewal and metastasis potentials of oral carcinoma stem cells through NF-κB signaling. J Formos Med Assoc 2022; 121:1900-1907. [DOI: 10.1016/j.jfma.2022.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/24/2022] Open
|
12
|
Target Deconvolution of Fenofibrate in Nonalcoholic Fatty Liver Disease Using Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2022; 2021:3654660. [PMID: 34988225 PMCID: PMC8720586 DOI: 10.1155/2021/3654660] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/12/2021] [Accepted: 12/14/2021] [Indexed: 01/30/2023]
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is a prevalent form of liver damage, affecting ~25% of the global population. NAFLD comprises a spectrum of liver pathologies, from hepatic steatosis to nonalcoholic steatohepatitis (NASH), and may progress to liver fibrosis and cirrhosis. The presence of NAFLD correlates with metabolic disorders such as hyperlipidemia, obesity, blood hypertension, cardiovascular, and insulin resistance. Fenofibrate is an agonist drug for peroxisome proliferator-activated receptor alpha (PPARα), used principally for treatment of hyperlipidemia. However, fenofibrate has recently been investigated in clinical trials for treatment of other metabolic disorders such as diabetes, cardiovascular disease, and NAFLD. The evidence to date indicates that fenofibrate could improve NAFLD. While PPARα is considered to be the main target of fenofibrate, fenofibrate may exert its effect through impact on other genes and pathways thereby alleviating, and possibly reversing, NAFLD. In this study, using bioinformatics tools and gene-drug, gene-diseases databases, we sought to explore possible targets, interactions, and pathways involved in fenofibrate and NAFLD. Methods We first determined significant protein interactions with fenofibrate in the STITCH database with high confidence (0.7). Next, we investigated the identified proteins on curated targets in two databases, including the DisGeNET and DISEASES databases, to determine their association with NAFLD. We finally constructed a Venn diagram for these two collections (curated genes-NAFLD and fenofibrate-STITCH) to uncover possible primary targets of fenofibrate. Then, Gene Ontology (GO) and KEGG were analyzed to detect the significantly involved targets in molecular function, biological process, cellular component, and biological pathways. A P value < 0.01 was considered the cut-off criterion. We also estimated the specificity of targets with NAFLD by investigating them in disease-gene associations (STRING) and EnrichR (DisGeNET). Finally, we verified our findings in the scientific literature. Results We constructed two collections, one with 80 protein-drug interactions and the other with 95 genes associated with NAFLD. Using the Venn diagram, we identified 11 significant targets including LEP, SIRT1, ADIPOQ, PPARA, SREBF1, LDLR, GSTP1, VLDLR, SCARB1, MMP1, and APOC3 and then evaluated their biological pathways. Based on Gene Ontology, most of the targets are involved in lipid metabolism, and KEGG enrichment pathways showed the PPAR signaling pathway, AMPK signaling pathway, and NAFLD as the most significant pathways. The interrogation of those targets on authentic disease databases showed they were more specific to both steatosis and steatohepatitis liver injury than to any other diseases in these databases. Finally, we identified three significant genes, APOC3, PPARA, and SREBF1, that showed robust drug interaction with fenofibrate. Conclusion Fenofibrate may exert its effect directly or indirectly, via modulation of several key targets and pathways, in the treatment of NAFLD.
Collapse
|
13
|
Repositioning Fenofibrate to Reactivate p53 and Reprogram the Tumor-Immune Microenvironment in HPV+ Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14020282. [PMID: 35053444 PMCID: PMC8773501 DOI: 10.3390/cancers14020282] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary A critical need for optimal management of human papillomavirus-associated head and neck squamous cell carcinoma (HPV+ HNSCC) patients is the development of therapeutic strategies to exploit the inherent vulnerabilities of this unique disease. We identified fenofibrate, an FDA-approved drug, as a potent anti-cancer agent for HPV+ HNSCC. Fenofibrate induced the accumulation of the p53 tumor suppressor and re-programmed the tumor microenvironment to drive immune cell infiltration. We provide compelling evidence to reposition fenofibrate as a single agent or in combination with standard therapies for the HPV+ HNSCC setting. Abstract Human papillomavirus-associated head and neck squamous cell carcinoma (HPV+ HNSCC) is recognized as a distinct disease with unique etiology and clinical features. Current standard of care therapeutic modalities are identical for HPV+ and HPV− HNSCC and thus, there remains an opportunity to develop innovative pharmacologic approaches to exploit the inherent vulnerabilities of HPV+ HNSCC. In this study, using an inducible HPVE6E7 knockdown system, we found that HPV+ HNSCC cells are addicted to HPVE6E7, such that loss of these viral oncogenes impaired tumorigenicity in vitro and in vivo. A number of druggable pathways, including PPAR and Wnt, were modulated in response to HPVE6E7 loss. Fenofibrate showed significant anti-proliferative effects in a panel of HPV+ cancer cell lines. Additionally, fenofibrate impaired tumor growth as monotherapy and potentiated the activity of cisplatin in a pre-clinical HPV+ animal model. Systemic fenofibrate treatment induced p53 protein accumulation, and surprisingly, re-programmed the tumor-immune microenvironment to drive immune cell infiltration. Since fenofibrate is FDA-approved with a favorable long-term safety record, repositioning of this drug, as a single agent or in combination with cisplatin or checkpoint blockade, for the HPV+ HNSCC setting should be prioritized.
Collapse
|
14
|
Down-Regulation of miR-194-5p for Predicting Metastasis in Breast Cancer Cells. Int J Mol Sci 2021; 23:ijms23010325. [PMID: 35008751 PMCID: PMC8745262 DOI: 10.3390/ijms23010325] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs), as key negative regulators of gene expression, are closely related to tumor occurrence and progression. miR-194-5p (miR-194-1) has been shown to play a regulatory role in various cancers however, its biological function and mechanism of action in breast cancer have not yet been well explored. In this study, we use the UALCAN and LinkedOmics databases to analyze transcription expression in The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA). The epithelial-mesenchymal transition status of breast cancer cells was evaluated by wound-healing assay, trans-well assays, and gelatin zymography, while protein expression was assessed by Western blotting. miR-194-5p expression was found to be up-regulated in breast cancer clinical specimens but down-regulated in the triple-negative breast cancer (TNBC) cell line MDA-MB-231 and breast cancer clinical specimens in The Cancer Genome Atlas (TCGA). miR-194-5p significantly inhibited the expression of the epithelial marker ZO-1 and increased the expression of mesenchymal markers, including ZEB-1 and vimentin, in MDA-MB-231 cells. miR-194-5p significantly reduced the gelatin-degrading activity of matrix metalloproteinase-2 (MMP-2) and MMP-9 in zymography assays. In MDA-MB-231 cells and TCGA patient samples, ZEB-1 expression was significantly inversely correlated with miR-194-5p expression. High levels of miR-194-5p were associated with good overall survival. miR-194-5p regulates epithelial–mesenchymal transition (EMT) in TNBC. Our findings suggest that miR-194-5p functions as a tumor biomarker in breast cancer, providing new insights for the study of breast cancer development and metastasis.
