1
|
Jung O, Baek MJ, Wooldrik C, Johnson KR, Fisher KW, Lou J, Ricks TJ, Wen T, Best MD, Cryns VL, Anderson RA, Choi S. Nuclear phosphoinositide signaling promotes YAP/TAZ-TEAD transcriptional activity in breast cancer. EMBO J 2024; 43:1740-1769. [PMID: 38565949 PMCID: PMC11066040 DOI: 10.1038/s44318-024-00085-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
The Hippo pathway effectors Yes-associated protein 1 (YAP) and its homolog TAZ are transcriptional coactivators that control gene expression by binding to TEA domain (TEAD) family transcription factors. The YAP/TAZ-TEAD complex is a key regulator of cancer-specific transcriptional programs, which promote tumor progression in diverse types of cancer, including breast cancer. Despite intensive efforts, the YAP/TAZ-TEAD complex in cancer has remained largely undruggable due to an incomplete mechanistic understanding. Here, we report that nuclear phosphoinositides function as cofactors that mediate the binding of YAP/TAZ to TEADs. The enzymatic products of phosphoinositide kinases PIPKIα and IPMK, including phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and phosphatidylinositol 3,4,5-trisphosphate (P(I3,4,5)P3), bridge the binding of YAP/TAZ to TEAD. Inhibiting these kinases or the association of YAP/TAZ with PI(4,5)P2 and PI(3,4,5)P3 attenuates YAP/TAZ interaction with the TEADs, the expression of YAP/TAZ target genes, and breast cancer cell motility. Although we could not conclusively exclude the possibility that other enzymatic products of IPMK such as inositol phosphates play a role in the mechanism, our results point to a previously unrecognized role of nuclear phosphoinositide signaling in control of YAP/TAZ activity and implicate this pathway as a potential therapeutic target in YAP/TAZ-driven breast cancer.
Collapse
Affiliation(s)
- Oisun Jung
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Min-Jeong Baek
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Interdisciplinary Graduate Program in Biomedical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Colin Wooldrik
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Interdisciplinary Graduate Program in Biomedical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Keith R Johnson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Oral Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kurt W Fisher
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Tanei J Ricks
- Department of Chemistry, University of Memphis, 3744 Walker Avenue, Memphis, TN, 38152, USA
| | - Tianmu Wen
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Vincent L Cryns
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Suyong Choi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
2
|
Cao H, Wang Y, Zhang D, Liu B, Zhou H, Wang S. Periprostatic Adipose Tissue: A New Perspective for Diagnosing and Treating Prostate Cancer. J Cancer 2024; 15:204-217. [PMID: 38164282 PMCID: PMC10751678 DOI: 10.7150/jca.89750] [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: 09/01/2023] [Accepted: 10/26/2023] [Indexed: 01/03/2024] Open
Abstract
Prostate cancer (PCa) is the most common tumor of the male genitourinary system. It will eventually progress to fatal metastatic castration-resistant prostate cancer, for which treatment options are limited. Adipose tissues are distributed in various parts of the body. They have different morphological structures and functional characteristics and are associated with the development of various tumors. Periprostatic adipose tissue (PPAT) is the closest white visceral adipose tissue to the prostate and is part of the PCa tumor microenvironment. Studies have shown that PPAT is involved in PCa development, progression, invasion, and metastasis through the secretion of multiple active molecules. Factors such as obesity, diet, exercise, and organochlorine pesticides can affect the development of PCa indirectly or directly through PPAT. Based on the mechanism of PPAT's involvement in regulating PCa, this review summarized various diagnostic and therapeutic approaches for PCa with potential applications to assess the progression of patients' disease and improve clinical outcomes.
Collapse
Affiliation(s)
- Hongliang Cao
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, China
| | - Song Wang
- Department of Urology II, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
3
|
Shuvalov O, Kirdeeva Y, Daks A, Fedorova O, Parfenyev S, Simon HU, Barlev NA. Phytochemicals Target Multiple Metabolic Pathways in Cancer. Antioxidants (Basel) 2023; 12:2012. [PMID: 38001865 PMCID: PMC10669507 DOI: 10.3390/antiox12112012] [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: 09/12/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer metabolic reprogramming is a complex process that provides malignant cells with selective advantages to grow and propagate in the hostile environment created by the immune surveillance of the human organism. This process underpins cancer proliferation, invasion, antioxidant defense, and resistance to anticancer immunity and therapeutics. Perhaps not surprisingly, metabolic rewiring is considered to be one of the "Hallmarks of cancer". Notably, this process often comprises various complementary and overlapping pathways. Today, it is well known that highly selective inhibition of only one of the pathways in a tumor cell often leads to a limited response and, subsequently, to the emergence of resistance. Therefore, to increase the overall effectiveness of antitumor drugs, it is advisable to use multitarget agents that can simultaneously suppress several key processes in the tumor cell. This review is focused on a group of plant-derived natural compounds that simultaneously target different pathways of cancer-associated metabolism, including aerobic glycolysis, respiration, glutaminolysis, one-carbon metabolism, de novo lipogenesis, and β-oxidation of fatty acids. We discuss only those compounds that display inhibitory activity against several metabolic pathways as well as a number of important signaling pathways in cancer. Information about their pharmacokinetics in animals and humans is also presented. Taken together, a number of known plant-derived compounds may target multiple metabolic and signaling pathways in various malignancies, something that bears great potential for the further improvement of antineoplastic therapy.
Collapse
Affiliation(s)
- Oleg Shuvalov
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Yulia Kirdeeva
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Alexandra Daks
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Olga Fedorova
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Sergey Parfenyev
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland;
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Nickolai A. Barlev
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 20000, Kazakhstan
| |
Collapse
|
4
|
Deng B, Kong W, Suo H, Shen X, Newton MA, Burkett WC, Zhao Z, John C, Sun W, Zhang X, Fan Y, Hao T, Zhou C, Bae-Jump VL. Oleic Acid Exhibits Anti-Proliferative and Anti-Invasive Activities via the PTEN/AKT/mTOR Pathway in Endometrial Cancer. Cancers (Basel) 2023; 15:5407. [PMID: 38001668 PMCID: PMC10670880 DOI: 10.3390/cancers15225407] [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: 10/09/2023] [Revised: 11/08/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Reprogramming of fatty acid metabolism promotes cell growth and metastasis through a variety of processes that stimulate signaling molecules, energy storage, and membrane biosynthesis in endometrial cancer. Oleic acid is one of the most important monounsaturated fatty acids in the human body, which appears to have both pro- and anti-tumorigenic activities in various pre-clinical models. In this study, we evaluated the potential anti-tumor effects of oleic acid in endometrial cancer cells and the LKB1fl/flp53fl/fl mouse model of endometrial cancer. Oleic acid increased lipogenesis, inhibited cell proliferation, caused cell cycle G1 arrest, induced cellular stress and apoptosis, and suppressed invasion in endometrial cancer cells. Targeting of diacylglycerol acyltransferases 1 and 2 effectively increased the cytotoxicity of oleic acid. Moreover, oleic acid significantly increased the expression of wild-type PTEN, and knockdown of PTEN by shRNA partially reversed the anti-proliferative and anti-invasive effects of oleic acid. Inhibition of the AKT/mTOR pathway by ipatasertib effectively increased the anti-tumor activity of oleic acid in endometrial cancer cells. Oleic acid treatment (10 mg/kg, daily, oral) for four weeks significantly inhibited tumor growth by 52.1% in the LKB1fl/flp53fl/fl mice. Our findings demonstrated that oleic acid exhibited anti-tumorigenic activities, dependent on the PTEN/AKT/mTOR signaling pathway, in endometrial cancer.
Collapse
Affiliation(s)
- Boer Deng
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China; (B.D.); (H.S.); (X.S.); (Z.Z.); (X.Z.); (Y.F.)
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Weimin Kong
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Hongyan Suo
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China; (B.D.); (H.S.); (X.S.); (Z.Z.); (X.Z.); (Y.F.)
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Xiaochang Shen
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China; (B.D.); (H.S.); (X.S.); (Z.Z.); (X.Z.); (Y.F.)
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Meredith A. Newton
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Wesley C. Burkett
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Ziyi Zhao
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China; (B.D.); (H.S.); (X.S.); (Z.Z.); (X.Z.); (Y.F.)
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Catherine John
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Xin Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China; (B.D.); (H.S.); (X.S.); (Z.Z.); (X.Z.); (Y.F.)
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Yali Fan
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China; (B.D.); (H.S.); (X.S.); (Z.Z.); (X.Z.); (Y.F.)
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Tianran Hao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
- 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 at Chapel Hill, Chapel Hill, NC 27599, USA; (W.K.); (M.A.N.); (W.C.B.); (C.J.); (W.S.); (T.H.)
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
5
|
Kado T, Kusakari N, Tamaki T, Murota K, Tsujiuchi T, Fukushima N. Oleic acid stimulates cell proliferation and BRD4-L-MYC-dependent glucose transporter transcription through PPARα activation in ovarian cancer cells. Biochem Biophys Res Commun 2023; 657:24-34. [PMID: 36965420 DOI: 10.1016/j.bbrc.2023.03.051] [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: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023]
Abstract
Fatty acids (FAs) play important roles in cell membrane structure maintenance, energy production via β-oxidation, and as extracellular signaling molecules. Prior studies have demonstrated that exposure of cancer cells to FAs affects cell survival, cell proliferation, and cell motility. Oleic acid (OA) has somewhat controversial effects in cancer cells, with both pro- and anti-cancer effects, depending on cell type. Our prior findings suggested that OA enhances cell survival in serum starved HNOA ovarian cancer cells by activating glycolysis, but not β-oxidation. Here, we pharmacologically examined the cellular mechanisms by which OA stimulates glycolysis in HNOA cells. OA induced cell cycle progression, leading to increase in cell number through peroxisome proliferator activated receptor (PPAR) α activation. OA-induced glycolysis was mediated by increased GLUT expression, and increases in GLUT expression were mediated by increased L-MYC expression. Furthermore, L-MYC expression was due to BRD4 activation. These findings suggested involvement of the BRD4-L-MYC-GLUT axis in OA-stimulated glycolysis. These results suggested that OA could activate PPARα to stimulate two pathways: glycolysis and cell cycle progression, and provided insight into the role of OA in ovarian cancer cell growth.
Collapse
Affiliation(s)
- Tsuyoshi Kado
- Division of Molecular Neurobiology, Department of Life Science, Kindai University, Higashiosaka, Japan
| | - Naoki Kusakari
- Division of Molecular Neurobiology, Department of Life Science, Kindai University, Higashiosaka, Japan
| | - Takeru Tamaki
- Division of Molecular Neurobiology, Department of Life Science, Kindai University, Higashiosaka, Japan
| | - Kaeko Murota
- Division of Food and Nutritional Chemistry, Department of Life Science, Kindai University, Higashiosaka, Japan
| | - Toshifumi Tsujiuchi
- Division of Molecular Oncology, Department of Life Science, Kindai University, Higashiosaka, Japan
| | - Nobuyuki Fukushima
- Division of Molecular Neurobiology, Department of Life Science, Kindai University, Higashiosaka, Japan.
| |
Collapse
|
6
|
DiPasquale M, Nguyen MHL, Castillo SR, Dib IJ, Kelley EG, Marquardt D. Vitamin E Does Not Disturb Polyunsaturated Fatty Acid Lipid Domains. Biochemistry 2022; 61:2366-2376. [PMID: 36227768 DOI: 10.1021/acs.biochem.2c00405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The function of vitamin E in biomembranes remains a prominent topic of discussion. As its limitations as an antioxidant persist and novel functions are discovered, our understanding of the role of vitamin E becomes increasingly enigmatic. As a group of lipophilic molecules (tocopherols and tocotrienols), vitamin E has been shown to influence the properties of its host membrane, and a wealth of research has connected vitamin E to polyunsaturated fatty acid (PUFA) lipids. Here, we use contrast-matched small-angle neutron scattering and differential scanning calorimetry to integrate these fields by examining the influence of vitamin E on lipid domain stability in PUFA-based lipid mixtures. The influence of α-tocopherol, γ-tocopherol, and α-tocopherylquinone on the lateral organization of a 1:1 lipid mixture of saturated distearoylphosphatidylcholine (DSPC) and polyunsaturated palmitoyl-linoleoylphosphatidylcholine (PLiPC) with cholesterol provides a complement to our growing understanding of the influence of tocopherol on lipid phases. Characterization of domain melting suggests a slight depression in the transition temperature and a decrease in transition cooperativity. Tocopherol concentrations that are an order of magnitude higher than anticipated physiological concentrations (2 mol percent) do not significantly perturb lipid domains; however, addition of 10 mol percent is able to destabilize domains and promote lipid mixing. In contrast to this behavior, increasing concentrations of the oxidized product of α-tocopherol (α-tocopherylquinone) induces a proportional increase in domain stabilization. We speculate how the contrasting effect of the oxidized product may supplement the antioxidant response of vitamin E.
