1
|
Nakamoto H, Shichi S, Shirakawa C, Suzuki T, Kitamura H, Taketomi A. Diacylglycerol kinase alpha regulates post-hepatectomy liver regeneration. Sci Rep 2025; 15:555. [PMID: 39747625 DOI: 10.1038/s41598-024-84403-2] [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: 08/27/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
Diacylglycerol kinases (DGKs) phosphorylate diacylglycerol to generate phosphatidic acid, which plays important roles in intracellular signal transduction. DGKα is reportedly associated with progression of tumors, including hepatocellular carcinomas, but its relationship with liver regeneration has not been examined. The purpose of this research is to elucidate the role of DGKα in liver regeneration. Here, we provide a detailed examination of C57BL/6 wild-type and DGKα knockout (KO) mice subjected to 70% partial hepatectomy (70% PH) modeling, including survival rates, hematological marker and gene expression levels, and histological analyses of factors related to liver regeneration. Following 70% PH, DGKα KO mice produce higher levels of hepatobiliary enzymes and have a higher incidence of jaundice compared with wild-type mice, with a death rate of ~ 40%. Furthermore, they exhibit impaired glycogen and lipid consumption, low liver energy charge, and hepatocyte hypertrophy disorder, accompanied by significantly reduced liver expression of proliferating cell nuclear antigen and cyclin D. We conclude that DGKα is a key molecule in the post-PH liver regeneration process and may have potential as a therapeutic target for the acceleration of liver regeneration.
Collapse
Affiliation(s)
- Hiroki Nakamoto
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Shunsuke Shichi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Chisato Shirakawa
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Takuto Suzuki
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Hidemitsu Kitamura
- Department of Biomedical Engineering, Faculty of Life Sciences, Toyo University, Saitama, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
2
|
Manouchehri JM, Datta J, Marcho LM, Reardon JJ, Stover D, Wesolowski R, Borate U, Cheng TYD, Schnell PM, Ramaswamy B, Sizemore GM, Rubinstein MP, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. Breast Cancer Res 2024; 26:153. [PMID: 39506780 PMCID: PMC11539583 DOI: 10.1186/s13058-024-01906-6] [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: 04/17/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Breast cancer, one of the most common forms of cancer, is associated with the highest cancer-related mortality among women worldwide. In comparison to other types of breast cancer, patients diagnosed with the triple-negative breast cancer (TNBC) subtype have the worst outcome because current therapies do not produce long-lasting responses. Hence, innovative therapies that produce persisting responses are a critical need. We previously discovered that hyperactivating purinergic receptors (P2RXs) by increasing extracellular adenosine triphosphate (eATP) concentrations enhances TNBC cell lines' response to chemotherapy. Heparan sulfate inhibits multiple extracellular ATPases, so it is a molecule of interest in this regard. In turn, heparanase degrades polysulfated polysaccharide heparan sulfate. Importantly, previous work suggests that breast cancer and other cancers express heparanase at high levels. Hence, as heparan sulfate can inhibit extracellular ATPases to facilitate eATP accumulation, it may intensify responses to chemotherapy. We postulated that heparanase inhibitors would exacerbate chemotherapy-induced decreases in TNBC cell viability by increasing heparan sulfate in the cellular microenvironment and hence, augmenting eATP. METHODS We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with paclitaxel (cytotoxic chemotherapeutic) with or without the heparanase inhibitor OGT 2115 and/or supplemental heparan sulfate. We evaluated cell viability and the release of eATP. Also, we compared the expression of heparanase protein in cell lines and tissues by immunoblot and immunohistochemistry, respectively. In addition, we examined breast-cancer-initiating cell populations using tumorsphere formation efficiency assays on treated cells. RESULTS We found that combining heparanase inhibitor OGT 2115 with chemotherapy decreased TNBC cell viability and tumorsphere formation through increases in eATP and activation of purinergic receptors as compared to TNBC cells treated with single-agent paclitaxel. CONCLUSION Our data shows that by preventing heparan sulfate breakdown, heparanase inhibitors make TNBC cells more susceptible to chemotherapy by enhancing eATP concentrations.
Collapse
Affiliation(s)
- Jasmine M Manouchehri
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jharna Datta
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Lynn M Marcho
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jesse J Reardon
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Uma Borate
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Ting-Yuan David Cheng
- Division of Cancer Prevention and Control, Department of Internal Medicine, The Ohio State University, Suite 525, 1590 North High St., Columbus, OH, 43201, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, 1841 Neil Ave., Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mark P Rubinstein
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mathew A Cherian
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA.