Collapse
|
15
|
Lin C, Lai SW, Shen CK, Chen CW, Tsai CF, Liu YS, Lu DY, Huang BR. Fenofibrate inhibits hypoxia-inducible factor-1 alpha and carbonic anhydrase expression through activation of AMP-activated protein kinase/HO-1/Sirt1 pathway in glioblastoma cells. ENVIRONMENTAL TOXICOLOGY 2021; 36:2551-2561. [PMID: 34520103 DOI: 10.1002/tox.23369] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 06/13/2023]
Abstract
Cancer and its associated conditions have significant impacts on public health at many levels worldwide, and cancer is the leading cause of death among adults. Peroxisome proliferator-activated receptor α (PPARα)-specific agonists, fibrates, have been approved by the Food and Drug Administration for managing hyperlipidemia. PPARα-specific agonists exert anti-cancer effects in many human cancer types, including glioblastoma (GBM). Recently, we have reported that the hypoxic state in GBM stabilizes hypoxia-inducible factor-1 alpha (HIF-1α), thus contributing to tumor escape from immune surveillance by activating the expression of the pH-regulating protein carbonic anhydrase IX (CA9). In this study, we aimed to study the regulatory effects of the PPARα agonist fibrate on the regulation of HIF-1α expression and its downstream target protein in GBM. Our findings showed that fenofibrate is the high potency compound among the various fibrates that inhibit hypoxia-induced HIF-1α and CA9 expression in GBM. Moreover, fenofibrate-inhibited HIF-1α expression is mediated by HO-1 activation in GBM cells through the AMP-activated protein kinase (AMPK) pathway. In addition, fenofibrate-enhanced HO-1 upregulation activates SIRT1 and leads to subsequent accumulation of SIRT1 in the nucleus, which further promotes HIF-1α deacetylation and inhibits CA9 expression. Using a protein synthesis inhibitor, cycloheximide, we also observed that fenofibrate inhibited HIF-1α protein synthesis. In addition, the administration of the proteasome inhibitor MG132 showed that fenofibrate promoted HIF-1α protein degradation in GBM. Hence, our results indicate that fenofibrate is a useful anti-GBM agent that modulates hypoxia-induced HIF-1α expression through multiple cellular pathways.
Collapse
Affiliation(s)
- Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Sheng-Wei Lai
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Kai Shen
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chao-Wei Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
16
|
Tsai SC, Wu WC, Yang JS. Tetrandrine Inhibits Epithelial-Mesenchymal Transition in IL-6-Induced HCT116 Human Colorectal Cancer Cells. Onco Targets Ther 2021; 14:4523-4536. [PMID: 34456573 PMCID: PMC8387317 DOI: 10.2147/ott.s324552] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction Patients with colorectal cancer (CRC) often develop distant metastases, which significantly reduces the 5-year survival rate. Epithelial-mesenchymal transition (EMT) is a crucial process for the invasion and metastasis of cancer cells. Tetrandrine has been reported to inhibit the viability and EMT of CRC cells; however, to the best of our knowledge, the molecular mechanism remains undetermined. Methods The MTT assay was used to determine HCT116 cell viability. Wound healing and Transwell assays were used to determine that cell migration and invasion, respectively. Western blotting analysis was performed to detect the expression of migration-related genes. Four different lengths of the E-cadherin gene promoter were constructed and cloned into pGL3 reporter plasmids to evaluate E-cadherin gene promoter activity. Results The results of the MTT assay revealed that tetrandrine inhibited HCT116 cell viability, with an IC50 value of 7.2 μM following 24 h of treatment. Tetrandrine inhibited IL-6-induced cell migration and invasion, respectively. Tetrandrine regulates the expression of migration-related genes in IL-6-stimulated HCT116 cells. Tetrandrine significantly downregulated the expression and enzyme activity of MMP-2 in IL-6-stimulated HCT116 cells. In addition, tetrandrine restored E-cadherin gene promoter activity. Conclusion The findings of the present study suggested that tetrandrine may inhibit EMT in IL-6-stimulated HCT116 cells; therefore, it may represent a potential drug for CRC.
Collapse
Affiliation(s)
- Shih-Chang Tsai
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, Republic of China
| | - Wei-Chei Wu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, Republic of China
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
17
|
Qiao X, Gao YY, Zheng LX, Ding XJ, Xu LW, Hu JJ, Gao WZ, Xu JY. Targeting ROS-AMPK pathway by multiaction Platinum(IV) prodrugs containing hypolipidemic drug bezafibrate. Eur J Med Chem 2021; 223:113730. [PMID: 34388483 DOI: 10.1016/j.ejmech.2021.113730] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022]
Abstract
Alterations in lipid metabolism, commonly disregarded in the past, have been accepted as a hallmark for cancer. Exploring cancer therapeutics that interrupt the lipid metabolic pathways by monotherapy or combination with conventional chemotherapy or immunotherapy is of great importance. Here we modified cisplatin with an FDA-approved hypolipidemic drug, bezafibrate (BEZ), via the well-established Pt(IV) strategy, affording two multi-functional Pt(IV) anticancer agents cis,cis,trans-[Pt(NH3)2Cl2(BEZ)(OH)] (CB) and cis,cis,trans-[Pt(NH3)2Cl2(BEZ)2] (CP) (BEZ = bezafibrate). The Pt(IV) prodrug CB exhibited an enhanced anticancer activity up to 187-fold greater than the clinical anticancer drug cisplatin. Both CB and CP had less toxicity to normal cells, showing higher efficacies and superior therapeutic indexes than cisplatin. Mechanism studies revealed that the bezafibrate-conjugated Pt(IV) complex CB, as a representative, could massively accumulate in A549 cells and genomic DNA, induce DNA damage, elevate intracellular ROS levels, perturb mitochondrial transmembrane potentials, activate the cellular metabolic sensor AMPK, and result in profound proliferation inhibition and apoptosis. Further cellular data also provided evidence that phosphorylation of AMPK, as a metabolic sensor, could suppress the downstream HMGB1, NF-κB, and VEGFA, which may contribute to the inhibition of angiogenesis and metastasis. Our study suggests that the antitumor action of CB and CP mechanistically distinct from the conventional platinum drugs and that functionalizing platinum-based agents with lipid-modulating agents may represent a novel practical strategy for cancer treatment.