Collapse
Affiliation(s)
- Mitchell DiPasquale
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, OntarioN9B3P4, Canada
| | - Michael H L Nguyen
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, OntarioN9B3P4, Canada
| | - Stuart R Castillo
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, OntarioN9B3P4, Canada
| | - Isabelle J Dib
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, OntarioN9B3P4, Canada
| | - Elizabeth G Kelley
- NIST Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, Maryland20878, United States
| | - Drew Marquardt
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, OntarioN9B3P4, Canada.,Department of Physics, University of Windsor, Windsor, OntarioN9B3P4, Canada
| |
Collapse
|
7
|
O’Neill KC, Liapis E, Harris BT, Perlin DS, Carter CL. Mass spectrometry imaging discriminates glioblastoma tumor cell subpopulations and different microvascular formations based on their lipid profiles. Sci Rep 2022; 12:17069. [PMID: 36224354 PMCID: PMC9556690 DOI: 10.1038/s41598-022-22093-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 10/10/2022] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma is a prevalent malignant brain tumor and despite clinical intervention, tumor recurrence is frequent and usually fatal. Genomic investigations have provided a greater understanding of molecular heterogeneity in glioblastoma, yet there are still no curative treatments, and the prognosis has remained unchanged. The aggressive nature of glioblastoma is attributed to the heterogeneity in tumor cell subpopulations and aberrant microvascular proliferation. Ganglioside-directed immunotherapy and membrane lipid therapy have shown efficacy in the treatment of glioblastoma. To truly harness these novel therapeutics and develop a regimen that improves clinical outcome, a greater understanding of the altered lipidomic profiles within the glioblastoma tumor microenvironment is urgently needed. In this work, high resolution mass spectrometry imaging was utilized to investigate lipid heterogeneity in human glioblastoma samples. Data presented offers the first insight into the histology-specific accumulation of lipids involved in cell metabolism and signaling. Cardiolipins, phosphatidylinositol, ceramide-1-phosphate, and gangliosides, including the glioblastoma stem cell marker, GD3, were shown to differentially accumulate in tumor and endothelial cell subpopulations. Conversely, a reduction in sphingomyelins and sulfatides were detected in tumor cell regions. Cellular accumulation for each lipid class was dependent upon their fatty acid residue composition, highlighting the importance of understanding lipid structure-function relationships. Discriminating ions were identified and correlated to histopathology and Ki67 proliferation index. These results identified multiple lipids within the glioblastoma microenvironment that warrant further investigation for the development of predictive biomarkers and lipid-based therapeutics.
Collapse
Affiliation(s)
- Kelly C. O’Neill
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA
| | - Evangelos Liapis
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA
| | - Brent T. Harris
- grid.411667.30000 0001 2186 0438Departments of Neurology and Pathology, Georgetown University Medical Center, Washington, D.C. 20007 USA
| | - David S. Perlin
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA ,grid.429392.70000 0004 6010 5947Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ 07110 USA
| | - Claire L. Carter
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA ,grid.429392.70000 0004 6010 5947Department of Pathology, Hackensack Meridian School of Medicine, Nutley, NJ 07110 USA
| |
Collapse
|
8
|
Newell M, Goruk S, Schueler J, Mazurak V, Postovit LM, Field CJ. Docosahexaenoic acid enrichment of tumor phospholipid membranes increases tumor necroptosis in mice bearing triple negative breast cancer patient-derived xenografts. J Nutr Biochem 2022; 107:109018. [PMID: 35489658 DOI: 10.1016/j.jnutbio.2022.109018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 10/04/2021] [Accepted: 03/18/2022] [Indexed: 11/24/2022]
Abstract
Docosahexaenoic acid (DHA) reduces breast cancer tumor growth in preclinical models. To better understand how DHA amplifies the actions of docetaxel (TXT) chemotherapy, we examined the effects of two doses of dietary DHA on tumor size, membrane DHA content and necroptosis using a drug resistant triple negative breast cancer (TNBC) patient derived xenograft (PDX) model. Female NSG mice bearing TNBC PDXs were randomized to one of three nutritionally complete diets (20% w/w fat): control (0% DHA), high DHA (3.8% HDHA), or low DHA (1.6% LDHA) with or without intraperitoneal injections of 5 mg/kg TXT, twice weekly for 6 weeks (n=8 per group). Tumors from mice fed either HDHA+TXT or LDHA+TXT were similar in size to each other, but were 36% and 32% smaller than tumors from mice fed control+TXT, respectively (P<0.05). A dose effect of DHA incorporation was observed in plasma total phospholipids and in phosphatidylethanolamine and phosphatidylinositol. Both doses of DHA resulted in similarly increased necrotic tissue and decreased NFκB protein expression compared to control tumors, however only the HDHA+TXT had increased expression of necroptosis related proteins: RIPK1, RIPK3 and MLKL (P<0.05). Increased MLKL was observed in the lipid raft portion of HDHA+TXT tumor extracts. This work confirms the efficacy of a combination therapy consisting of DHA supplementation and TXT chemotherapy using two doses of DHA as indicated by reduced tumor growth in a TNBC PDX model. Moreover, the results suggest that decreased growth may occur through increased DHA incorporation into tumor phospholipid membranes and necroptosis.
Collapse
Affiliation(s)
- Marnie Newell
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E1
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E1
| | - Julia Schueler
- Charles River Discovery Research Services Germany, Freiburg, Germany
| | - Vera Mazurak
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E1
| | - Lynne-Marie Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R7; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E1.
| |
Collapse
|
9
|
Wei L, Wu Z, Chen YQ. Multi-targeted therapy of cancer by omega-3 fatty acids-an update. Cancer Lett 2022; 526:193-204. [PMID: 34843864 DOI: 10.1016/j.canlet.2021.11.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022]
Abstract
Low in dietary ω3 polyunsaturated fatty acid (PUFA) consumption has been associated with increased incidence of cancers. Many basic and clinical studies have been conducted over the last several decades. We previously reviewed multi-targeted therapy of cancer by omega-3 fatty acids in 2008, and since hundreds of new clinical trials are being conducted to validate the effectiveness of ω3 PUFA in cancer therapy. Because of the availability of such large amount of clinical trial data, in this update we summarize clinical data, sort out trials that show promising results, and discuss potential mechanism(s) responsible for the clinical outcomes. It appears that ω3 PUFA mainly affects cancer-associated symptoms, namely cachexia, inflammation, neuropathy, post operative complications and quality of life. Mechanisms responsible for these effects are possible regulation of skeletal muscle protein turnover, inflammatory response and neuron cell survive by ω3 PUFA.
Collapse
Affiliation(s)
- Lengyun Wei
- Wuxi School of Medicine, Jiangnan University, Jiangsu Province, 214122, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China; School of Food Science and Technology, Jiangnan University, Jiangsu Province, 214122, China
| | - Zhipeng Wu
- Wuxi School of Medicine, Jiangnan University, Jiangsu Province, 214122, China
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, Jiangsu Province, 214122, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China; School of Food Science and Technology, Jiangnan University, Jiangsu Province, 214122, China.
| |
Collapse
|
10
|
Zhao Y, Ma T, Zhang Z, Chen X, Zhou C, Zhang L, Zou D. Resolvin D1 attenuates acid-induced DNA damage in esophageal epithelial cells and rat models of acid reflux. Eur J Pharmacol 2021; 912:174571. [PMID: 34656605 DOI: 10.1016/j.ejphar.2021.174571] [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: 06/06/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 11/15/2022]
Abstract
The role of resolvin D1 (RvD1) in gastroesophageal reflux disease (GERD) remains largely unknown. Here, we investigated the potential role of RvD1 in acid-induced DNA damage in esophageal epithelial cells, patients with refractory GERD and a rat model of acid reflux. Weak acid exposure induced longer comet tails, reactive oxygen species (ROS) generation, oxidative DNA damage and DNA double-strand breaks (DSBs) in cells and RvD1 (0.1 μM) blocked all these effects. Mechanistic analyses showed that apart from ROS-reducing effects, RvD1 possessed a strong capacity to promote DNA damage repair, augmenting cell cycle checkpoint activity and DSB repair by modulating phosphatase and tensin homolog (PTEN) in cells. We also detected the surface expression of formyl peptide receptor 2 (FPR2), a receptor for RvD1, in the esophageal epithelial cells, and inhibition of FPR2 abrogated the protective effects of RvD1 on cells. Furthermore, a positive correlation between RvD1 and PTEN was observed predominantly in the esophageal epithelium from patients with refractory GERD (r = 0.67, P < 0.05). Additionally, RvD1 administration upregulated PTEN, suppressed DNA DSBs and alleviated microscopic damage in the rat model of gastric reflux. FPR2 gene silencing abolished the therapeutic effects of RvD1 on the rat model. Taken together, RvD1 binding to FPR2 protects the esophageal epithelium from acid reflux-induced DNA damage via a mechanism involving the inhibition of ROS production and facilitation of DSB repair. These findings support RvD1 as a promising approach that may be valuable for the treatment of GERD.
Collapse
Affiliation(s)
- Ye Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Teng Ma
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Zhihan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xi Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunhua Zhou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ling Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
11
|
Huang M, Koizumi A, Narita S, Nakanishi H, Sato H, Kashima S, Nara T, Kanda S, Numakura K, Saito M, Satoh S, Nanjo H, Sasaki T, Habuchi T. Altering phosphoinositides in high-fat diet-associated prostate tumor xenograft growth. MedComm (Beijing) 2021; 2:756-764. [PMID: 34977875 PMCID: PMC8706770 DOI: 10.1002/mco2.89] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/08/2022] Open
Abstract
The metabolic reprogramming of phospholipids may affect intracellular signal transduction pathways. A high-fat diet (HFD) is attributed to prostate cancer (PCa) progression, but the expression pattern and role of phospholipids in HFD-mediated PCa progression remains unclear. In this study, HFD enhanced LNCaP xenograft tumor growth by upregulating the phosphatidylinositol (PI) 3-kinase (PI3K)/AKT signaling pathway. A lipidomic analysis using xenograft tumors showed that phosphoinositides, especially PI (3,4,5)-trisphosphate (PIP3), including several species containing C38:4, C38:3, and C40:4 fatty acids, increased in the HFD group compared to control. Fatty acid synthase (FASN) was significantly upregulated in xenograft tumors under HFD in both gene and protein levels. PCa cell growth was significantly inhibited through the decreased AKT signaling pathway by treatment with cerulenin, a chemical FASN inhibitor, which also downregulated PIP, PIP2, and PIP3 but not PI. Thus, dietary fat influences PCa progression and alters phosphoinositides, especially PIP3, a critical player in the PI3K/AKT pathway. These results may offer appropriate targets, such as FASN, for dietary intervention and/or chemoprevention to reduce PCa incidence and progression.