- Division of Medical Oncology, 460 W 12th Ave., 888 BRT, Columbus, OH, 43210, USA.
| |
Collapse
|
3
|
Wu Y, Gong Y, Liu Y, Chen F, Chen S, Zhang F, Wang C, Li S, Hu M, Huang R, Guo X, Wang X, Ning Y, Yang L. Comparative Analysis of Differentially Expressed Genes in Chondrocytes from Rats Exposed to Low Selenium and T-2 Toxin. Biol Trace Elem Res 2024; 202:1020-1030. [PMID: 37326932 DOI: 10.1007/s12011-023-03725-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
The aim of this study was to construct rat models of environmental risk factors for Kashin-Beck disease (KBD) with low selenium and T-2 toxin levels and to screen the differentially expressed genes (DEGs) between the rat models exposed to environmental risk factors. The Se-deficient (SD) group and T-2 toxin exposure (T-2) group were constructed. Knee joint samples were stained with hematoxylin-eosin, and cartilage tissue damage was observed. Illumina high-throughput sequencing technology was used to detect the gene expression profiles of the rat models in each group. Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway enrichment analysis were performed and five differential gene expression results were verified by quantitative real-time polymerase chain reaction (qRT‒PCR). A total of 124 DEGs were identified from the SD group, including 56 upregulated genes and 68 downregulated genes. A total of 135 DEGs were identified in the T-2 group, including 68 upregulated genes and 67 downregulated genes. The DEGs were significantly enriched in 4 KEGG pathways in the SD group and 9 KEGG pathways in the T-2 group. The expression levels of Dbp, Pc, Selenow, Rpl30, and Mt2A were consistent with the results of transcriptome sequencing by qRT‒PCR. The results of this study confirmed that there were some differences in DEGs between the SD group and the T-2 group and provided new evidence for further exploration of the etiology and pathogenesis of KBD.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yi Gong
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yanli Liu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Feihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Sijie Chen
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Feiyu Zhang
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Chaowei Wang
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Shujin Li
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Minhan Hu
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Ruitian Huang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xiong Guo
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
- Clinical Research Center for Endemic Disease of Shaanxi Province, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 Xi Wu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Xi Wang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China.
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China.
| | - Yujie Ning
- School of Public Health, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
| | - Lei Yang
- School of Nursing, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
4
|
Martin-Salgado M, Ochoa-Echeverría A, Mérida I. Diacylglycerol kinases: A look into the future of immunotherapy. Adv Biol Regul 2024; 91:100999. [PMID: 37949728 DOI: 10.1016/j.jbior.2023.100999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Cancer still represents the second leading cause of death right after cardiovascular diseases. According to the World Health Organization (WHO), cancer provoked around 10 million deaths in 2020, with lung and colon tumors accounting for the deadliest forms of cancer. As tumor cells become resistant to traditional therapeutic approaches, immunotherapy has emerged as a novel strategy for tumor control. T lymphocytes are key players in immune responses against tumors. Immunosurveillance allows identification, targeting and later killing of cancerous cells. Nevertheless, tumors evolve through different strategies to evade the immune response and spread in a process called metastasis. The ineffectiveness of traditional strategies to control tumor growth and expansion has led to novel approaches considering modulation of T cell activation and effector functions. Program death receptor 1 (PD-1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) showed promising results in the early 90s and nowadays are still being exploited together with other drugs for several cancer types. Other negative regulators of T cell activation are diacylglycerol kinases (DGKs) a family of enzymes that catalyze the conversion of diacylglycerol (DAG) into phosphatidic acid (PA). In T cells, DGKα and DGKζ limit the PLCγ/Ras/ERK axis thus attenuating DAG mediated signaling and T cell effector functions. Upregulation of either of both isoforms results in impaired Ras activation and anergy induction, whereas germline knockdown mice showed enhanced antitumor properties and more effective immune responses against pathogens. Here we review the mechanisms used by DGKs to ameliorate T cell activation and how inhibition could be used to reinvigorate T cell functions in cancer context. A better knowledge of the molecular mechanisms involved upon T cell activation will help to improve current therapies with DAG promoting agents.