Collapse
Affiliation(s)
- Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yu-Yang Gao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Li-Xia Zheng
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xiao-Jing Ding
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Ling-Wen Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Juan-Juan Hu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Wei-Zhen Gao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China; Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
18
|
Tan Y, Wang M, Yang K, Chi T, Liao Z, Wei P. PPAR-α Modulators as Current and Potential Cancer Treatments. Front Oncol 2021; 11:599995. [PMID: 33833983 PMCID: PMC8021859 DOI: 10.3389/fonc.2021.599995] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of mortality worldwide. PPAR modulators may hold great potential for the management of cancer patients. Indeed, PPARs are critical sensors and regulators of lipid, and they are able to promote eNOS activation, regulate immunity and inflammation response, and affect proliferation and differentiation of cancer cells. Cancer, a name given to a group of diseases, is characterized by multiple distinctive biological behaviors, including angiogenesis, abnormal cell proliferation, aerobic glycolysis, inflammation, etc. In the last decade, emerging evidence has shown that PPAR-α, a nuclear hormone receptor, can modulate carcinogenesis via exerting effects on one or several characteristic pathological behaviors of cancer. Therefore, the multi-functional PPAR modulators have substantial promise in various types of cancer therapies. This review aims to consolidate the functions of PPAR-α, as well as discuss the current and potential applications of PPAR-α agonists and antagonists in tackling cancer.
Collapse
Affiliation(s)
- Yan Tan
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ke Yang
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tiange Chi
- The First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Stockholm, Sweden
- Zehuan Liao
| | - Peng Wei
- School of Traditional Chinese Medicine and School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Peng Wei
| |
Collapse
|
19
|
Hu Y, Wu MZ, Gu NJ, Xu HT, Li QC, Wu GP. Human papillomavirus 16 (HPV 16) E6 but not E7 inhibits the antitumor activity of LKB1 in lung cancer cells by downregulating the expression of KIF7. Thorac Cancer 2020; 11:3175-3180. [PMID: 32945133 PMCID: PMC7606012 DOI: 10.1111/1759-7714.13640] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The E6 and E7 proteins in human papillomavirus 16 (HPV 16) are the main oncogenes in the occurrence of lung cancer. In recent studies, we found that E6 and E7 downregulated the expression of LKB1 in lung cancer cells. However, it is still unclear how E6 and E7 regulate LKB1 in lung cancer cells. METHODS Double directional genetic manipulation and nuclear plasma separation technology were performed to explore the molecular mechanism of E6 and E7 inhibiting the antitumor activity of LKB1 in well-established lung cancer cell lines. RESULTS E6 but not E7 significantly downregulated the expression of tumor suppressor KIF7 at protein level, and the inhibition of KIF7 further reduced the expression of LKB1 both in the nuclei and in the cytoplasm, whereas reduced the expression of p-LKB1 in the cytoplasm only. This suggested that HPV 16 E6 but not E7 downregulates the antitumor activity of LKB1 by downregulating the expression of p-LKB1 in the cytoplasm only. CONCLUSIONS Here, we demonstrated for the first time that E6 but not E7 inhibits the antitumor activity of LKB1 in lung cancer cells by downregulating the expression of KIF7. Our findings provide new evidence to support the important role of KIF7 in the pathogenesis of lung cancer and suggests new therapeutic targets.
Collapse
Affiliation(s)
- Yue Hu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Ming-Zhe Wu
- Departments of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Na-Jin Gu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hong-Tao Xu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qing-Chang Li
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Guang-Ping Wu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
20
|
Shang LM, Liao XW, Zhu GZ, Huang KT, Han CY, Yang CK, Wang XK, Zhou X, Su H, Ye XP, Peng T. Genome-wide RNA-sequencing dataset reveals the prognostic value and potential molecular mechanisms of lncRNA in non-homologous end joining pathway 1 in early stage Pancreatic Ductal Adenocarcinoma. J Cancer 2020; 11:5556-5567. [PMID: 32913451 PMCID: PMC7477440 DOI: 10.7150/jca.39888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/07/2020] [Indexed: 12/13/2022] Open
Abstract
Objective: Our current study is to explore the prognostic value and molecular mechanisms underlying the role of lncRNA in non-homologous end joining pathway 1 (LINP1) in early stage pancreatic ductal adenocarcinoma (PDAC). Methods: Genome-wide RNA-seq datasets of 112 early stage PDAC patients were got from The Cancer Genome Atlas and analyzed using multiple online tools. Results: Overall survival in high LINP1 expression patients was shorter than those with low expression (high-LINP1 vs. low-LINP1=481 vs. 592 days, log-rank P=0.0432). The multivariate Cox proportional hazard regression model suggested that high-LINP1 patients had a markedly higher risk of death than low-LINP1 patients (adjusted P=0.004, hazard ratio=2.214, 95% confidence interval=1.283-3.820). Analysis of genome-wide co-expressed genes, screening of differentially expressed genes, and gene set enrichment analysis indicated that LINP1 may be involved in the regulation of cell proliferation-, cell adhesion- and cell cycle-related biological processes in PDAC. Six small-molecule compounds including STOCK1N-35874, fenofibrate, exisulind, NU-1025, vinburnine, and doxylamine were identified as potential LINP1-targeted drugs for the treatment of PDAC. Conclusions: Our study indicated that LINP1 may serve as a prognostic biomarker of early stage PDAC. Analysis of genome-wide datasets led to the elucidation of the underlying mechanisms and identified six potential targeted drugs for the treatment of early PDAC.
Collapse
Affiliation(s)
- Li-Ming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xi-Wen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Guang-Zhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Ke-Tuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Chuang-Ye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Cheng-Kun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiang-Kun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xin-Ping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
21
|
Li L, Zhang ZT. Hsa_circ_0086414 Might Be a Diagnostic Biomarker of Oral Squamous Cell Carcinoma. Med Sci Monit 2020; 26:e919383. [PMID: 31933490 PMCID: PMC6978993 DOI: 10.12659/msm.919383] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Circular RNAs (circRNAs), a newly-discovered class of non-coding RNAs, have a significant role in the progression of cancers, but the effect of hsa_circ_0086414 in human oral squamous cell carcinoma (OSCC) is still unclear. Material/Methods The circRNAs expression profile in OSCC tissue samples was assessed by high-throughput sequencing. The hsa_circ_0086414 expression level in 55 paired OSCC tissue samples and 2 kinds of OSCC cells was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the correlation between the hsa_circ_0086414 expression and clinicopathological characteristics of individuals with OSCC was studied. We used receiver operating characteristic (ROC) curves to observe the hsa_circ_0086414 value of diagnosis in OSCC. The network of hsa_circ_0086414-miRNAs-mRNAs was constructed. Gene Ontology (GO), Disease Oncology (DO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were carried out based on sequencing data and bioinformatics predictions. Results Hsa_circ_0086414 expression in OSCC tissue samples and OSCC cells was first discovered to be significantly downregulated compared with the adjacent healthy tissues (AHTs) and normal (HaCaT) cells, respectively. Moreover, its expression level was significantly correlated with stage in TNM, size of tumor, and lymph node metastasis. The area below the ROC curve was 0.749. Hsa_circ_0086414-miRNAs-mRNAs network analysis and GO, DO, and KEGG analyses all demonstrated that hsa_circ_0086414 is correlated with cancer progression to a certain extent. Conclusions We discovered that hsa_circ_0086414 might be an essential diagnostic biomarker in OSCC. Furthermore, hsa_circ_0086414 could be a target for OSCC therapy.