Collapse
Affiliation(s)
- Mingguo Huang
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Atsushi Koizumi
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Shintaro Narita
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Hiroki Nakanishi
- Research Center for BiosignalAkita University Graduate School of MedicineAkitaJapan
| | - Hiromi Sato
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Soki Kashima
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Taketoshi Nara
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Sohei Kanda
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Kazuyuki Numakura
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Mitsuru Saito
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Shigeru Satoh
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Hiroshi Nanjo
- Department of Clinical PathologyAkita University Graduate School of MedicineAkitaJapan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental UniversityBunkyo‐kuTokyoJapan
| | - Tomonori Habuchi
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| |
Collapse
|
12
|
Liu F, Zuo X, Liu Y, Deguchi Y, Moussalli MJ, Chen W, Yang P, Wei B, Tan L, Lorenzi PL, Gao S, Jaoude JC, Mehdizadeh A, Valentin LA, Wei D, Shureiqi I. Suppression of Membranous LRP5 Recycling, Wnt/β-Catenin Signaling, and Colon Carcinogenesis by 15-LOX-1 Peroxidation of Linoleic Acid in PI3P. Cell Rep 2021; 32:108049. [PMID: 32814052 DOI: 10.1016/j.celrep.2020.108049] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/27/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
APC mutation activation of Wnt/β-catenin drives initiation of colorectal carcinogenesis (CRC). Additional factors potentiate β-catenin activation to promote CRC. Western diets are enriched in linoleic acid (LA); LA-enriched diets promote chemically induced CRC in rodents. 15-Lipoxygenase-1 (15-LOX-1), the main LA-metabolizing enzyme, is transcriptionally silenced during CRC. Whether LA and 15-LOX-1 affect Wnt/β-catenin signaling is unclear. We report that high dietary LA promotes CRC in mice treated with azoxymethane or with an intestinally targeted Apc mutation (ApcΔ580) by upregulating Wnt receptor LRP5 protein expression and β-catenin activation. 15-LOX-1 transgenic expression in mouse intestinal epithelial cells suppresses LRP5 protein expression, β-catenin activation, and CRC. 15-LOX-1 peroxidation of LA in phosphatidylinositol-3-phosphates (PI3P_LA) leads to PI3P_13-HODE formation, which decreases PI3P binding to SNX17 and LRP5 and inhibits LRP5 recycling from endosomes to the plasma membrane, thereby increasing LRP5 lysosomal degradation. This regulatory mechanism of LRP5/Wnt/β-catenin signaling could be therapeutically targeted to suppress CRC.
Collapse
Affiliation(s)
- Fuyao Liu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiangsheng Zuo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi Liu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasunori Deguchi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Micheline J Moussalli
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weidong Chen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peiying Yang
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bo Wei
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shen Gao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan C Jaoude
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amir Mehdizadeh
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lovie Ann Valentin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Desale SE, Chinnathambi S. Phosphoinositides signaling modulates microglial actin remodeling and phagocytosis in Alzheimer's disease. Cell Commun Signal 2021; 19:28. [PMID: 33627135 PMCID: PMC7905611 DOI: 10.1186/s12964-021-00715-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/26/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease is one of the neurodegenerative diseases, characterized by the accumulation of abnormal protein deposits, which disrupts signal transduction in neurons and other glia cells. The pathological protein in neurodegenerative diseases, Tau and amyloid-β contribute to the disrupted microglial signaling pathways, actin cytoskeleton, and cellular receptor expression. The important secondary messenger lipids i.e., phosphatidylinositols are largely affected by protein deposits of amyloid-β in Alzheimer's disease. Phosphatidylinositols are the product of different phosphatidylinositol kinases and the state of phosphorylation at D3, D4, and D5 positions of inositol ring. Phosphatidylinositol 3,4,5-triphosphate (PI 3, 4, 5-P3) involves in phagocytic cup formation, cell polarization, whereas Phosphatidylinositol 4,5-bisphosphate (PI 4, 5-P2)-mediates the process of phagosomes formation and further its fusion with early endosome.. The necessary activation of actin-binding proteins such as Rac, WAVE complex, and ARP2/3 complex for the actin polymerization in the process of phagocytosis, migration is regulated and maintained by PI 3, 4, 5-P3 and PI 4, 5-P2. The ratio and types of fatty acid intake can influence the intracellular secondary lipid messengers along with the cellular content of phaphatidylcholine and phosphatidylethanolamine. The Amyloid-β deposits and extracellular Tau seeds disrupt phosphatidylinositides level and actin cytoskeletal network that hamper microglial-signaling pathways in AD. We hypothesize that being a lipid species intracellular levels of phosphatidylinositol would be regulated by dietary fatty acids. Further we are interested to understand phosphoinositide-based signaling cascades in phagocytosis and actin remodeling. Video Abstract.
Collapse
Affiliation(s)
- Smita Eknath Desale
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008 India
| |
Collapse
|
14
|
Story MJ. Zinc, ω-3 polyunsaturated fatty acids and vitamin D: An essential combination for prevention and treatment of cancers. Biochimie 2020; 181:100-122. [PMID: 33307154 DOI: 10.1016/j.biochi.2020.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 11/14/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
Zinc, ω-3 polyunsaturated fatty acids (PUFAs) and vitamin D are essential nutrients for health, maturation and general wellbeing. Extensive literature searches have revealed the widespread similarity in molecular biological properties of zinc, ω-3 PUFAs and vitamin D, and their similar anti-cancer properties, even though they have different modes of action. These three nutrients are separately essential for good health, especially in the aged. Zinc, ω-3 PUFAs and vitamin D are inexpensive and safe as they are fundamentally natural and have the properties of correcting and inhibiting undesirable actions without disturbing the normal functions of cells or their extracellular environment. This review of the anticancer properties of zinc, ω-3 PUFAs and vitamin D is made in the context of the hallmarks of cancer. The anticancer properties of zinc, ω-3 PUFAs and vitamin D can therefore be used beneficially through combined treatment or supplementation. It is proposed that sufficiency of zinc, ω-3 PUFAs and vitamin D is a necessary requirement during chemotherapy treatment and that clinical trials can have questionable integrity if this sufficiency is not checked and maintained during efficacy trials.
Collapse
Affiliation(s)
- Michael J Story
- Story Pharmaceutics Pty Ltd, PO Box 6086, Linden Park, South Australia, 5065, Australia.
| |
Collapse
|
15
|
Tamarindo GH, Góes RM. Docosahexaenoic acid differentially modulates the cell cycle and metabolism- related genes in tumor and pre-malignant prostate cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158766. [PMID: 32712248 DOI: 10.1016/j.bbalip.2020.158766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) has different molecular features along progression, including androgen profile, which is associated to therapy inefficiency leading to more aggressive phenotype. Docosahexaenoic acid (DHA) has antiproliferative and pro-apoptotic properties in different cancers associated to cell metabolism modulation. The latter is of particular interest since metabolic reprogramming is one of PCa hallmarks, but is not clear how this occurs among disease progression. Therefore, we evaluated DHA antiproliferative potential in distinct androgenic backgrounds associated to metabolism modulation and androgen-regulated genes. For this purpose, pre-malignant PNT1A and tumor AR-positive 22rv1, and AR-negative PC3 cells were incubated with DHA at 100 μM-48 h. DHA reduced at least 26% cell number for all lineages due to S-phase decrease in AR-positive and G2/M arrest in AR-negative. Mitochondrial metabolic rate decreased in PNT1A (~38%) and increased in tumor cells (at least 40%). This was associated with ROS overproduction (1.6-fold PNT1A; 2.1 22rv1; 2.2 PC3), lipid accumulation (3-fold PNT1A; 1.8 22rv1; 3.6 PC3) and mitochondria damage in all cell lines. AKT, AMPK and PTEN were not activated in any cell line, but p-ERK1/2 increased (1.5-fold) in PNT1A. Expression of androgen-regulated and nuclear receptors genes showed that DHA affected them in a distinct pattern in each cell line, but most converged to metabolism regulation, response to hormones, lipids and stress. In conclusion, regardless of androgenic or PTEN background DHA exerted antiproliferative effect associated to cell cycle impairment, lipid deregulation and oxidative stress, but differentially regulated gene expression probably due to distinct molecular features of each pathologic stage.
Collapse
Affiliation(s)
| | - Rejane Maira Góes
- Institute of Biology, University of Campinas, Campinas, SP, Brazil; Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP, Brazil.
| |
Collapse
|
16
|
Newell M, Patel D, Goruk S, Field CJ. Docosahexaenoic Acid Incorporation Is Not Affected by Doxorubicin Chemotherapy in either Whole Cell or Lipid Raft Phospholipids of Breast Cancer Cells in vitro and Tumor Phospholipids in vivo. Lipids 2020; 55:549-565. [PMID: 32588470 DOI: 10.1002/lipd.12252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/07/2022]
Abstract
To better understand how docosahexaenoic acid (DHA) improves the effects of doxorubicin (DOX), we examined DHA ± DOX on changes in whole cell and lipid raft phospholipids (PL) of MDA-MB-231 and MCF-7 breast cancer cells. We sought to confirm whether the relative changes in PL DHA content of MDA-MB-231 cells could be extended to PL from MDA-MB-231 tumors grown in mice fed a DHA supplemented diet ±DOX. Treatment with DHA did not change PL composition yet DOX increased the proportion of phosphatidylserine in MCF-7 cell lipid rafts by two-fold (p < 0.001). Regardless of DOX, the relative percent incorporation of DHA was higher in MDA-MB-231 cells compared to MCF-7 cells in phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine (whole cell and lipid rafts); and higher in phosphatidylethanolamine vs. phosphatidylcholine (4.4-fold in MCF-7 and 6-fold in MDA-MB-231 cells respectively). DHA treatment increased eicosapentaenoic acid and docosapentaenoic acid in MDA-MB-231 cells but not MCF-7 cells. Increased DHA content in MDA-MB-231 cells, MCF-7 cells, and MDA-MB-231 tumors in all PL moieties (except sphingomyelin) corresponded with reduced arachidonic acid (p < 0.05). Feeding mice 2.8% (w/w of fat) DHA ± DOX increased tumor necrotic regions (p < 0.05). This study established differential incorporation of DHA into whole cell and lipid rafts between human breast cancer cell lines. However, within each cell line, this incorporation was not altered by DOX confirming that DOX does not change membrane lipid composition. Furthermore, our findings indicate that membrane changes observed in vitro are translatable to in vivo changes and that DHA + DOX could contribute to the anticancer effects through increased necrosis.
Collapse
Affiliation(s)
- Marnie Newell
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Dhruvesh Patel
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| |
Collapse
|
17
|
Fontaine D, Figiel S, Félix R, Kouba S, Fromont G, Mahéo K, Potier-Cartereau M, Chantôme A, Vandier C. Roles of endogenous ether lipids and associated PUFAs in the regulation of ion channels and their relevance for disease. J Lipid Res 2020; 61:840-858. [PMID: 32265321 PMCID: PMC7269763 DOI: 10.1194/jlr.ra120000634] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Indexed: 12/16/2022] Open
Abstract
Ether lipids (ELs) are lipids characterized by the presence of either an ether linkage (alkyl lipids) or a vinyl ether linkage [i.e., plasmalogens (Pls)] at the sn1 position of the glycerol backbone, and they are enriched in PUFAs at the sn2 position. In this review, we highlight that ELs have various biological functions, act as a reservoir for second messengers (such as PUFAs) and have roles in many diseases. Some of the biological effects of ELs may be associated with their ability to regulate ion channels that control excitation-contraction/secretion/mobility coupling and therefore cell physiology. These channels are embedded in lipid membranes, and lipids can regulate their activities directly or indirectly as second messengers or by incorporating into membranes. Interestingly, ELs and EL-derived PUFAs have been reported to play a key role in several pathologies, including neurological disorders, cardiovascular diseases, and cancers. Investigations leading to a better understanding of their mechanisms of action in pathologies have opened a new field in cancer research. In summary, newly identified lipid regulators of ion channels, such as ELs and PUFAs, may represent valuable targets to improve disease diagnosis and advance the development of new therapeutic strategies for managing a range of diseases and conditions.
Collapse
Affiliation(s)
- Delphine Fontaine
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sandy Figiel
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Romain Félix
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sana Kouba
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Gaëlle Fromont
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Department of Pathology, CHRU Bretonneau, F-37044 Tours CEDEX 9, France
| | - Karine Mahéo
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | | | - Aurélie Chantôme
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | - Christophe Vandier
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France. mailto:
| |
Collapse
|
18
|
Liu J, Huang H, Yang Q, Zhao J, Zhang H, Chen W, Peng X, Gu Z. Dietary Supplementation of n-3 LCPUFAs Prevents Salmonellosis in a Murine Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:128-137. [PMID: 31825613 DOI: 10.1021/acs.jafc.9b05899] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Salmonellosis is a world-wide epidemic, and n-3 long chain polyunsaturated fatty acids (LCPUFAs) possess various health benefits. This study is aimed to investigate the preventive effects of n-3 LCPUFAs against Salmonella infection. By pretreatment with n-3 LCPUFAs, but not n-6 LCPUFAs, the survival rate of the infected mice was increased. Further studies showed that n-3 LCPUFAs significantly increased the fecal contents of short-chain fatty acids (SCFAs). The cytokine expression in the liver and production in serum were both modulated by n-3 LCPUFAs into an anti-inflammatory profile against infection. Moreover, the changes in gut microbiota by n-3 LCPUFAs favored the host against pathogens, closely related to the modified SCFA production and immune responses. In conclusion, n-3 LCPUFAs prevented Salmonella infection through multiple mechanisms, especially by the interaction with gut microbiota and host immunology. Our results suggested great perspectives for n-3 LCPUFAs and their related products to control the prevalence of Salmonella, a most predominant food-borne pathogen.