Collapse
Affiliation(s)
- Miguel Martin-Salgado
- Department of Immunology and Oncology. National Centre for Biotechnology. Spanish Research Council (CNB-CSIC), Spain
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology. National Centre for Biotechnology. Spanish Research Council (CNB-CSIC), Spain
| | - Isabel Mérida
- Department of Immunology and Oncology. National Centre for Biotechnology. Spanish Research Council (CNB-CSIC), Spain.
| |
Collapse
|
5
|
Manouchehri JM, Marcho L, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556819. [PMID: 37745355 PMCID: PMC10515779 DOI: 10.1101/2023.09.08.556819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Background Among women worldwide, breast cancer has the highest incidence and is the leading cause of cancer-related death. Patients with the triple-negative breast cancer (TNBC) subtype have an inferior prognosis in comparison to other breast cancers because current therapies do not facilitate long-lasting responses. Thus, there is a demand for more innovative therapies that induce durable responses.In our previous research, we discovered that augmenting the concentration of extracellular ATP (eATP) greatly enhances the chemotherapeutic response of TNBC cell lines by activating purinergic receptors (P2RXs), leading to cell death through the induction of non-selective membrane permeability. However, eATP levels are limited by several classes of extracellular ATPases. One endogenous molecule of interest that can inhibit multiple classes of extracellular ATPases is heparan sulfate. Polysulfated polysaccharide heparan sulfate itself is degraded by heparanase, an enzyme that is known to be highly expressed in various cancers, including breast cancer. Heparan sulfate has previously been shown to regulate several cancer-related processes such as fibroblast growth factor signaling, neoangiogenesis by sequestering vascular endothelial growth factors in the extracellular matrix, hedgehog signaling and cell adhesion. In this project, we identified an additional mechanism for a tumor suppressor role of heparan sulfate: inhibition of extracellular ATPases, leading to augmented levels of eATP.Several heparanase inhibitors have been previously identified, including OGT 2115, suramin, PI-88, and PG 545. We hypothesized that heparanase inhibitors would augment eATP concentrations in TNBC by increasing heparan sulfate in the tumor microenvironment, resulting in enhanced cell death in response to chemotherapy. Methods We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with increasing concentrations of the chemotherapeutic agent paclitaxel in the presence of heparan sulfate and/or the heparanase inhibitor OGT 2115 while analyzing eATP release and cell viability. Moreover, to verify that the effects of OGT 2115 are mediated through eATP, we applied specific antagonists to the purinergic receptors P2RX4 and P2RX7. In addition, the protein expression of heparanase was compared in the cell lines by Western blot analysis. We also evaluated the consequences of this therapeutic strategy on the breast cancer-initiating cell population in the treated cells using flow cytometry and tumorsphere formation efficiency assays. Results Heparanase was found to be highly expressed in immortal mammary epithelial cells in comparison to TNBC cell lines. The heparanase inhibitor OGT 2115 augmented chemotherapy-induced TNBC cell death and eATP release. Conclusion These results demonstrate that inhibiting the degradation of heparan sulfate in the tumor microenvironment augments the susceptibility of TNBC cell lines to chemotherapy by increasing extracellular ATP concentrations. This strategy could potentially be applied to induce more enhanced and enduring responses in TNBC patients.
Collapse
|
6
|
Mendez R, Shaikh M, Lemke MC, Yuan K, Libby AH, Bai DL, Ross MM, Harris TE, Hsu KL. Predicting small molecule binding pockets on diacylglycerol kinases using chemoproteomics and AlphaFold. RSC Chem Biol 2023; 4:422-430. [PMID: 37292058 PMCID: PMC10246554 DOI: 10.1039/d3cb00057e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/13/2023] [Indexed: 06/10/2023] Open
Abstract
Diacylglycerol kinases (DGKs) are metabolic kinases involved in regulating cellular levels of diacylglycerol and phosphatidic lipid messengers. The development of selective inhibitors for individual DGKs would benefit from discovery of protein pockets available for inhibitor binding in cellular environments. Here we utilized a sulfonyl-triazole probe (TH211) bearing a DGK fragment ligand for covalent binding to tyrosine and lysine sites on DGKs in cells that map to predicted small molecule binding pockets in AlphaFold structures. We apply this chemoproteomics-AlphaFold approach to evaluate probe binding of DGK chimera proteins engineered to exchange regulatory C1 domains between DGK subtypes (DGKα and DGKζ). Specifically, we discovered loss of TH211 binding to a predicted pocket in the catalytic domain when C1 domains on DGKα were exchanged that correlated with impaired biochemical activity as measured by a DAG phosphorylation assay. Collectively, we provide a family-wide assessment of accessible sites for covalent targeting that combined with AlphaFold revealed predicted small molecule binding pockets for guiding future inhibitor development of the DGK superfamily.