Collapse
Affiliation(s)
- Lin Li
- VIP Department, School of Stomatology, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Zhong-Ti Zhang
- VIP Department, School of Stomatology, China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
22
|
Chang NW, Huang YP. The RNA degradation pathway is involved in PPARα-modulated anti-oral tumorigenesis. Biomedicine (Taipei) 2019; 9:27. [PMID: 31724941 PMCID: PMC6855187 DOI: 10.1051/bmdcn/2019090427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
Background: The activation of peroxisome proliferator-activated receptor alpha (PPARα) has been shown to reprogram tumor metabolism and exhibits great potential for treating anti-oral tumorigenesis. Methods: In this study, we used a pathway-based strategy to explore possible functional pathways involved in the anticancer activity of PPARα in oral cancer cells through next-generation sequencing (NGS) and bioinformatic approaches. Results: We found that 3919 genes were upregulated and 1060 genes were downregulated through PPARα activation. These genes were mainly involved in the proteasomal, mRNA surveillance, spliceosomal, RNA transport, and RNA degradation pathways, as indicated by GO and KEGG enrichment analysis. Importantly, a total of 13 upregulated genes in the RNA degradation pathway were identified including 3 core exosome factor genes (RRP43, RRP42, and CSL4), 2 TRAMP complex genes (TRF4 and Mtr4), 2 exosome cofactor genes (RRP6 and MPP6), 2 CCR4-NOT complex genes (CNOT2 and CNOT3), 2 Ski complex genes (SKI2 and Ski3), 1 decapping complex gene (EDC4), and 1 gene involved in 5’ exoribonuclease activity (XRN1). Conclusion: Our findings suggest that the activation of PPARα to upregulate the RNA degradation pathway might provide a new strategy for oral cancer treatment.
Collapse
Affiliation(s)
- Nai-Wen Chang
- Department of Biochemistry, College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
23
|
A novel reporter construct for screening small molecule inhibitors that specifically target self-renewing cancer cells. Exp Cell Res 2019; 383:111551. [PMID: 31401066 DOI: 10.1016/j.yexcr.2019.111551] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a subset of cancer cells, which possess self-renewal ability, and lead to tumor progression, metastasis, and resistance to therapy. Live detection and isolation of CSCs are important to understand the biology of CSCs as well as to screen drugs that target them. Even though CSCs are detected using surface markers, there is a lot of inconsistencies for that in a given cancer type. At the same time, self-renewal markers like ALDH1A1, OCT4A and SOX2, which are intracellular molecules, are reliable markers for CSCs in different cancers. In the present study, we generated a reporter construct for self-renewing CSCs, based on ALDH1A1 expression. Oral cancer cells harboring ALDH1A1-DsRed2 were used to screen inhibitors that target CSCs. Our results showed that Comb1, a cocktail of inhibitors for EGF and TGF-β pathways and their intermediates, effectively reduced the DsRed2 population to 34%. Our immunohistochemical analysis on primary oral cancer corroborated the importance of EGF and TGF-β pathways in sustaining CSCs. Since these two pathways are also critical for the self-renewal and differentiation of normal stem cells, Comb1 might abolish them as well. On analysis of the effect of Comb1 on normal murine bone marrow cells, there was no significant change in the stem cell self-renewal and differentiation potential in the treated group compared to untreated cells. To conclude, we claim that ALDH1A1-DsRed2 is a useful tool to detect CSCs, and Comb1 is effective in targeting CSCs without affecting normal stem cells.
Collapse
|
24
|
Metabolic Regulation of Glycolysis and AMP Activated Protein Kinase Pathways during Black Raspberry-Mediated Oral Cancer Chemoprevention. Metabolites 2019; 9:metabo9070140. [PMID: 31336728 PMCID: PMC6680978 DOI: 10.3390/metabo9070140] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/15/2019] [Accepted: 07/08/2019] [Indexed: 12/26/2022] Open
Abstract
Oral cancer is a public health problem with an incidence of almost 50,000 and a mortality of 10,000 each year in the USA alone. Black raspberries (BRBs) have been shown to inhibit oral carcinogenesis in several preclinical models, but our understanding of how BRB phytochemicals affect the metabolic pathways during oral carcinogenesis remains incomplete. We used a well-established rat oral cancer model to determine potential metabolic pathways impacted by BRBs during oral carcinogenesis. F344 rats were exposed to the oral carcinogen 4-nitroquinoline-1-oxide in drinking water for 14 weeks, then regular drinking water for six weeks. Carcinogen exposed rats were fed a 5% or 10% BRB supplemented diet or control diet for six weeks after carcinogen exposure. RNA-Seq transcriptome analysis on rat tongue, and mass spectrometry and NMR metabolomics analysis on rat urine were performed. We tentatively identified 57 differentially or uniquely expressed metabolites and over 662 modulated genes in rats being fed with BRB. Glycolysis and AMPK pathways were modulated during BRB-mediated oral cancer chemoprevention. Glycolytic enzymes Aldoa, Hk2, Tpi1, Pgam2, Pfkl, and Pkm2 as well as the PKA-AMPK pathway genes Prkaa2, Pde4a, Pde10a, Ywhag, and Crebbp were downregulated by BRBs during oral cancer chemoprevention. Furthermore, the glycolysis metabolite glucose-6-phosphate decreased in BRB-administered rats. Our data reveal the novel metabolic pathways modulated by BRB phytochemicals that can be targeted during the chemoprevention of oral cancer.
Collapse
|
25
|
Small molecule inhibition of matrix metalloproteinases as a potential therapeutic for metastatic activity in squamous cell carcinoma. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s41548-019-00017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Fenofibrate induces human hepatoma Hep3B cells apoptosis and necroptosis through inhibition of thioesterase domain of fatty acid synthase. Sci Rep 2019; 9:3306. [PMID: 30824767 PMCID: PMC6397239 DOI: 10.1038/s41598-019-39778-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/29/2019] [Indexed: 12/31/2022] Open
Abstract
This study demonstrated that fenofibrate, a lipid-lowering drug, induced a significant time-dependent cytotoxicity of hepatoma Hep3B cells. Hep3B cells are significantly more sensitive to cell killing by fenofibrate than hepatoma HepG2, lung cancer CH27 and oral cancer HSC-3 cells. From the result of docking simulation, fenofibrate can bind excellently to the thioesterase domain of fatty acid synthase (FASN) binding site as orlistat, a FASN inhibitor, acts. The fenofibrate-induced cell cytotoxicity was protected by addition of palmitate, indicating that the cytotoxic effect of fenofibrate is due to starvation of Hep3B cells by inhibiting the formation of end product in the FASN reaction. Inhibition of lipid metabolism-related proteins expression, such as proteins containing thioesterase domain and fatty acid transport proteins, was involved in the fenofibrate-induced Hep3B cell death. Fenofibrate caused S and G2/M cell cycle arrest by inducing cyclin A/Cdk2 and reducing cyclin D1 and E protein levels in Hep3B cells. The anti-tumor roles of fenofibrate on Hep3B cells by inducing apoptosis and necroptosis were dependent on the expression of Bcl-2/caspase family members and RIP1/RIP3 proteins, respectively. These results suggest that fenofibrate has an anti-cancer effect in Hep3B cells and inhibition of lipid metabolism may be involved in fenofibrate-induced Hep3B cells apoptosis and necroptosis.