Collapse
Affiliation(s)
- Junsheng Liu
- Department of Food Science and Engineering , Jinan University , Guangzhou , Guangdong 510632 , P. R. China
| | | | | | | | | | - Wei Chen
- Beijing Innovation Centre of Food Nutrition and Human Health , Beijing Technology and Business University (BTBU) , Beijing 100048 , P. R. China
| | - Xichun Peng
- Department of Food Science and Engineering , Jinan University , Guangzhou , Guangdong 510632 , P. R. China
| | | |
Collapse
|
19
|
Abstract
Our own studies and those of others have shown that defects in essential fatty acid (EFA) metabolism occurs in age-related disorders such as obesity, type 2 diabetes mellitus, hypertension, atherosclerosis, coronary heart disease, immune dysfunction and cancer. It has been noted that in all these disorders there could occur a defect in the activities of desaturases, cyclo-oxygenase (COX), and lipoxygenase (LOX) enzymes leading to a decrease in the formation of their long-chain products gamma-linolenic acid (GLA), arachidonic acid, eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA). This leads to an increase in the production of pro-inflammatory prostaglandin E2 (PGE2), thromboxanes (TXs), and leukotrienes (LTs) and a decrease in anti-inflammatory lipoxin A4, resolvins, protectins and maresins. All these bioactive molecules are termed as bioactive lipids (BALs). This imbalance in the metabolites of EFAs leads to low-grade systemic inflammation and at times acute inflammatory events at specific local sites that trigger the development of various age-related disorders such as obesity, type 2 diabetes mellitus, hypertension, coronary heart disease, atherosclerosis, and immune dysfunction as seen in rheumatoid arthritis, lupus, nephritis and other localized inflammatory conditions. This evidence implies that methods designed to restore BALs to normal can prevent age-related disorders and enhance longevity and health.
Collapse
|
20
|
Kawashima M, Tokiwa M, Nishimura T, Kawata Y, Sugimoto M, Kataoka TR, Sakurai T, Iwaisako K, Suzuki E, Hagiwara M, Harris AL, Toi M. High-resolution imaging mass spectrometry combined with transcriptomic analysis identified a link between fatty acid composition of phosphatidylinositols and the immune checkpoint pathway at the primary tumour site of breast cancer. Br J Cancer 2020; 122:245-257. [PMID: 31819188 PMCID: PMC7051979 DOI: 10.1038/s41416-019-0662-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The fatty acid (FA) composition of phosphatidylinositols (PIs) is tightly regulated in mammalian tissue since its disruption impairs normal cellular functions. We previously found its significant alteration in breast cancer by using matrix-assisted laser desorption and ionisation imaging mass spectrometry (MALDI-IMS). METHODS We visualised the histological distribution of PIs containing different FAs in 65 primary breast cancer tissues using MALDI-IMS and investigated its association with clinicopathological features and gene expression profiles. RESULTS Normal ductal cells (n = 7) predominantly accumulated a PI containing polyunsaturated FA (PI-PUFA), PI(18:0/20:4). PI(18:0/20:4) was replaced by PIs containing monounsaturated FA (PIs-MUFA) in all non-invasive cancer cells (n = 12). While 54% of invasive cancer cells (n = 27) also accumulated PIs-MUFA, 46% of invasive cancer cells (n = 23) accumulated the PIs-PUFA, PI(18:0/20:3) and PI(18:0/20:4). The accumulation of PI(18:0/20:3) was associated with higher incidence of lymph node metastasis and activation of the PD-1-related immune checkpoint pathway. Fatty acid-binding protein 7 was identified as a putative molecule controlling PI composition. CONCLUSIONS MALDI-IMS identified PI composition associated with invasion and nodal metastasis of breast cancer. The accumulation of PI(18:0/20:3) could affect the PD-1-related immune checkpoint pathway, although its precise mechanism should be further validated.
Collapse
Affiliation(s)
- Masahiro Kawashima
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan.
- Molecular Oncology Laboratories, Wheaterall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - Mariko Tokiwa
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Tomomi Nishimura
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Yukiko Kawata
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Masahiro Sugimoto
- Health Promotion and Preemptive Medicine, Research and Development Center for Minimally Invasive Therapies, Tokyo Medical University, Sinjuku-ku, Tokyo, 160-8402, Japan
| | - Tatsuki R Kataoka
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takaki Sakurai
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Keiko Iwaisako
- Department of Target Therapy Oncology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eiji Suzuki
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Adrian L Harris
- Molecular Oncology Laboratories, Wheaterall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| |
Collapse
|
21
|
Effect of dietary omega-3 fatty acids on castrate-resistant prostate cancer and tumor-associated macrophages. Prostate Cancer Prostatic Dis 2019; 23:127-135. [PMID: 31439889 PMCID: PMC7031053 DOI: 10.1038/s41391-019-0168-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/01/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022]
Abstract
Background M2-like macrophages are associated with the pathogenesis of castrate resistant prostate cancer (CRPC). We sought to determine if dietary omega-3 fatty acids (ω−3 FAs) delay the development and progression of CRPC and inhibit tumor associated M2-like macrophages. Methods MycCap cells were grown subcutaneously in immunocompetent FVB mice. Mice were castrated when tumors reached 300mm3. To study effects of dietary ω−3 FAs on development of CRPC, ω−3 or ω−6 diets were started two days after castration and mice sacrificed after early regrowth of tumors. To study ω−3 FA effects on progression of CRPC, tumors were allowed to regrow after castration before starting the diets. M2 (CD206+) macrophages were isolated from allografts to examine ω−3 FA effects on macrophage function. Omega-3 fatty acid effects on androgen-deprived RAW264.7 M2 macrophages was studied by RTqPCR and a migration/ invasion assay. Results The ω−3 diet combined with castration lead to greater MycCap tumor regression (182.2±33.6 mm3) than the ω−6 diet (148.3±35.2; p=0.003) and significantly delayed the time to CRPC (p=0.006). Likewise, the ω−3 diet significantly delayed progression of established castrate resistant MycCaP tumors (p=0.003). The ω−3 diet (as compared to the ω−6 diet) significantly reduced tumor associated M2-like macrophage expression of CSF-1R in the CRPC development model, and matrix metallopeptidase-9 (MMP-9) and vascular endothelial growth factor (VEGF) in the CRPC progression model. Migration of androgen depleted RAW264.7 M2 macrophages towards MycCaP cells was reversed by addition of docosahexaenoic acid (ω−3). Conclusions Dietary omega-3 FAs (as compared to omega-6 FAs) decreased the development and progression of CRPC in an immunocompetent mouse model, and had inhibitory effects on M2-like macrophage function. Clinical trials are warranted evaluating if a fish oil-based diet can delay the time to castration resistance in men on androgen deprivation therapy, whereas further pre-clinical studies are warranted evaluating fish oil for more advanced CRPC.
Collapse
|
22
|
Mansi IA, Al-Sha'er MA, Mhaidat NM, Taha MO, Shahin R. Investigation of Binding Characteristics of Phosphoinositide-dependent Kinase-1 (PDK1) Co-crystallized Ligands Through Virtual Pharmacophore Modeling Leading to Novel Anti-PDK1 Hits. Med Chem 2019; 16:860-880. [PMID: 31339076 DOI: 10.2174/1573406415666190724131048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND 3-Phosphoinositide Dependent Protein Kinase-1 (PDK1) is being lately considered as an attractive and forthcoming anticancer target. A Protein Data Bank (PDB) cocrystallized crystal provides not only rigid theoretical data but also a realistic molecular recognition data that can be explored and used to discover new hits. OBJECTIVE This incited us to investigate the co-crystallized ligands' contacts inside the PDK1 binding pocket via a structure-based receptor-ligand pharmacophore generation technique in Discovery Studio 4.5 (DS 4.5). METHODS Accordingly, 35 crystals for PDK1 were collected and studied. Every single receptorligand interaction was validated and the significant ones were converted into their corresponding pharmacophoric features. The generated pharmacophores were scored by the Receiver Operating Characteristic (ROC) curve analysis. RESULTS Consequently, 169 pharmacophores were generated and sorted, 11 pharmacophores acquired good ROC-AUC results of 0.8 and a selectivity value above 8. Pharmacophore 1UU3_2_01 was used in particular as a searching filter to screen NCI database because of its acceptable validity criteria and its distinctive positive ionizable feature. Several low micromolar PDK1 inhibitors were revealed. The most potent hit illustrated anti-PDK1 IC50 values of 200 nM with 70% inhibition against SW480 cell lines. CONCLUSION Eventually, the active hits were docked inside the PDK1 binding pocket and the recognition points between the active hits and the receptor were analyzed that led to the discovery of new scaffolds as potential PDK1 inhibitors.
Collapse
Affiliation(s)
- Iman A Mansi
- Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. Box 330127 Zarqa, 13133 Jordan
| | | | - Nizar M Mhaidat
- Clinical Pharmacy Department, Faculty of Pharmacy, Jordan University of Science & Technology, Irbid, Jordan
| | - Mutasem O Taha
- Drug Design Center, Faculty of Pharmacy, University of Jordan, Amman, Jordan,Faculty of Pharmacy, Applied Science University, Amman, Jordan
| | - Rand Shahin
- Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. Box 330127 Zarqa, 13133 Jordan
| |
Collapse
|
23
|
Xu F, Song Y, Guo A. Anti-Apoptotic Effects of Docosahexaenoic Acid in IL-1β-Induced Human Chondrosarcoma Cell Death through Involvement of the MAPK Signaling Pathway. Cytogenet Genome Res 2019; 158:17-24. [PMID: 31261155 DOI: 10.1159/000500290] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2018] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease characterized by progressive articular cartilage destruction and joint marginal osteophyte formation with different degrees of synovitis. Docosahexaenoic acid (DHA) is an unsaturated fatty acid with anti-inflammatory, antioxidant, and antiapoptotic functions. In this study, the human chondrosarcoma cell line SW1353 was cultured in vitro, and an OA cell model was constructed with inflammatory factor IL-1β stimulation. After cells were treated with DHA, cell apoptosis was measured. Western blot assay was used to detect protein expression of apoptosis-related factors (Bax, Bcl-2, and cleaved caspase-3) and mitogen-activated protein kinase (MAPK) signaling pathway family members, including extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK), and p38 MAPK. Our results show that IL-1β promotes the apoptosis of SW1353 cells, increases the expression of Bax and cleaved caspase-3, and activates the MAPK signaling pathway. In contrast, DHA inhibits the expression of IL-1β, inhibits IL-1β-induced cell apoptosis, and has a certain inhibitory effect on the activation of the MAPK signaling pathway. When the MAPK signaling pathway is inhibited by its inhibitors, the effects of DHA on SW1353 cells are weakened. Thus, DHA enhances the apoptosis of SW1353 cells through the MAPK signaling pathway.