Collapse
Affiliation(s)
- Roberto Mendez
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
| | - Minhaj Shaikh
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
| | - Michael C Lemke
- Department of Pharmacology, University of Virginia School of Medicine Charlottesville Virginia 22908 USA
| | - Kun Yuan
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
| | - Adam H Libby
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
- University of Virginia Cancer Center, University of Virginia Charlottesville VA 22903 USA
| | - Dina L Bai
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
| | - Mark M Ross
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia School of Medicine Charlottesville Virginia 22908 USA
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia Charlottesville Virginia 22904 USA +1 434-297-4864
- Department of Pharmacology, University of Virginia School of Medicine Charlottesville Virginia 22908 USA
- Department of Molecular Physiology and Biological Physics, University of Virginia Charlottesville Virginia 22908 USA
- University of Virginia Cancer Center, University of Virginia Charlottesville VA 22903 USA
| |
Collapse
|
7
|
Grunt TW, Heller G. A critical appraisal of the relative contribution of tissue architecture, genetics, epigenetics and cell metabolism to carcinogenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023:S0079-6107(23)00056-1. [PMID: 37268024 DOI: 10.1016/j.pbiomolbio.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023]
Abstract
Here we contrast several carcinogenesis models. The somatic-mutation-theory posits mutations as main causes of malignancy. However, inconsistencies led to alternative explanations. For example, the tissue-organization-field-theory considers disrupted tissue-architecture as main cause. Both models can be reconciled using systems-biology-approaches, according to which tumors hover in states of self-organized criticality between order and chaos, are emergent results of multiple deviations and are subject to general laws of nature: inevitable variation(mutation) explainable by increased entropy(second-law-of-thermodynamics) or indeterminate decoherence upon measurement of superposed quantum systems(quantum mechanics), followed by Darwinian-selection. Genomic expression is regulated by epigenetics. Both systems cooperate. So cancer is neither just a mutational nor an epigenetic problem. Rather, epigenetics links environmental cues to endogenous genetics engendering a regulatory machinery that encompasses specific cancer-metabolic-networks. Interestingly, mutations occur at all levels of this machinery (oncogenes/tumor-suppressors, epigenetic-modifiers, structure-genes, metabolic-genes). Therefore, in most cases, DNA mutations may be the initial and crucial cancer-promoting triggers.
Collapse
Affiliation(s)
- Thomas W Grunt
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090, Vienna, Austria; Comprehensive Cancer Center, 1090, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology, 1090, Vienna, Austria.
| | - Gerwin Heller
- Comprehensive Cancer Center, 1090, Vienna, Austria; Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
8
|
Colacci A, Corvi R, Ohmori K, Paparella M, Serra S, Da Rocha Carrico I, Vasseur P, Jacobs MN. The Cell Transformation Assay: A Historical Assessment of Current Knowledge of Applications in an Integrated Approach to Testing and Assessment for Non-Genotoxic Carcinogens. Int J Mol Sci 2023; 24:ijms24065659. [PMID: 36982734 PMCID: PMC10057754 DOI: 10.3390/ijms24065659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
The history of the development of the cell transformation assays (CTAs) is described, providing an overview of in vitro cell transformation from its origin to the new transcriptomic-based CTAs. Application of this knowledge is utilized to address how the different types of CTAs, variously addressing initiation and promotion, can be included on a mechanistic basis within the integrated approach to testing and assessment (IATA) for non-genotoxic carcinogens. Building upon assay assessments targeting the key events in the IATA, we identify how the different CTA models can appropriately fit, following preceding steps in the IATA. The preceding steps are the prescreening transcriptomic approaches, and assessment within the earlier key events of inflammation, immune disruption, mitotic signaling and cell injury. The CTA models address the later key events of (sustained) proliferation and change in morphology leading to tumor formation. The complementary key biomarkers with respect to the precursor key events and respective CTAs are mapped, providing a structured mechanistic approach to represent the complexity of the (non-genotoxic) carcinogenesis process, and specifically their capacity to identify non-genotoxic carcinogenic chemicals in a human relevant IATA.