Collapse
|
27
|
Sun J, Zheng Z, Chen Q, Pan Y, Quan M, Dai Y. Fenofibrate potentiates chemosensitivity to human breast cancer cells by modulating apoptosis via AKT/NF-κB pathway. Onco Targets Ther 2019; 12:773-783. [PMID: 30774365 PMCID: PMC6353220 DOI: 10.2147/ott.s191239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Cumulatively, evidences revealed that fenofibrate used in the therapy of hyperlipidemia and hypercholesterolemia has anti-cancer effect in multiple cancer types. However, its function and underlying mechanism of chemosensitization in breast cancer remain poorly understood. Materials and methods The cytotoxicity of fenofibrate and anti-cancer drugs in breast cancer cells was determined by MTT. Apoptosis and mitochondrial membrane potential were measured using flow cytometry. Caspases and PARP cleavage, the Bcl-2 family members’ protein expression, as well as the activation of AKT and NF-κB signaling pathways were evaluated using Western blot assay. Real-time PCR was used to determine the mRNA expression of Bcl-2 family members. Results Our data indicated that fenofibrate suppressed SKBR3 and MDA-MB-231 cell growth in a dose-dependent manner, in the same way as paclitaxel, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), ABT-737, and doxorubicin. Subtoxic levels of fenofibrate significantly augmented paclitaxel, TRAIL, ABT-737, and doxorubicin-induced apoptosis in both these two cell lines. Fenofibrate-promoted chemosensitivity is predominantly mediated by caspase-9 and caspase-3 activation and mitochondrial outer membrane permeabilization. Meanwhile, chemosensitivity promoted by fenofibrate also increased the expression of Bax and Bok and decreased the expression of Mcl-1 and Bcl-xl. Mechanistically, fenofibrate effectively reduced the phosphorylation levels of AKT and NF-κB. In addition, imiquimod, an NF-κB activator, could reverse fenofibrate-induced susceptibility to ABT-737-triggered apoptosis. Conclusion The present study provided the evidence of the underlying mechanisms on chemosensitization of fenofibrate by inducing the apoptosis of breast cancer in an AKT/NF-κB-dependent manner and implicated the potential application of fenofibrate in potentiating chemosensitivity in breast cancer therapy.
Collapse
Affiliation(s)
- Jianguo Sun
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China, .,Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China
| | - Zhibao Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China,
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China
| | - Yin Pan
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China,
| | - Mingming Quan
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China,
| | - Yuechu Dai
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, People's Republic of China,
| |
Collapse
|
28
|
Wang YF, Li BW, Sun S, Li X, Su W, Wang ZH, Wang F, Zhang W, Yang HY. Circular RNA Expression in Oral Squamous Cell Carcinoma. Front Oncol 2018; 8:398. [PMID: 30349810 PMCID: PMC6187971 DOI: 10.3389/fonc.2018.00398] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/03/2018] [Indexed: 01/22/2023] Open
Abstract
Circular RNA (circRNA) is a type of non-coding RNA molecule that affects the cellular regulatory network by sequestering microRNA (miRNA) like a sponge. This study was performed to identify differentially-expressed circRNA in oral squamous cell carcinomas (OSCCs). By high-throughput sequencing, microarray circRNA expression profiles were acquired from patients with OSCCs (n = 8) and controls (n = 8), which totaled 1921 existing circRNA molecules and 10021 novel circRNA molecules. Most of the circular RNA is from exons and distributed in the No. 1 and 2 chromosomes. Eight up-regulated and down-regulated circRNA molecules were identified as differentially-expressed in OSCCs. Among this, the expression of circ_000334, circ_006740, and circ_006371 are significantly down-regulated in 42 pairs of samples, which means that these circRNA molecules might be implicated in oncogenesis and development of OSCCs.
Collapse
Affiliation(s)
- Yu-Fan Wang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bo-Wen Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Clinical School, Peking University Shenzhen Hospital, Anhui Medical University, Hefei, China
| | - Shuai Sun
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiang Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wen Su
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhi-Hong Wang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Feng Wang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wei Zhang
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hong-Yu Yang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Clinical School, Peking University Shenzhen Hospital, Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
Wu W, Wang S, Liu Q, Shan T, Wang Y. Metformin Protects against LPS-Induced Intestinal Barrier Dysfunction by Activating AMPK Pathway. Mol Pharm 2018; 15:3272-3284. [DOI: 10.1021/acs.molpharmaceut.8b00332] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Weiche Wu
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Sisi Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Qing Liu
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Tizhong Shan
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yizhen Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| |
Collapse
|
30
|
Vallard A, Rancoule C, Espenel S, Garcia MA, Langrand-Escure J, He MY, Ben Mrad M, El Meddeb Hamrouni A, Ouni S, Trone JC, Rehailia-Blanchard A, Guillaume E, Vial N, Riocreux C, Guy JB, Magné N. Harnessing drug/radiation interaction through daily routine practice: Leverage medical and methodological point of view (MORSE 02-17 study). Radiother Oncol 2018; 129:471-478. [PMID: 29937210 DOI: 10.1016/j.radonc.2018.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 06/02/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Safety profile of the interaction between anticancer drugs and radiation is a recurrent question. However, there are little data regarding the non-anticancer treatment (NACT)/radiation combinations. The aim of the present study was to investigate concomitant NACTs in patients undergoing radiotherapy in a French comprehensive cancer center. METHODS A prospective cross-sectional study was conducted. All cancer patients undergoing a palliative or curative radiotherapy were consecutively screened for six weeks in 2016. Data on NACTs were collected. RESULTS Out of 214 included patients, a NACT was concomitantly prescribed to 155 patients (72%), with a median number of 5 NACTs per patient (range: 1-12). The most prescribed drugs were anti-hypertensive drugs (101 patients, 47.2%), psychotropic drugs (n = 74, 34.6%), analgesics (n = 78, 36.4%), hypolipidemic drugs (n = 57, 26.6%), proton pump inhibitors (n = 46, 21.5%) and antiplatelet drugs (n = 38, 17.8%). Although 833 different molecules were reported, only 20 possible modifiers of cancer biological pathways (prescribed to 74 patients (34.5%)) were identified. Eight out of the 833 molecules (0.9%), belonging to six drug families, have been investigated in 28 ongoing or published clinical trials in combo with radiotherapy. They were prescribed to 63 patients (29.4%). CONCLUSION Drug-radiation interaction remains a subject of major interest, not only for conventional anticancer drugs, but also for NACTs. New trial designs are thus required.