Collapse
|
24
|
Rahmani B, Hamedi Asl D, Naserpour Farivar T, Azad M, Sahmani M, Gheibi N. Omega-3 PUFA Alters the Expression Level but Not the Methylation Pattern of the WIF1 Gene Promoter in a Pancreatic Cancer Cell Line (MIA PaCa-2). Biochem Genet 2019; 57:477-486. [PMID: 30649640 DOI: 10.1007/s10528-018-9895-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 11/14/2018] [Indexed: 01/17/2023]
Abstract
Pancreatic cancer is the fourth leading cause of death in both males and females, with a 5-year relative survival rate of 8%. The Wnt signaling pathway has a significant role in the pathogenesis of many tumors, including those of pancreatic cancer. Hypermethylation of the Wnt inhibitory Factor-1 (WIF1) gene promoter have been detected in different types of cancer. In contrast, the anticancer effects of long-chain omega-3 PUFA (ALA) have been reported. Regarding its anticancer effects, in this study, we investigated the effects of various concentrations of omega-3 PUFA on expression level and promoter methylation of the WIF1 gene in MIA PaCa-2 cells in 24, 48, and 72 h after treatment. MIA PaCa-2 cells were treated with different concentrations of omega-3 PUFA (25, 50, 100, 250, 500, and 1000 μM). Cell viability assay was carried out followed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and methylation-specific PCR (MSP). This investigation suggested that dietary consumption of omega-3 PUFAs (250-1000 μM) has a significant effect on the proliferation and WIF1 gene expression of the MIA PaCa-2 cancer cell line but no effect on the promoter methylation of this gene. Changes in promoter methylation were not observed in any of the treatments.
Collapse
Affiliation(s)
- Babak Rahmani
- Department of Molecular Medicine, Faculty of Medical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Dariush Hamedi Asl
- Department of Molecular Medicine, Faculty of Medical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Mehdi Azad
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehdi Sahmani
- Department of Clinical Biochemistry and Genetic, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Nematollah Gheibi
- Cellular and Molecular Research Centre, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
25
|
Gevariya N, Besançon M, Robitaille K, Picard V, Diabaté L, Alesawi A, Julien P, Fradet Y, Bergeron A, Fradet V. Omega-3 fatty acids decrease prostate cancer progression associated with an anti-tumor immune response in eugonadal and castrated mice. Prostate 2019; 79:9-20. [PMID: 30073695 DOI: 10.1002/pros.23706] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 07/13/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Several lines of evidence suggest effects of dietary fat on prostate cancer (PCa) development and progression. Targeting omega (ω)-3:ω6 fatty acids (FA) ratio could be beneficial against PCa by favorably modulating inflammation. Here, we studied the effects of ω3- and ω6-enriched diets on prostate tumor growth and inflammatory response in androgen-deprived and non-deprived conditions. METHODS Immune-competent eugonadal and castrated C57BL/6 mice were injected with TRAMP-C2 prostate tumor cells and daily fed with ω3- or ω6-enriched diet. FA and cytokine profiles were measured in blood and tumors using gas chromatography and multiplex immunoassay, respectively. Immune cell infiltration in tumors was profiled by multicolor flow cytometry. RESULTS ω3-enriched diet decreased prostate TRAMP-C2 tumor growth in immune-competent eugonadal and castrated mice. Cytokines associated with Th1 immune response (IL-12 [p70], IFN-γ, GM-CSF) and eosinophil recruitment (eotaxin-1, IL-5, and IL-13) were significantly elevated in tumors of ω3-fed mice. Using in vitro experiments, we confirmed ω3 FA-induced eotaxin-1 secretion by tumor cells and that eotaxin-1 secretion was regulated by androgens. Analysis of immune cell infiltrating tumors showed no major difference of immune cells' abundance between ω3- and ω6-enriched diets. CONCLUSIONS ω3-enriched diet reduces prostate tumor growth independently of androgen levels. ω3 FA can inhibit tumor cell growth and induce a local anti-tumor inflammatory response. These findings warrant further examination of dietary ω3's potential to slow down the progression of androgen-sensitive and castrate-resistant PCa by modulating immune cell function in tumors.
Collapse
Affiliation(s)
- Nikunj Gevariya
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
| | - Marjorie Besançon
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
| | - Karine Robitaille
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Valérie Picard
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Lamoussa Diabaté
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
| | - Anwar Alesawi
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Pierre Julien
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
- Endocrinology and Nephrology Axis, Centre de recherche du CHU de Québec-Université Laval-CHUL, Québec, Quebec, Canada
| | - Yves Fradet
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
| | - Alain Bergeron
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
| | - Vincent Fradet
- Laboratoire d'Uro-Oncologie Expérimentale, Oncology Axis, Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
- Faculté de Medicine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
26
|
Figiel S, Pinault M, Domingo I, Guimaraes C, Guibon R, Besson P, Tavernier E, Blanchet P, Multigner L, Bruyère F, Haillot O, Mathieu R, Vincendeau S, Rioux-Leclercq N, Lebdai S, Azzouzi AR, Perrouin-Verbe MA, Fournier G, Doucet L, Rigaud J, Renaudin K, Mahéo K, Fromont G. Fatty acid profile in peri-prostatic adipose tissue and prostate cancer aggressiveness in African-Caribbean and Caucasian patients. Eur J Cancer 2018; 91:107-115. [PMID: 29413967 DOI: 10.1016/j.ejca.2017.12.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/29/2017] [Accepted: 12/11/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Genetic and nutritional factors have been linked to the risk of aggressive prostate cancer (PCa). The fatty acid (FA) composition of peri-prostatic adipose tissue (PPAT), which reflects the past FA intake, is potentially involved in PCa progression. We analysed the FA composition of PPAT, in correlation with the ethno-geographical origin of the patients and markers of tumour aggressiveness. METHODS From a cohort of 1000 men treated for PCa by radical prostatectomy, FA composition of PPAT was analysed in 156 patients (106 Caucasians and 50 African-Caribbeans), 78 with an indolent tumour (ISUP group 1 + pT2 + PSA <10 ng/mL) and 78 with an aggressive tumour (ISUP group 4-5 + pT3). The effect of FA extracted from PPAT on in-vitro migration of PCa cells DU145 was studied in 72 patients, 36 Caucasians, and 36 African-Caribbeans. RESULTS FA composition differed according to the ethno-geographical origin. Linoleic acid, an essential n-6 FA, was 2-fold higher in African-Caribbeans compared with Caucasian patients, regardless of disease aggressiveness. In African-Caribbeans, the FA profile associated with PCa aggressiveness was characterised by low level of linoleic acid along with high levels of saturates. In Caucasians, a weak and negative association was observed between eicosapentaenoic acid level (an n-3 FA) and disease aggressiveness. In-vitro migration of PCa cells using PPAT from African-Caribbean patients was associated with lower content of linoleic acid. CONCLUSION These results highlight an important ethno-geographical variation of PPAT, in both their FA content and association with tumour aggressiveness.
Collapse
Affiliation(s)
- Sandy Figiel
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France
| | - Michelle Pinault
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France
| | - Isabelle Domingo
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France
| | - Cyrille Guimaraes
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France
| | - Roseline Guibon
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France; CHRU Bretonneau, Department of Pathology, Tours, France; CHRU Bretonneau, Department of Urology, Tours, France
| | - Pierre Besson
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France
| | - Elsa Tavernier
- CHRU Tours, Clinical Investigation Center - INSERM 1415, Tours, France
| | - Pascal Blanchet
- CHU Pointe à Pitre, Department of Urology, Guadeloupe, France; Inserm UMR1085 - IRSET, Rennes, France
| | | | - Franck Bruyère
- CHRU Bretonneau, Department of Pathology, Tours, France; CHRU Bretonneau, Department of Urology, Tours, France
| | - Olivier Haillot
- CHRU Bretonneau, Department of Pathology, Tours, France; CHRU Bretonneau, Department of Urology, Tours, France
| | - Romain Mathieu
- CHU Rennes, Department of Pathology, Rennes, France; CHU Rennes, Department of Urology, Rennes, France
| | - Sebastien Vincendeau
- CHU Rennes, Department of Pathology, Rennes, France; CHU Rennes, Department of Urology, Rennes, France
| | - Nathalie Rioux-Leclercq
- CHU Rennes, Department of Pathology, Rennes, France; CHU Rennes, Department of Urology, Rennes, France
| | | | | | | | - Georges Fournier
- CHU Brest, Department of Pathology, Brest, France; CHU Brest, Department of Urology, Brest, France
| | - Laurent Doucet
- CHU Brest, Department of Pathology, Brest, France; CHU Brest, Department of Urology, Brest, France
| | - Jerome Rigaud
- CHU Nantes, Department of Pathology, Nantes, France; CHU Nantes, Department of Urology, Nantes, France
| | - Karine Renaudin
- CHU Nantes, Department of Pathology, Nantes, France; CHU Nantes, Department of Urology, Nantes, France
| | - Karine Mahéo
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France
| | - Gaëlle Fromont
- Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Faculté de Médecine, 10 Bd Tonnellé, 37032 Tours, France; CHRU Bretonneau, Department of Pathology, Tours, France; CHRU Bretonneau, Department of Urology, Tours, France.
| |
Collapse
|
27
|
Discovery of a small-molecule inhibitor of specific serine residue BAD phosphorylation. Proc Natl Acad Sci U S A 2018; 115:E10505-E10514. [PMID: 30309962 DOI: 10.1073/pnas.1804897115] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human BCL-2-associated death promoter (hBAD) is an apoptosis-regulatory protein mediating survival signals to carcinoma cells upon phosphorylation of Ser99, among other residues. Herein, we screened multiple small-molecule databases queried in a Laplacian-modified naive Bayesian-based cheminformatics platform and identified a Petasis reaction product as a site-specific inhibitor for hBAD phosphorylation. Based on apoptotic efficacy against mammary carcinoma cells, N-cyclopentyl-3-((4-(2,3-dichlorophenyl) piperazin-1-yl) (2-hydroxyphenyl) methyl) benzamide (NPB) was identified as a potential lead compound. In vitro biochemical analyses demonstrated that NPB inhibited the phosphorylation of hBAD specifically on Ser99. NPB was observed to exert this effect independently of AKT and other kinase activities despite the demonstration of AKT-mediated BAD-Ser99 phosphorylation. Using a structure-based bioinformatics platform, we observed that NPB exhibited predicted interactions with hBAD in silico and verified the same by direct binding kinetics. NPB reduced phosphorylation of BAD-Ser99 and enhanced caspase 3/7 activity with associated loss of cell viability in various human cancer cell lines derived from mammary, endometrial, ovarian, hepatocellular, colon, prostatic, and pancreatic carcinoma. Furthermore, by use of a xenograft model, it was observed that NPB, as a single agent, markedly diminished BAD phosphorylation in tumor tissue and significantly inhibited tumor growth. Similar doses of NPB utilized in acute toxicity studies in mice did not exhibit significant effects. Hence, we report a site-specific inhibitor of BAD phosphorylation with efficacy in tumor models.
Collapse
|
28
|
Diedrich JD, Herroon MK, Rajagurubandara E, Podgorski I. The Lipid Side of Bone Marrow Adipocytes: How Tumor Cells Adapt and Survive in Bone. Curr Osteoporos Rep 2018; 16:443-457. [PMID: 29869753 PMCID: PMC6853185 DOI: 10.1007/s11914-018-0453-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Bone marrow adipocytes have emerged in recent years as key contributors to metastatic progression in bone. In this review, we focus specifically on their role as the suppliers of lipids and discuss pro-survival pathways that are closely linked to lipid metabolism, affected by the adipocyte-tumor cell interactions, and likely impacting the ability of the tumor cell to thrive in bone marrow space and evade therapy. RECENT FINDINGS The combined in silico, pre-clinical, and clinical evidence shows that in adipocyte-rich tissues such as bone marrow, tumor cells rely on exogenous lipids for regulation of cellular energetics and adaptation to harsh metabolic conditions of the metastatic niche. Adipocyte-supplied lipids have a potential to alter the cell's metabolic decisions by regulating glycolysis and respiration, fatty acid oxidation, lipid desaturation, and PPAR signaling. The downstream effects of lipid signaling on mitochondrial homeostasis ultimately control life vs. death decisions, providing a mechanism for gaining survival advantage and reduced sensitivity to treatment. There is a need for future research directed towards identifying the key metabolic and signaling pathways that regulate tumor dependence on exogenous lipids and consequently drive the pro-survival behavior in the bone marrow niche.
Collapse
Affiliation(s)
- Jonathan D Diedrich
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield, Rm 6304, Detroit, MI, 48201, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mackenzie K Herroon
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield, Rm 6304, Detroit, MI, 48201, USA
| | - Erandi Rajagurubandara
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield, Rm 6304, Detroit, MI, 48201, USA
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield, Rm 6304, Detroit, MI, 48201, USA.