Collapse
Affiliation(s)
- Annamaria Colacci
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Via Po 5, I-40139 Bologna, Italy
- Correspondence:
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), I-21027 Ispra, Italy
| | - Kyomi Ohmori
- Chemical Division, Kanagawa Prefectural Institute of Public Health, Chigasaki 253-0087, Japan
- Research Initiatives and Promotion Organization, Yokohama National University, Yokohama 240-8501, Japan
| | - Martin Paparella
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, A-6020 Innbruck, Austria
| | - Stefania Serra
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Via Po 5, I-40139 Bologna, Italy
| | | | - Paule Vasseur
- Universite de Lorraine, CNRS UMR 7360 LIEC, Laboratoire Interdisciplinaire des Environnements Continentaux, 57070 Metz, France
| | - Miriam Naomi Jacobs
- Radiation, Chemical and Environmental Hazards, UK Health Security Agency, Harwell Science and Innovation Campus, Chilton OX11 0RQ, UK
| |
Collapse
|
9
|
Yachida N, Hoshino F, Murakami C, Ebina M, Miura Y, Sakane F. Saturated fatty acid- and/or monounsaturated fatty acid-containing phosphatidic acids selectively interact with heat shock protein 27. J Biol Chem 2023; 299:103019. [PMID: 36791913 PMCID: PMC10023972 DOI: 10.1016/j.jbc.2023.103019] [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: 08/11/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Diacylglycerol kinase (DGK) α, which is a key enzyme in the progression of cancer and, in contrast, in T-cell activity attenuation, preferentially produces saturated fatty acid (SFA)- and/or monounsaturated fatty acid (MUFA)-containing phosphatidic acids (PAs), such as 16:0/16:0-, 16:0/18:0-, and 16:1/16:1-PA, in melanoma cells. In the present study, we searched for the target proteins of 16:0/16:0-PA in melanoma cells and identified heat shock protein (HSP) 27, which acts as a molecular chaperone and contributes to cancer progression. HSP27 more strongly interacted with PA than other phospholipids, including phosphatidylcholine, phosphatidylserine, phosphatidylglycerol, cardiolipin, phosphatidylinositol, phosphatidylinositol 4-monophosphate, and phosphatidylinositol 4,5-bisphosphate. Moreover, HSP27 is more preferentially bound to SFA- and/or MUFA-containing PAs, including 16:0/16:0- and 16:0/18:1-PAs, than PUFA-containing PAs, including 18:0/20:4- and 18:0/22:6-PA. Furthermore, HSP27 and constitutively active DGKα expressed in COS-7 cells colocalized in a DGK activity-dependent manner. Notably, 16:0/16:0-PA, but not phosphatidylcholine or 16:0/16:0-phosphatidylserine, induced oligomer dissociation of HSP27, which enhances its chaperone activity. Intriguingly, HSP27 protein was barely detectable in Jurkat T cells, while the protein band was intensely detected in AKI melanoma cells. Taken together, these results strongly suggest that SFA- and/or MUFA-containing PAs produced by DGKα selectively target HSP27 and regulate its cancer-progressive function in melanoma cells but not in T cells.
Collapse
Affiliation(s)
- Naoto Yachida
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Fumi Hoshino
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Masayuki Ebina
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan.
| |
Collapse
|
10
|
DGKB mediates radioresistance by regulating DGAT1-dependent lipotoxicity in glioblastoma. Cell Rep Med 2023; 4:100880. [PMID: 36603576 PMCID: PMC9873821 DOI: 10.1016/j.xcrm.2022.100880] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/08/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023]
Abstract
Glioblastoma (GBM) currently has a dismal prognosis. GBM cells that survive radiotherapy contribute to tumor progression and recurrence with metabolic advantages. Here, we show that diacylglycerol kinase B (DGKB), a regulator of the intracellular concentration of diacylglycerol (DAG), is significantly downregulated in radioresistant GBM cells. The downregulation of DGKB increases DAG accumulation and decreases fatty acid oxidation, contributing to radioresistance by reducing mitochondrial lipotoxicity. Diacylglycerol acyltransferase 1 (DGAT1), which catalyzes the formation of triglycerides from DAG, is increased after ionizing radiation. Genetic inhibition of DGAT1 using short hairpin RNA (shRNA) or microRNA-3918 (miR-3918) mimic suppresses radioresistance. We discover that cladribine, a clinical drug, activates DGKB, inhibits DGAT1, and sensitizes GBM cells to radiotherapy in vitro and in vivo. Together, our study demonstrates that DGKB downregulation and DGAT1 upregulation confer radioresistance by reducing mitochondrial lipotoxicity and suggests DGKB and DGAT1 as therapeutic targets to overcome GBM radioresistance.