Collapse
Affiliation(s)
- A Vallard
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France; Cellular and Molecular Radiobiology Laboratory, CNRS UMR 5822, IPNL, 69622 Villeurbanne, France
| | - C Rancoule
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France; Cellular and Molecular Radiobiology Laboratory, CNRS UMR 5822, IPNL, 69622 Villeurbanne, France
| | - S Espenel
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France; Cellular and Molecular Radiobiology Laboratory, CNRS UMR 5822, IPNL, 69622 Villeurbanne, France
| | - M-A Garcia
- General Health Department, Hygée Institute, Avenue Albert Raimond, BP 60008, 42271 Saint-Priest en Jarez, France
| | - J Langrand-Escure
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - M Y He
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - M Ben Mrad
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - A El Meddeb Hamrouni
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - S Ouni
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - J-C Trone
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - A Rehailia-Blanchard
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - E Guillaume
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - N Vial
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - C Riocreux
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France
| | - J-B Guy
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France; Cellular and Molecular Radiobiology Laboratory, CNRS UMR 5822, IPNL, 69622 Villeurbanne, France
| | - N Magné
- Radiotherapy Department, Lucien Neuwirth Cancer Institute, 42270 St Priest en Jarez, France; Cellular and Molecular Radiobiology Laboratory, CNRS UMR 5822, IPNL, 69622 Villeurbanne, France.
| |
Collapse
|
31
|
Lian X, Wang G, Zhou H, Zheng Z, Fu Y, Cai L. Anticancer Properties of Fenofibrate: A Repurposing Use. J Cancer 2018; 9:1527-1537. [PMID: 29760790 PMCID: PMC5950581 DOI: 10.7150/jca.24488] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/25/2018] [Indexed: 12/22/2022] Open
Abstract
Cancer is a leading cause of death throughout the world, and cancer therapy remains a big medical challenge in terms of both its therapeutic efficacy and safety. Therefore, to find out a safe anticancer drug has been long goal for oncologist and medical scientists. Among clinically used medicines with no or little toxicity, fenofibrate is a drug of the fibrate class that plays an important role in lowering the levels of serum cholesterol and triglycerides while elevating the levels of high-density lipoproteins. Recently, several studies have implied that fenofibrate may exert anticancer effects via a variety of pathways involved in apoptosis, cell-cycle arrest, invasion, and migration. Given the great potential that fenofibrate may have anticancer effects, this review was to investigate all published works which directly or indirectly support the anticancer activity of fenofibrate. These studies provide evidence that fenofibrate exerted antitumor effects in several human cancer cell lines, such as breast, liver, glioma, prostate, pancreas, and lung cancer cell lines. Among these studies some have further confirmed the possibility and efficacy of fenofibrate anticancer in xenograft mouse models. In the last part of this review, we also discuss the potential mechanisms of action of fenofibrate based on the available information. Overall, we may repurpose fenofibrate as an anticancer drug in cancer treatment, which urgently need further and comprehensively investigated.
Collapse
Affiliation(s)
- Xin Lian
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Gang Wang
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China
| | - Zongyu Zheng
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Yaowen Fu
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China
| | - Lu Cai
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.,Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
32
|
Chiu YJ, Hour MJ, Jin YA, Lu CC, Tsai FJ, Chen TL, Ma H, Juan YN, Yang JS. Disruption of IGF‑1R signaling by a novel quinazoline derivative, HMJ‑30, inhibits invasiveness and reverses epithelial-mesenchymal transition in osteosarcoma U‑2 OS cells. Int J Oncol 2018; 52:1465-1478. [PMID: 29568964 PMCID: PMC5873869 DOI: 10.3892/ijo.2018.4325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most common primary malignancy of the bone and is characterized by local invasion and distant metastasis. Over the past 20 years, long-term outcomes have reached a plateau even with aggressive therapy. Overexpression of insulin-like growth factor 1 receptor (IGF‑1R) is associated with tumor proliferation, invasion and migration in osteosarcoma. In the present study, our group developed a novel quinazoline derivative, 6-fluoro‑2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline (HMJ‑30), in order to disrupt IGF‑1R signaling and tumor invasiveness in osteosarcoma U‑2 OS cells. Molecular modeling, immune-precipitation, western blotting and phosphorylated protein kinase sandwich ELISA assays were used to confirm this hypothesis. The results demonstrated that HMJ‑30 selectively targeted the ATP-binding site of IGF‑1R and inhibited its downstream phosphoinositide 3-kinase/protein kinase B, Ras/mitogen-activated protein kinase, and IκK/nuclear factor-κB signaling pathways in U‑2 OS cells. HMJ‑30 inhibited U‑2 OS cell invasion and migration and downregulated protein levels and activities of matrix metalloproteinase (MMP)‑2 and MMP-9. An increase in protein levels of tissue inhibitor of metalloproteinase (TIMP)‑1 and TIMP‑2 was also observed. Furthermore, HMJ‑30 caused U‑2 OS cells to aggregate and form tight clusters, and these cells were flattened, less elongated and displayed cobblestone-like shapes. There was an increase in epithelial markers and a decrease in mesenchymal markers, indicating that the cells underwent the reverse epithelial-mesenchymal transition (EMT) process. Overall, these results demonstrated the potential molecular mechanisms underlying the effects of HMJ‑30 on invasiveness and EMT in U‑2 OS cells, suggesting that this compound deserves further investigation as a potential anti-osteosarcoma drug.