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
29
|
Acland M, Mittal P, Lokman NA, Klingler-Hoffmann M, Oehler MK, Hoffmann P. Mass Spectrometry Analyses of Multicellular Tumor Spheroids. Proteomics Clin Appl 2018; 12:e1700124. [PMID: 29227035 DOI: 10.1002/prca.201700124] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/13/2017] [Indexed: 12/21/2022]
Abstract
Multicellular tumor spheroids (MCTS) are a powerful biological in vitro model, which closely mimics the 3D structure of primary avascularized tumors. Mass spectrometry (MS) has established itself as a powerful analytical tool, not only to better understand and describe the complex structure of MCTS, but also to monitor their response to cancer therapeutics. The first part of this review focuses on traditional mass spectrometry approaches with an emphasis on elucidating the molecular characteristics of these structures. Then the mass spectrometry imaging (MSI) approaches used to obtain spatially defined information from MCTS is described. Finally the analysis of primary spheroids, such as those present in ovarian cancer, and the great potential that mass spectrometry analysis of these structures has for improved understanding of cancer progression and for personalized in vitro therapeutic testing is discussed.
Collapse
Affiliation(s)
- Mitchell Acland
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Institute of Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, Australia
| | - Parul Mittal
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Institute of Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, Australia
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Manuela Klingler-Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Peter Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
30
|
Tanaka A, Yamamoto A, Murota K, Tsujiuchi T, Iwamori M, Fukushima N. Polyunsaturated fatty acids induce ovarian cancer cell death through ROS-dependent MAP kinase activation. Biochem Biophys Res Commun 2017; 493:468-473. [DOI: 10.1016/j.bbrc.2017.08.168] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 01/10/2023]
|
31
|
Sun Y, Jia X, Hou L, Liu X, Gao Q. Involvement of apoptotic pathways in docosahexaenoic acid-induced benefit in prostate cancer: Pathway-focused gene expression analysis using RT 2 Profile PCR Array System. Lipids Health Dis 2017; 16:59. [PMID: 28330470 PMCID: PMC5363041 DOI: 10.1186/s12944-017-0442-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/07/2017] [Indexed: 11/12/2022] Open
Abstract
Background Present study aimed to better understand the potential apoptotic pathways that involved in docosahexaenoic acid (DHA)-induced apoptosis of prostate cancer cells. Methods Human prostate cancer DU145 cells were treated with different concentrations of fish oil, omega-3 PUFA (DHA, and Eicosapentaenoic acid, EPA), or omega-6 PUFA (Arachidonic acid, AA). Cell viability and apoptosis were evaluated by MTT assay and Hoechst staining. Pathway-focused gene expression profiling of DU145 cells was analyzed with the RT2 Profile PCR Array System. The results were verified by real time quantitative polymerase chain reaction (RT-qPCR). Results AA exposure showed no obvious effect on viability of DU145 cells. However, exposure with fish oil, EPA, or DHA for 24 h significantly affected cell viability. The growth inhibition of DHA was more pronounced than that of EPA and showed a time-dependent increase. DHA exposure caused typical apoptotic characteristics. Ten genes were more expressed, while 5 genes were less expressed following DHA exposure. RT-qPCR confirmed the time dependent effect of DHA on the expression of these differentially expressed genes. KEGG pathway analysis showed that DHA may induce the apoptosis of cancer cells preferentially through mediating P53, MAPK, TNF, PI3K/AKT, and NF-κB signaling pathways. Conclusion Our study demonstrated the beneficial action of DHA on human prostate carcinoma cell line DU145. The pro-apoptotic effect of DHA on DU145 cells may involve mediation various pathways, especially P53, MAPK, TNF, PI3K/AKT, and NF-κB signaling pathways. Molecular mechanisms of DHA on apoptosis of cancer cells still need to be further clarified. Electronic supplementary material The online version of this article (doi:10.1186/s12944-017-0442-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanan Sun
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital of the People's Liberation Army, Shijiazhuang, 050082, China
| | - Xiaopeng Jia
- Department of Urology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| | - Lianguo Hou
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xing Liu
- Department of Orthopaedic Trauma, Section II, The Third Hospital of Shijiazhuang City, Shijiazhuang, Hebei, 050011, China
| | - Qiang Gao
- Department of Nutrition and Food hygiene, Hebei Medical University, Shijiazhuang, 050017, China
| |
Collapse
|
32
|
Aucoin M, Cooley K, Knee C, Fritz H, Balneaves LG, Breau R, Fergusson D, Skidmore B, Wong R, Seely D. Fish-Derived Omega-3 Fatty Acids and Prostate Cancer: A Systematic Review. Integr Cancer Ther 2017; 16:32-62. [PMID: 27365385 PMCID: PMC5736071 DOI: 10.1177/1534735416656052] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/16/2016] [Accepted: 05/18/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The use of natural health products in prostate cancer (PrCa) is high despite a lack of evidence with respect to safety and efficacy. Fish-derived omega-3 fatty acids possess anti-inflammatory effects and preclinical data suggest a protective effect on PrCa incidence and progression; however, human studies have yielded conflicting results. METHODS A search of OVID MEDLINE, Pre-MEDLINE, Embase, and the Allied and Complementary Medicine Database (AMED) was completed for human interventional or observational data assessing the safety and efficacy of fish-derived omega-3 fatty acids in the incidence and progression of PrCa. RESULTS Of 1776 citations screened, 54 publications reporting on 44 studies were included for review and analysis: 4 reports of 3 randomized controlled trials, 1 nonrandomized clinical trial, 20 reports of 14 cohort studies, 26 reports of 23 case-control studies, and 3 case-cohort studies. The interventional studies using fish oil supplements in patients with PrCa showed no impact on prostate-specific antigen levels; however, 2 studies showed a decrease in inflammatory or other cancer markers. A small number of mild adverse events were reported and interactions with other interventions were not assessed. Cohort and case-control studies assessing the relationship between dietary fish intake and the risk of PrCa were equivocal. Cohort studies assessing the risk of PrCa mortality suggested an association between higher intake of fish and decreased risk of prostate cancer-related death. CONCLUSIONS Current evidence is insufficient to suggest a relationship between fish-derived omega-3 fatty acid and risk of PrCa. An association between higher omega-3 intake and decreased PrCa mortality may be present but more research is needed. More intervention trials or observational studies with precisely measured exposure are needed to assess the impact of fish oil supplements and dietary fish-derived omega-3 fatty acid intake on safety, PrCa incidence, treatment, and progression.
Collapse
Affiliation(s)
- Monique Aucoin
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | - Kieran Cooley
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | - Christopher Knee
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | - Heidi Fritz
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | | | - Rodney Breau
- Ottawa Hospital General Campus, Ottawa, Ontario, Canada
| | - Dean Fergusson
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Becky Skidmore
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | | | - Dugald Seely
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Ottawa Integrative Cancer Centre, Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Modulation of Ras/ERK and Phosphoinositide Signaling by Long-Chain n-3 PUFA in Breast Cancer and Their Potential Complementary Role in Combination with Targeted Drugs. Nutrients 2017; 9:nu9030185. [PMID: 28241486 PMCID: PMC5372848 DOI: 10.3390/nu9030185] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/16/2017] [Accepted: 02/19/2017] [Indexed: 12/14/2022] Open
Abstract
A potential complementary role of the dietary long-chain n-3 polyunsaturated fatty acids (LCn-3 PUFA) in combination with innovative mono-targeted therapies has recently been proposed. These compounds are thought to act pleiotropically to prevent the development and progression of a variety of cancers, including breast cancer. We hereinafter critically analyze the reports investigating the ability of LCn-3 PUFA to modulate the Ras/ERK and the phosphoinositide survival signaling pathways often aberrantly activated in breast cancer and representing the main targets of innovative therapies. The in vitro or in vivo animal and human interventional studies published up to January 2017 investigating the effects of LCn-3 PUFA on these pathways in normal and cancerous breast cells or tissues were identified through a systematic search of literature in the PubMed database. We found that, in most cases, both the in vitro and in vivo studies demonstrated the ability of LCn-3 PUFA to inhibit the activation of these pro-survival pathways. Altogether, the analyzed results strongly suggest a potential role of LCn-3 PUFA as complementary agents in combination with mono-targeted therapies. Moreover, the results indicate the need for further in vitro and human interventional studies designed to unequivocally prove the potential adjuvant role of these fatty acids.
Collapse
|
34
|
Zhao H, Pflug BR, Lai X, Wang M. Pyruvate dehydrogenase alpha 1 as a target of omega-3 polyunsaturated fatty acids in human prostate cancer through a global phosphoproteomic analysis. Proteomics 2016; 16:2419-31. [PMID: 27357730 DOI: 10.1002/pmic.201600166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/23/2016] [Accepted: 06/27/2016] [Indexed: 01/26/2023]
Abstract
Prostate cancer is one of the leading cancers in men. Taking dietary supplements, such as fish oil (FO), which is rich in n-3 polyunsaturated fatty acids (PUFAs), has been employed as a strategy to lower prostate cancer risk and control disease progression. In this study, we investigated the global phosphoproteomic changes induced by FO using a combination of phosphoprotein-enrichment strategy and high-resolution tandem mass spectrometry. We found that FO induces many more phosphorylation changes than oleic acid when they both are compared to control group. Quantitative comparison between untreated group and FO- or oleic acid-treated groups uncovered a number of important protein phosphorylation changes induced by n-3PUFAs. This phosphoproteomic discovery study and the follow-up Western Blot validation study elucidate that phosphorylation levels of the two regulatory serine residues in pyruvate dehydrogenase alpha 1 (PDHA1), serine-232 and serine-300, are significantly decreased upon FO treatment. As expected, increased pyruvate dehydrogenase activity was also observed. This study suggests that FO-induced phosphorylation changes in PDHA1 is more likely related to the glucose metabolism pathway, and n-3 PUFAs may have a role in controlling the balance between lipid and glucose oxidation.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Beth R Pflug
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xianyin Lai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mu Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
35
|
Cinnamon E, Makki R, Sawala A, Wickenberg LP, Blomquist GJ, Tittiger C, Paroush Z, Gould AP. Drosophila Spidey/Kar Regulates Oenocyte Growth via PI3-Kinase Signaling. PLoS Genet 2016; 12:e1006154. [PMID: 27500738 PMCID: PMC4976899 DOI: 10.1371/journal.pgen.1006154] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 06/08/2016] [Indexed: 11/19/2022] Open
Abstract
Cell growth and proliferation depend upon many different aspects of lipid metabolism. One key signaling pathway that is utilized in many different anabolic contexts involves Phosphatidylinositide 3-kinase (PI3K) and its membrane lipid products, the Phosphatidylinositol (3,4,5)-trisphosphates. It remains unclear, however, which other branches of lipid metabolism interact with the PI3K signaling pathway. Here, we focus on specialized fat metabolizing cells in Drosophila called larval oenocytes. In the presence of dietary nutrients, oenocytes undergo PI3K-dependent cell growth and contain very few lipid droplets. In contrast, during starvation, oenocytes decrease PI3K signaling, shut down cell growth and accumulate abundant lipid droplets. We now show that PI3K in larval oenocytes, but not in fat body cells, functions to suppress lipid droplet accumulation. Several enzymes of fatty acid, triglyceride and hydrocarbon metabolism are required in oenocytes primarily for lipid droplet induction rather than for cell growth. In contrast, a very long chain fatty-acyl-CoA reductase (FarO) and a putative lipid dehydrogenase/reductase (Spidey, also known as Kar) not only promote lipid droplet induction but also inhibit oenocyte growth. In the case of Spidey/Kar, we show that the growth suppression mechanism involves inhibition of the PI3K signaling pathway upstream of Akt activity. Together, the findings in this study show how Spidey/Kar and FarO regulate the balance between the cell growth and lipid storage of larval oenocytes. Lipids play diverse roles in health and disease. Some types of lipids function as metabolic fuels for energy homeostasis, whereas others act as components of cell membranes or serve as signals regulating cell behaviors. Much, however, remains to be discovered about the molecular connections between different categories of lipids. Phosphatidylinositide 3-kinase (PI3K) is an enzyme that synthesizes phosphatidylinositide lipids, which act as signals essential for growth during normal development and cancer. Using genetics in the fruit fly, Drosophila, we identify new regulatory links between phosphatidylinositides and lipid oxidoreductases in specialized fat-metabolizing cells called oenocytes. We find that an enzyme metabolizing very long chain fatty acids (VLCFAs) and also a putative lipid dehydrogenase/reductase both act to prevent the inappropriate overgrowth of oenocytes. In the case of the latter enzyme, it suppresses cell growth by inhibiting phosphatidylinositide signaling. Future studies will determine whether similar lipid enzymes regulate PI3K signaling in other cell and tissue types during normal development and tumorigenesis.
Collapse
Affiliation(s)
- Einat Cinnamon
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, United Kingdom
| | - Rami Makki
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, United Kingdom
| | - Annick Sawala
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, United Kingdom
| | - Leah P. Wickenberg
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, Nevada, United States of America
| | - Gary J. Blomquist
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, Nevada, United States of America
| | - Claus Tittiger
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Reno, Nevada, United States of America
| | - Ze'ev Paroush
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, the Hebrew University, Jerusalem, Israel
| | - Alex P. Gould
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Hiraide T, Ikegami K, Sakaguchi T, Morita Y, Hayasaka T, Masaki N, Waki M, Sugiyama E, Shinriki S, Takeda M, Shibasaki Y, Miyazaki S, Kikuchi H, Okuyama H, Inoue M, Setou M, Konno H. Accumulation of arachidonic acid-containing phosphatidylinositol at the outer edge of colorectal cancer. Sci Rep 2016; 6:29935. [PMID: 27435310 PMCID: PMC4951683 DOI: 10.1038/srep29935] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence indicates that cancer cells show specific alterations in phospholipid metabolism that contribute to tumour progression in several types of cancer, including colorectal cancer. Questions still remain as to what lipids characterize the outer edge of cancer tissues and whether those cancer outer edge-specific lipid compositions emerge autonomously in cancer cells. Cancer tissue-originated spheroids (CTOSs) that are composed of pure primary cancer cells have been developed. In this study, we aimed to seek out the cancer cell-autonomous acquisition of cancer outer edge-characterizing lipids in colorectal cancer by analysing phospholipids in CTOSs derived from colorectal cancer patients with matrix-assisted laser desorption/ionization (MALDI)-imaging mass spectrometry (IMS). A signal at m/z 885.5 in negative ion mode was detected specifically at the surface regions. The signal was identified as an arachidonic acid (AA)-containing phosphatidylinositol (PI), PI(18:0/20:4), by tandem mass spectrometry analysis. Quantitative analysis revealed that the amount of PI(18:0/20:4) in the surface region of CTOSs was two-fold higher than that in the medial region. Finally, PI(18:0/20:4) was enriched at the cancer cells/stromal interface in colorectal cancer patients. These data imply a possible importance of AA-containing PI for colorectal cancer progression, and suggest cells expressing AA-containing PI as potential targets for anti-cancer therapy.
Collapse
Affiliation(s)
- Takanori Hiraide
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Koji Ikegami
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Takanori Sakaguchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Yoshifumi Morita
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Takahiro Hayasaka
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Noritaka Masaki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Michihiko Waki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Eiji Sugiyama
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Satoru Shinriki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Makoto Takeda
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Yasushi Shibasaki
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Shinichiro Miyazaki
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Hirotoshi Kikuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | - Hiroaki Okuyama
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka, Japan
| | - Masahiro Inoue
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,Preeminent Medical Photonics Education &Research Center, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan.,Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR.,Division of Neural Systematics, National Institute for Physiological Sciences, Myodaiji, Okazaki, Japan.,Riken Center for Molecular Imaging Science, Chuo-ku, Kobe, Japan
| | - Hiroyuki Konno
- Second Department of Surgery, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| |
Collapse
|
37
|
Nowinski SM, Solmonson A, Mills EM. Chewing the fat for Akt1 inhibition and oncosuppression. Mol Cell Oncol 2016; 3:e1102795. [PMID: 27308618 DOI: 10.1080/23723556.2015.1102795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 10/22/2022]
Abstract
The catabolic and energy-dissipating actions of mitochondrial uncoupling proteins (UCPs) conflict with many of the bioenergetic hallmarks of malignancy. We have recently demonstrated that overexpression of mitochondrial uncoupling protein 3 (Ucp3) in the basal epidermis impedes skin tumorigenesis through a novel pathway of thymoma viral proto-oncogene 1 (Akt1) inhibition via increased mitochondrial lipid catabolism.
Collapse
Affiliation(s)
- Sara M Nowinski
- Department of Biochemistry, University of Utah School of Medicine , Salt Lake City, UT, USA
| | - Ashley Solmonson
- Division of Pharmacology and Toxicology, College of Pharmacy and; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Edward M Mills
- Division of Pharmacology and Toxicology, College of Pharmacy and; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
38
|
Long chain n-3 polyunsaturated fatty acids increase the efficacy of docetaxel in mammary cancer cells by downregulating Akt and PKCε/δ-induced ERK pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:380-90. [DOI: 10.1016/j.bbalip.2016.01.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/15/2016] [Accepted: 01/22/2016] [Indexed: 12/21/2022]
|
39
|
|
40
|
Inhibition of colon cancer growth by docosahexaenoic acid involves autocrine production of TNFα. Oncogene 2016; 35:4611-22. [PMID: 26853468 DOI: 10.1038/onc.2015.523] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 12/02/2015] [Accepted: 12/11/2015] [Indexed: 12/20/2022]
Abstract
The omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) has anti-inflammatory and anti-cancer properties. Among pro-inflammatory mediators, tumor necrosis factor α (TNFα) plays a paradoxical role in cancer biology with induction of cancer cell death or survival depending on the cellular context. The objective of the study was to evaluate the role of TNFα in DHA-mediated tumor growth inhibition and colon cancer cell death. The treatment of human colorectal cancer cells, HCT-116 and HCT-8 cells, with DHA triggered apoptosis in autocrine TNFα-dependent manner. We demonstrated that DHA-induced increased content of TNFα mRNA occurred through a post-transcriptional regulation via the down-regulation of microRNA-21 (miR-21) expression. Treatment with DHA led to nuclear accumulation of Foxo3a that bounds to the miR-21 promoter triggering its transcriptional repression. Moreover, inhibition of RIP1 kinase and AMP-activated protein kinase α reduced Foxo3a nuclear-cytoplasmic shuttling and subsequent increase of TNFα expression through a decrease of miR-21 expression in DHA-treated colon cancer cells. Finally, we were able to show in HCT-116 xenograft tumor-bearing nude mice that a DHA-enriched diet induced a decrease of human miR-21 expression and an increase of human TNFα mRNA expression limiting tumor growth in a cancer cell-derived TNFα dependent manner. Altogether, the present work highlights a novel mechanism for anti-cancer action of DHA involving colon cancer cell death mediated through autocrine action of TNFα.
Collapse
|
41
|
Omega-3 Fatty Acids and Cancer Cell Cytotoxicity: Implications for Multi-Targeted Cancer Therapy. J Clin Med 2016; 5:jcm5020015. [PMID: 26821053 PMCID: PMC4773771 DOI: 10.3390/jcm5020015] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/24/2022] Open
Abstract
Cancer is a major disease worldwide. Despite progress in cancer therapy, conventional cytotoxic therapies lead to unsatisfactory long-term survival, mainly related to development of drug resistance by tumor cells and toxicity towards normal cells. n-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), can exert anti-neoplastic activity by inducing apoptotic cell death in human cancer cells either alone or in combination with conventional therapies. Indeed, n-3 PUFAs potentially increase the sensitivity of tumor cells to conventional therapies, possibly improving their efficacy especially against cancers resistant to treatment. Moreover, in contrast to traditional therapies, n-3 PUFAs appear to cause selective cytotoxicity towards cancer cells with little or no toxicity on normal cells. This review focuses on studies investigating the cytotoxic activity of n-3 PUFAs against cancer cells via apoptosis, analyzing the molecular mechanisms underlying this effective and selective activity. Here, we highlight the multiple molecules potentially targeted by n-3 PUFAs to trigger cancer cell apoptosis. This analysis can allow a better comprehension of the potential cytotoxic therapeutic role of n-3 PUFAs against cancer, providing specific information and support to design future pre-clinical and clinical studies for a better use of n-3 PUFAs in cancer therapy, mainly combinational therapy.
Collapse
|
42
|
n-3 polyunsaturated fatty acids suppress CD4(+) T cell proliferation by altering phosphatidylinositol-(4,5)-bisphosphate [PI(4,5)P2] organization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:85-96. [PMID: 26476105 DOI: 10.1016/j.bbamem.2015.10.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023]
Abstract
The mechanisms by which n-3 polyunsaturated fatty acids (n-3 PUFA), abundant in fish oil, exert their anti-inflammatory effects have not been rigorously defined. We have previously demonstrated that n-3 PUFA decrease the amount of phosphatidylinositol-(4,5)-bisphosphate, [PI(4,5)P2], in CD4(+) T cells, leading to suppressed actin remodeling upon activation. Since discrete pools of PI(4,5)P2 exist in the plasma membrane, we determined whether n-3 PUFA modulate spatial organization of PI(4,5)P2 relative to raft and non-raft domains. We used Förster resonance energy transfer (FRET) to demonstrate that lipid raft mesodomains in the plasma membrane of CD4(+) T cells enriched in n-3 PUFA display increased co-clustering of Lck(N10) and LAT(ΔCP), markers of lipid rafts. CD4(+) T cells enriched in n-3 PUFA also exhibited a depleted plasma membrane non-raft PI(4,5)P2 pool as detected by decreased co-clustering of Src(N15), a non-raft marker, and PH(PLC-δ), a PI(4,5)P2 reporter. Incubation with exogenous PI(4,5)P2 rescued the effects on the non-raft PI(4,5)P2 pool, and reversed the suppression of T cell proliferation in CD4(+) T cells enriched with n-3 PUFA. Furthermore, CD4(+) T cells isolated from mice fed a 4% docosahexaenoic acid (DHA)-enriched diet exhibited a decrease in the non-raft pool of PI(4,5)P2, and exogenous PI(4,5)P2 reversed the suppression of T cell proliferation. Finally, these effects were not due to changes to post-translational lipidation, since n-3 PUFA did not alter the palmitoylation status of signaling proteins. These data demonstrate that n-3 PUFA suppress T cell proliferation by altering plasma membrane topography and the spatial organization of PI(4,5)P2.
Collapse
|
43
|
Mitochondrial uncoupling links lipid catabolism to Akt inhibition and resistance to tumorigenesis. Nat Commun 2015; 6:8137. [PMID: 26310111 PMCID: PMC4552083 DOI: 10.1038/ncomms9137] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 07/22/2015] [Indexed: 12/12/2022] Open
Abstract
To support growth, tumour cells reprogramme their metabolism to simultaneously upregulate macromolecular biosynthesis while maintaining energy production. Uncoupling proteins (UCPs) oppose this phenotype by inducing futile mitochondrial respiration that is uncoupled from ATP synthesis, resulting in nutrient wasting. Here using a UCP3 transgene targeted to the basal epidermis, we show that forced mitochondrial uncoupling inhibits skin carcinogenesis by blocking Akt activation. Similarly, Akt activation is markedly inhibited in UCP3 overexpressing primary human keratinocytes. Mechanistic studies reveal that uncoupling increases fatty acid oxidation and membrane phospholipid catabolism, and impairs recruitment of Akt to the plasma membrane. Overexpression of Akt overcomes metabolic regulation by UCP3, rescuing carcinogenesis. These findings demonstrate that mitochondrial uncoupling is an effective strategy to limit proliferation and tumorigenesis through inhibition of Akt, and illuminate a novel mechanism of crosstalk between mitochondrial metabolism and growth signalling.
Collapse
|
44
|
Gu Z, Shan K, Chen H, Chen YQ. n-3 Polyunsaturated Fatty Acids and their Role in Cancer Chemoprevention. ACTA ACUST UNITED AC 2015; 1:283-294. [PMID: 26457243 DOI: 10.1007/s40495-015-0043-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polyunsaturated fatty acids (PUFAs), including omega-3 (n-3) and omega-6 (n-6) PUFAs, are essential for human health. Recent research shows n-3 PUFAs and their mediators can inhibit inflammation, angiogenesis and cancer via multiple mechanisms, including reduced release of n-6 fatty acid arachidonic acid from cell membranes, inhibition of enzymatic activities, and direct competition with arachidonic acid for enzymatic conversions. In this review, we discuss inflammation-related cancer, anti-inflammatory effects of n-3 PUFA lipid mediators, antineoplastic activities of n-3 PUFA in vitro and in vivo, and present an update on recent human trials.