Collapse
Affiliation(s)
- Yu-Jen Chiu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veteran General Hospital, Taipei 112, Taiwan, R.O.C
| | - Mann-Jen Hour
- School of Pharmacy, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yi-An Jin
- Department of Dermatology, Taipei Medical University Hospital, Taipei 110, Taiwan, R.O.C
| | - Chi-Cheng Lu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan, R.O.C
| | - Fuu-Jen Tsai
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Tai-Lin Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 112, Taiwan, R.O.C
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veteran General Hospital, Taipei 112, Taiwan, R.O.C
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan, R.O.C
| |
Collapse
|
33
|
Huang YP, Chang NW. Proteomic analysis of oral cancer reveals new potential therapeutic targets involved in the Warburg effect. Clin Exp Pharmacol Physiol 2018; 44:880-887. [PMID: 28453233 DOI: 10.1111/1440-1681.12774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/28/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Activation of peroxisome proliferator-activated receptor alpha (PPARα) has been reported to disrupt tumour metabolism and to promote anticancer activity through interfering with the Warburg effect. This study is to investigate whether Warburg effect-related proteins also could be identified in oral tumour lesions and to explore the functional significance of PPARα in metabolic shift. Five pairs of tongue tumour tissues and adjacent reference tissues obtained from 4-NQO/arecoline induced mouse model were analyzed by 2-d-gel-electrophoresis and LC-MS. Further, the hexokinase II level, pyruvate dehydrogenase (PDH) activity, and metabolites of glycolysis and TCA cycle were all examined in order to validate the effect of PPARα on metabolic shift. Changes in protein expression levels revealed that seven proteins, which were involved in glycolysis, the tricarboxylic acid cycle, and the respiratory chain, were down-regulated in tumour tissues. We found that activation of PPARα through fenofibrate could inhibit oral cancer cell growth and switch the way of energy production from the Warburg effect to oxidative phosphorylation. Fenofibrate induced a reduction of hexokinase II protein levels, increases in PDH activity and metabolites of the TCA cycle, and an impairment of ATP production. These findings suggested that activation of the PPARα to reprogram the metabolic pathway might impair the Warburg effect and trigger cancer cell death. The study provides a novel view of changes in protein expression profiles involved in the Warburg effect during oral tumourigenesis. Activation of the PPARα to impair the Warburg effect might offer a new strategy for oral cancer treatment.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Nai-Wen Chang
- Department of Biochemistry, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
34
|
Nguyen CH, Huttary N, Atanasov AG, Chatuphonprasert W, Brenner S, Fristiohady A, Hong J, Stadler S, Holzner S, Milovanovic D, Dirsch VM, Kopp B, Saiko P, Krenn L, Jäger W, Krupitza G. Fenofibrate inhibits tumour intravasation by several independent mechanisms in a 3-dimensional co-culture model. Int J Oncol 2017; 50:1879-1888. [PMID: 28393180 DOI: 10.3892/ijo.2017.3956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 01/16/2017] [Indexed: 11/06/2022] Open
Abstract
Lymph node metastasis of breast cancer is a clinical marker of poor prognosis. Yet, there exist no therapies targeting mechanisms of intravasation into lymphatics. Herein we report on an effect of the antidyslipidemic drug fenofibrate with vasoprotective activity, which attenuates breast cancer intravasation in vitro, and describe the potential mechanisms. To measure intravasation in a 3-dimensional co-culture model MDA-MB231 and MCF-7 breast cancer spheroids were placed on immortalised lymphendothelial cell (LEC) monolayers. This provokes the formation of circular chemorepellent induced defects (CCIDs) in the LEC barrier resembling entry ports for the intravasating tumour. Furthermore, the expression of adhesion molecules ICAM-1, CD31 and FAK was investigated in LECs by western blotting as well as cell-cell adhesion and NF-κB activity by respective assays. In MDA-MB231 cells the activity of CYP1A1 was measured by EROD assay. Fenofibrate inhibited CCID formation in the MDA-MB231/LEC- and MCF-7/LEC models and the activity of NF-κB, which in turn downregulated ICAM-1 in LECs and the adhesion of cancer cells to LECs. Furthermore, CD31 and the activity of FAK were inhibited. In MDA-MB231 cells, fenofibrate attenuated CYP1A1 activity. Combinations with other FDA-approved drugs, which reportedly inhibit different ion channels, attenuated CCID formation additively or synergistically. In summary, fenofibrate inhibited NF-κB and ICAM-1, and inactivated FAK, thereby attenuating tumour intravasation in vitro. A combination with other FDA-approved drugs further improved this effect. Our new concept may lead to a novel therapy for cancer patients.
Collapse
Affiliation(s)
- Chi Huu Nguyen
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Nicole Huttary
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | | | | | - Stefan Brenner
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Adryan Fristiohady
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Junli Hong
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Serena Stadler
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Silvio Holzner
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Daniela Milovanovic
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Brigitte Kopp
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Philipp Saiko
- Department of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Liselotte Krenn
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Georg Krupitza
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Zhang L, Han J, Jackson AL, Clark LN, Kilgore J, Guo H, Livingston N, Batchelor K, Yin Y, Gilliam TP, Gehrig PA, Sheng X, Zhou C, Bae-Jump VL. NT1014, a novel biguanide, inhibits ovarian cancer growth in vitro and in vivo. J Hematol Oncol 2016; 9:91. [PMID: 27655410 PMCID: PMC5031332 DOI: 10.1186/s13045-016-0325-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/15/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND NT1014 is a novel biguanide and AMPK activator with a high affinity for the organic cation-specific transporters, OCT1 and OCT3. We sought to determine the anti-tumorigenic effects of NT1014 in human ovarian cancer cell lines as well as in a genetically engineered mouse model of high-grade serous ovarian cancer. METHODS The effects of NT1014 and metformin on cell proliferation were assessed by MTT assay using the human ovarian cancer cell lines, SKOV3 and IGROV1, as well as in primary cultures. In addition, the impact of NT1014 on cell cycle progression, apoptosis, cellular stress, adhesion, invasion, glycolysis, and AMPK activation/mTOR pathway inhibition was also explored. The effects of NT1014 treatment in vivo was evaluated using the K18 - gT121+/-; p53fl/fl; Brca1fl/fl (KpB) mouse model of high-grade serous ovarian cancer. RESULTS NT1014 significantly inhibited cell proliferation in both ovarian cancer cell lines as well as in primary cultures. In addition, NT1014 activated AMPK, inhibited downstream targets of the mTOR pathway, induced G1 cell cycle arrest/apoptosis/cellular stress, altered glycolysis, and reduced invasion/adhesion. Similar to its anti-tumorigenic effects in vitro, NT1014 decreased ovarian cancer growth in the KpB mouse model of ovarian cancer. NT1014 appeared to be more potent than metformin in both our in vitro and in vivo studies. CONCLUSIONS NT1014 inhibited ovarian cancer cell growth in vitro and in vivo, with greater efficacy than the traditional biguanide, metformin. These results support further development of NT1014 as a useful therapeutic approach for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Jinan, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Jianjun Han
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Surgical Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Amanda L Jackson
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Leslie N Clark
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua Kilgore
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Hui Guo
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Jinan, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Nick Livingston
- NovaTarg Therapeutics, Research Triangle Park, Durham, NC, 27709, USA
| | - Kenneth Batchelor
- NovaTarg Therapeutics, Research Triangle Park, Durham, NC, 27709, USA
| | - Yajie Yin
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Jinan, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Timothy P Gilliam
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Paola A Gehrig
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiugui Sheng
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Jinan, People's Republic of China
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
36
|
Jan CI, Tsai MH, Chiu CF, Huang YP, Liu CJ, Chang NW. Fenofibrate Suppresses Oral Tumorigenesis via Reprogramming Metabolic Processes: Potential Drug Repurposing for Oral Cancer. Int J Biol Sci 2016; 12:786-98. [PMID: 27313493 PMCID: PMC4910598 DOI: 10.7150/ijbs.13851] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 04/01/2016] [Indexed: 02/07/2023] Open
Abstract
One anticancer strategy suggests targeting mitochondrial metabolism to trigger cell death through slowing down energy production from the Warburg effect. Fenofibrate is a clinical lipid-lowering agent and an effective anticancer drug. In the present study, we demonstrate that fenofibrate provided novel mechanisms for delaying oral tumor development via the reprogramming of metabolic processes. Fenofibrate induced cytotoxicity by decreasing oxygen consumption rate (OCR) that was accompanied with increasing extracellular acidification rate (ECAR) and reducing ATP content. Moreover, fenofibrate caused changes in the protein expressions of hexokinase II (HK II), pyruvate kinase, pyruvate dehydrogenase, and voltage-dependent anion channel (VDAC), which are associated with the Warburg effect. In addition, fenofibrate reprogrammed the metabolic pathway by interrupting the binding of HK II to VDAC. In an oral cancer mouse model, fenofibrate exhibited both preventive and therapeutic efficacy on oral tumorigenesis. Fenofibrate administration suppressed the incidence rate of tongue lesions, reduced the tumor sizes, decreased the tumor multiplicity, and decreased the immunoreactivities of VDAC and mTOR. The molecular mechanisms involved in fenofibrate's ability to delay tumor development included the down-regulation of mTOR activity via TSC1/2-dependent signaling through activation of AMPK and inactivation of Akt, or via a TSC1/2-independent pathway through direct suppression of raptor. Our findings provide a molecular rationale whereby fenofibrate exerts anticancer and additional beneficial effects for the treatment of oral cancer patients.