Collapse
Affiliation(s)
- Zhennan Gu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China ; The Synergistic Innovation Center for Food Safety and Nutrition, Wuxi 214122, P.R. China ; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Kai Shan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China ; The Synergistic Innovation Center for Food Safety and Nutrition, Wuxi 214122, P.R. China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China ; The Synergistic Innovation Center for Food Safety and Nutrition, Wuxi 214122, P.R. China
| | - Yong Q Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P.R. China ; The Synergistic Innovation Center for Food Safety and Nutrition, Wuxi 214122, P.R. China ; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
45
|
Liu Z, Hopkins MM, Zhang Z, Quisenberry CB, Fix LC, Galvan BM, Meier KE. Omega-3 fatty acids and other FFA4 agonists inhibit growth factor signaling in human prostate cancer cells. J Pharmacol Exp Ther 2014; 352:380-94. [PMID: 25491146 DOI: 10.1124/jpet.114.218974] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Omega-3 fatty acids (n-3 FAs) are proposed to have many beneficial effects on human health. However, the mechanisms underlying their potential cancer preventative effects are unclear. G protein-coupled receptors (GPCRs) of the free fatty acid receptor (FFAR) family, FFA1/GPR40 and FFA4/GPR120, specifically bind n-3 FAs as agonist ligands. In this study, we examined the effects of n-3 FAs in human prostate cancer cell lines. Initial studies established that the long-chain n-3 FAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid, inhibit proliferation of DU145 cells in response to lysophosphatidic acid (LPA), a mitogenic lipid mediator. When added alone to serum-starved DU145 cells, EPA transiently activates signaling events, including p70S6K phosphorylation. However, when added 15 minutes prior to LPA, EPA suppresses LPA-induced activating phosphorylations of ERK, FAK, and p70S6K, and expression of the matricellular protein CCN1. The rapid onset of the inhibitory action of EPA suggested involvement of a GPCR. Further studies showed that DU145 and PC-3 cells express mRNA and protein for both FFA4 and FFA1. TUG-891 (4-[(4-fluoro-4'-methyl[1,1'-biphenyl]-2-yl)methoxy]-benzenepropanoic acid), a selective agonist for FFA4, exerts inhibitory effects on LPA- and epidermal growth factor-induced proliferation and migration, similar to EPA, in DU145 and PC-3 cells. The effects of TUG-891 and EPA are readily reversible. The FFA1/FFA4 agonist GW9508 (4-[[(3-phenoxyphenyl)methyl]amino]-benzenepropranoic acid) likewise inhibits proliferation at doses that block FFA4. Knockdown of FFA4 expression prevents EPA- and TUG-891-induced inhibition of growth and migration. Together, these results indicate that activation of FFA4 initiates signaling events that can inhibit growth factor-induced signaling, providing a novel mechanism for suppression of cancer cell proliferation.
Collapse
Affiliation(s)
- Ze Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Mandi M Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Zhihong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Chrystal B Quisenberry
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Louise C Fix
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Brianna M Galvan
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Kathryn E Meier
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| |
Collapse
|
46
|
Guéguinou M, Gambade A, Félix R, Chantôme A, Fourbon Y, Bougnoux P, Weber G, Potier-Cartereau M, Vandier C. Lipid rafts, KCa/ClCa/Ca2+ channel complexes and EGFR signaling: Novel targets to reduce tumor development by lipids? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2603-20. [PMID: 25450343 DOI: 10.1016/j.bbamem.2014.10.036] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/15/2014] [Accepted: 10/22/2014] [Indexed: 12/29/2022]
Abstract
Membrane lipid rafts are distinct plasma membrane nanodomains that are enriched with cholesterol, sphingolipids and gangliosides, with occasional presence of saturated fatty acids and phospholipids containing saturated acyl chains. It is well known that they organize receptors (such as Epithelial Growth Factor Receptor), ion channels and their downstream acting molecules to regulate intracellular signaling pathways. Among them are Ca2+ signaling pathways, which are modified in tumor cells and inhibited upon membrane raft disruption. In addition to protein components, lipids from rafts also contribute to the organization and function of Ca2+ signaling microdomains. This article aims to focus on the lipid raft KCa/ClCa/Ca2+ channel complexes that regulate Ca2+ and EGFR signaling in cancer cells, and discusses the potential modification of these complexes by lipids as a novel therapeutic approach in tumor development. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Maxime Guéguinou
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Audrey Gambade
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Romain Félix
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Aurélie Chantôme
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Yann Fourbon
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Philippe Bougnoux
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France; Centre HS Kaplan, CHRU Tours, Tours F-37032, France
| | - Günther Weber
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Marie Potier-Cartereau
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France
| | - Christophe Vandier
- Inserm, UMR1069, Nutrition, Croissance et Cancer, Tours F-37032, France; Université François Rabelais, Tours F-37032, France.
| |
Collapse
|
47
|
Essential role of PH domain and leucine-rich repeat protein phosphatase 2 in Nrf2 suppression via modulation of Akt/GSK3β/Fyn kinase axis during oxidative hepatocellular toxicity. Cell Death Dis 2014; 5:e1153. [PMID: 24675471 PMCID: PMC3973230 DOI: 10.1038/cddis.2014.118] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/21/2014] [Accepted: 02/21/2014] [Indexed: 12/30/2022]
Abstract
Instances of sustained oxidative activity have been shown to involve dysregulation of Nrf2-mediated transcriptional induction; however, mechanisms warranting Nrf2-repression remain unclear. In this study, using primary rat hepatocytes, we have attempted to identify factors that may negatively influence Nrf2 survival pathway. Though studies indicate a conspicuous association between Akt and Nrf2, a confirmatory link between the two is unaddressed. On inhibiting PI3K/Akt pathway, we observed compromised activities of antioxidant and detoxification enzymes culminating in oxidative cytotoxicity. This was accompanied by reduced nuclear retention of Nrf2 and its ARE binding affinity, increased Nrf2 ubiquitination and concurrent decline in its downstream targets. Moreover, Akt inhibition enhanced nuclear translocation as well as phosphorylation of Fyn kinase, an enzyme linked to Nrf2 degradation, by relieving GSK3β from phosphorylation-mediated repression. The involvement of Akt and Fyn kinase in influencing Nrf2 signaling was further confirmed in oxidatively stressed hepatocytes by using tert-butyl hydroperoxide (tBHP). tBHP-induced decrease in Nrf2 levels was associated with enhanced Fyn kinase phosphorylation, Fyn kinase nuclear translocation and decreased levels of phosphorylated GSK3β(Ser9) in a time-dependent manner. Interestingly, tBHP induced site-specific deactivation of Akt as only Akt(Ser473) phosphorylation was observed to be affected. Further, protein expression as well as nuclear localization of PHLPP2, a phosphatase specific for Akt(Ser473), was found to be significantly enhanced in tBHP-stressed hepatocytes. Silencing of PHLPP2 not only resulted in considerable restoration of Nrf2 signaling, enhanced Nrf2-ARE binding and reduced Nrf2 ubiquitination but also significantly suppressed tBHP-induced ROS generation and alterations in mitochondrial permeability. We infer that cellular PHLPP2 levels may aggravate oxidative toxicity by suppressing Nrf2/ARE transcriptional regulation via Akt(Se473)/GSK3β/Fyn kinase axis. The study indicates that PHLPP2 could serve as a new target for developing strategies to manage pathological conditions exacerbated due to oxidative stress.
Collapse
|
48
|
Khan S, Abu Jawdeh BG, Goel M, Schilling WP, Parker MD, Puchowicz MA, Yadav SP, Harris RC, El-Meanawy A, Hoshi M, Shinlapawittayatorn K, Deschênes I, Ficker E, Schelling JR. Lipotoxic disruption of NHE1 interaction with PI(4,5)P2 expedites proximal tubule apoptosis. J Clin Invest 2014; 124:1057-68. [PMID: 24531551 DOI: 10.1172/jci71863] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/11/2013] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease progression can be predicted based on the degree of tubular atrophy, which is the result of proximal tubule apoptosis. The Na+/H+ exchanger NHE1 regulates proximal tubule cell survival through interaction with phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2], but pathophysiologic triggers for NHE1 inactivation are unknown. Because glomerular injury permits proximal tubule luminal exposure and reabsorption of fatty acid/albumin complexes, we hypothesized that accumulation of amphipathic, long-chain acyl-CoA (LC-CoA) metabolites stimulates lipoapoptosis by competing with the structurally similar PI(4,5)P2 for NHE1 binding. Kidneys from mouse models of progressive, albuminuric kidney disease exhibited increased fatty acids, LC-CoAs, and caspase-2-dependent proximal tubule lipoapoptosis. LC-CoAs and the cytosolic domain of NHE1 directly interacted, with an affinity comparable to that of the PI(4,5)P2-NHE1 interaction, and competing LC-CoAs disrupted binding of the NHE1 cytosolic tail to PI(4,5)P2. Inhibition of LC-CoA catabolism reduced NHE1 activity and enhanced apoptosis, whereas inhibition of proximal tubule LC-CoA generation preserved NHE1 activity and protected against apoptosis. Our data indicate that albuminuria/lipiduria enhances lipotoxin delivery to the proximal tubule and accumulation of LC-CoAs contributes to tubular atrophy by severing the NHE1-PI(4,5)P2 interaction, thereby lowering the apoptotic threshold. Furthermore, these data suggest that NHE1 functions as a metabolic sensor for lipotoxicity.
Collapse
|
49
|
Yao QH, Zhang XC, Fu T, Gu JZ, Wang L, Wang Y, Lai YB, Wang YQ, Guo Y. ω-3 polyunsaturated fatty acids inhibit the proliferation of the lung adenocarcinoma cell line A549 in vitro. Mol Med Rep 2013; 9:401-6. [PMID: 24276408 DOI: 10.3892/mmr.2013.1829] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/11/2013] [Indexed: 12/24/2022] Open
Abstract
ω-3 polyunsaturated fatty acids (n-3 PUFA), in particular the marine-derived forms eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have been demonstrated to affect cancer cell replication, the cell cycle and cell death. Epidemiological studies have also suggested diets rich in n-3 PUFA were inversely correlated with the development of cancer. In the present study, we explored the effects of DHA and EPA on the proliferation activity and apoptosis of the human lung adenocarcinoma cell line A549. A methyl thiazolyl tetrazolium (MTT) assay was used to detect cell proliferation, apoptosis was detected by flow cytometry and morphological analysis was determined by fluorescence microscopy and transmission electron microscopy. A549 cells were treated with different doses of DHA (40, 45, 50 and 55 µg/ml) or EPA (45, 50, 55 and 60 µg/ml) for 24, 48 and 72 h. The results demonstrated that DHA and EPA significantly suppressed the proliferation of A549 cells and induced apoptosis of A549 cells in a dose- and time-dependent manner. The apoptotic phenomenon was also confirmed by fluorescence microscopy and transmission electron microscopy. Furthermore, compared with the control, the formation of autophagosomes was clearly enhanced in DHA‑ or EPA-treated cells. In conclusion, DHA and EPA inhibited the proliferation of A549 cells and induced cell apoptosis and autophagy, which may provide new safe and effective options for the treatment of lung cancer in the future.
Collapse
Affiliation(s)
- Qing-Hua Yao
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiao-Chen Zhang
- Department of Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ting Fu
- Department of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jian-Zhong Gu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Lu Wang
- Department of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yun Wang
- Department of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yue-Biao Lai
- Department of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yu-Qi Wang
- Department of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yong Guo
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
50
|
Mechanisms of omega-3 polyunsaturated fatty acids in prostate cancer prevention. BIOMED RESEARCH INTERNATIONAL 2013; 2013:824563. [PMID: 23762859 PMCID: PMC3676993 DOI: 10.1155/2013/824563] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/02/2013] [Accepted: 05/08/2013] [Indexed: 12/22/2022]
Abstract
This review focuses on several key areas where progress has been made recently to highlight the role of omega-3 polyunsaturated fatty acid in prostate cancer prevention.
Collapse
|