Collapse
Affiliation(s)
- Chia-Ing Jan
- 1. Department of Pathology, China Medical University and Hospital, Taichung, Taiwan, ROC; 2. Department of Pathology, China Medical University and Beigang Hospital, Yunlin, Taiwan.TOC
| | - Ming-Hsui Tsai
- 3. Department of Otolaryngology, China Medical University and Hospital, Taichung, Taiwan, ROC
| | - Chang-Fang Chiu
- 4. Department of Hematology Oncology, China Medical University and Hospital, Taichung, Taiwan, ROC
| | - Yi-Ping Huang
- 5. Department of Physiology, College of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Chia Jen Liu
- 6. Department of Biochemistry, College of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Nai Wen Chang
- 6. Department of Biochemistry, College of Medicine, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
37
|
Li S, Han D, Zhang Y, Xie X, Ke R, Zhu Y, Liu L, Song Y, Yang L, Li M. Activation of AMPK Prevents Monocrotaline-Induced Extracellular Matrix Remodeling of Pulmonary Artery. Med Sci Monit Basic Res 2016; 22:27-33. [PMID: 26978596 PMCID: PMC4795089 DOI: 10.12659/msmbr.897505] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background The current study was performed to investigate the effect of adenosine monophosphate (AMP) – activated protein kinase (AMPK) activation on the extracellular matrix (ECM) remodeling of pulmonary arteries in pulmonary arterial hypertension (PAH) and to address its potential mechanisms. Material/Methods PAH was induced by a single intraperitoneal injection of monocrotaline (MCT) into Sprague-Dawley rats. Metformin (MET) was administered to activate AMPK. Immunoblotting was used to determine the phosphorylation and expression of AMPK and expression of tissue inhibitor of metalloproteinase-1 (TIMP-1). Gelatin zymography was performed to determine the activity of matrix metalloproteinase-2 (MMP-2) and MMP-9. Results Activation of AMPK by MET significantly reduced the right ventricle systolic pressure and the right ventricular hypertrophy in MCT-induced rat PAH model, and partially inhibited the ECM remodeling of pulmonary arteries. These effects were coupled with the decrease of MMP-2/9 activity and TIMP-1 expression. Conclusions This study suggests that activation of AMPK benefits PAH by inhibiting ECM remodeling of pulmonary arteries. Enhancing AMPK activity might have potential value in clinical treatment of PAH.
Collapse
Affiliation(s)
- Shaojun Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Dong Han
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Yonghong Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Xinming Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Rui Ke
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Yanting Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Lu Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Yang Song
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Lan Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Manxiang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
38
|
Huang YP, Chang NW. PPARα modulates gene expression profiles of mitochondrial energy metabolism in oral tumorigenesis. Biomedicine (Taipei) 2016; 6:3. [PMID: 26869356 PMCID: PMC4751096 DOI: 10.7603/s40681-016-0003-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/15/2015] [Indexed: 11/16/2022] Open
Abstract
Metabolic reprogramming plays a crucial role in the development of cancer. The aim of this study was to explore the effect of fenofibrate, an agonist of peroxisome proliferator-activated receptor alpha (PPARα), on gene expression profiles of mitochondrial energy metabolism. Our results showed that PPARα expression was negatively correlated with tumor progression in an oral cancer mouse model. Activation of PPARα through fenofibrate suppressed migration of oral cancer cells. Differential protein profiling demonstrated that expressions of genes related to mitochondrial energy metabolism were either up-regulated (Atp5g3, Cyc1, Ndufa5, Ndufa10, and Sdhd) or down-regulated (Cox5b, Ndufa1, Ndufb7, and Uqcrh) through PPARα activation and response. Our results indicate that PPARα exhibits a great potential for anti-oral cancer therapies by modulating cancer cell mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, 404, Taichung, Taiwan
| | - Nai Wen Chang
- Department of Biochemistry, College of Medicine,, China Medical University, 404, No. 91, Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
39
|
Abstract
Objective: To review the mechanisms of anti-cancer activity of fenofibrate (FF) and other Peroxisome Proliferator Activator Receptor α (PPARα) agonists based on evidences reported in the published literature.Methods: We extensively reviewed the literature concerning FF as an off target anti-cancer drug. Controversies regarding conflicting findings were also addressed.Results: The main mechanism involved in anti-cancer activity is anti-angiogenesis through down-regulation of Vascular Endothelial Growth Factor (VEGF), Vascular Endothelial Growth Factor Receptor (VEGFR) and Hypoxia Inducible factor-1 α (HIF-1α), inhibition of endothelial cell migration, up-regulation of endostatin and thrombospondin-1, but there are many other contributing mechanisms like apoptosis and cell cycle arrest, down-regulation of Nuclear Factor Kappa B (NF-kB) and Protein kinase B (Akt) and decrease of cellular energy by impairing mitochondrial function. Growth impairment is related to down-regulation of Phospho-Inositol 3 Kinase (PI3K)/Akt axis and down-regulation of the p38 map kinase (MAPK) cascade. A possible role should be assigned to FF stimulated over-expression of Tribbles Homolog-3 (TRIB3) which inhibits Akt phosphorylation. Important anti-cancer and anti-metastatic activities are due to down-regulation of MCP-1 (monocyte chemotactic protein-1), decreased Metalloprotease-9 (MMP-9) production, weak down-regulation of adhesion molecules like E selectin, intercellular adhesion molecules (ICAM) and Vascular Endothelial Adhesion Molecules (VCAM), and decreased secretion of chemokines like Interleukin-6 (IL-6), and down-regulation of cyclin D-1. There is no direct link between FF activity in lipid metabolism and anticancer activity, except for the fact that many anticancer actions are dependent from PPARα agonism. FF exhibits also PPARα independent anti-cancer activities.Conclusions: There are strong evidences indicating that FF can disrupt growth-related activities in many different cancers, due to anti-angiogenesis and anti-inflammatory effects. Therefore FF may be useful as a complementary adjunct treatment of cancer, particularly included in anti-angiogenic protocols like those currently increasingly used in glioblastoma. There are sound reasons to initiate well planned phase II clinical trials for FF as a complementary adjunct treatment of cancer.
Collapse
|