Collapse
|
11
|
Aulakh SS, Bozelli JC, Epand RM. Exploring the AlphaFold Predicted Conformational Properties of Human Diacylglycerol Kinases. J Phys Chem B 2022; 126:7172-7183. [PMID: 36041230 DOI: 10.1021/acs.jpcb.2c04533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diacylglycerol kinases (DGKs) are important enzymes in molecular membrane biology, as they can lower the concentration of diacylglycerol through phosphorylation while at the same time producing phosphatidic acid. Dysfunction of DGK is linked with multiple diseases including cancer and autoimmune disorders. Currently, the high-resolution structures have not been determined for any of the 10 human DGK paralogs, which has made it difficult to gain a more complete understanding of the enzyme's mechanism of action and regulation. In the present study, we have taken advantage of the significant developments in protein structural prediction technology by artificial intelligence (i.e., Alphafold 2.0), to conduct a comprehensive investigation on the properties of all 10 human DGK paralogs. Structural alignment of the predictions reveals that the C1, catalytic, and accessory domains are conserved in their spatial arrangement relative to each other, across all paralogs. This suggests a critical role played by this domain architecture in DGK function. Moreover, docking studies corroborate the existence of a conserved ATP-binding site between the catalytic and accessory domains. Interestingly, the ATP bound to the interdomain cleft was also found to be in proximity of the conserved glycine-rich motif, which in protein kinases has been suggested to function in ATP binding. Lastly, the spatial arrangement of DGK, with respect to the membrane, reveals that most paralogs possess a more energetically favorable interaction with curved membranes. In conclusion, AlphaFold predictions of human DGKs provide novel insights into the enzyme's structural and functional properties while also paving the way for future experimentation.
Collapse
Affiliation(s)
- Sukhvershjit S Aulakh
- Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - José Carlos Bozelli
- Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
12
|
Takahashi D, Yonezawa K, Okizaki Y, Caaveiro JMM, Ueda T, Shimada A, Sakane F, Shimizu N. Ca 2+ -induced structural changes and intramolecular interactions in N-terminal region of diacylglycerol kinase alpha. Protein Sci 2022; 31:e4365. [PMID: 35762720 PMCID: PMC9202544 DOI: 10.1002/pro.4365] [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: 02/03/2022] [Revised: 05/09/2022] [Accepted: 05/27/2022] [Indexed: 11/05/2022]
Abstract
Diacylglycerol kinases (DGKs) are multi-domain lipid kinases that modulate the levels of lipid messengers, diacylglycerol, and phosphatidic acid. Recently, increasing attention has been paid to its α isozyme (DGKα) as a potential target for cancer immunotherapy. However, little progress has been made on the structural biology of DGKs, and a detailed understanding of the Ca2+ -triggered activation of DGKα, for which the N-terminal domains likely play a critical role, remains unclear. We have recently shown that Ca2+ binding to DGKα-EF induces conformational changes from a protease-susceptible "open" conformation in the apo state to a well-folded one in its holo state. Here, we further studied the structural properties of DGKα N-terminal (RVH and EF) domains using a series of biophysical techniques. We first revealed that the N-terminal RVH domain is a novel Ca2+ -binding domain, but the Ca2+ -induced conformational changes mainly occur in the EF domain. This was corroborated by NMR experiments showing that the EF domain adopts a molten-globule like structure in the apo state. Further analyses using SEC-SAXS and NMR indicate that the partially unfolded EF domain interacts with RVH domain, likely via hydrophobic interactions in the absence of Ca2+ , and this interaction is modified in the presence of Ca2+ . Taken together, these results present novel insights into the structural rearrangement of DGKα N-terminal domains upon binding to Ca2+ , which is essential for the activation of the enzyme.
Collapse
Affiliation(s)
- Daisuke Takahashi
- Department of Protein Structure, Function, and Design, Graduate School of Pharmaceutical ScienceKyushu UniversityFukuokaJapan
| | - Kento Yonezawa
- Photon Factory, Institute of Materials Structure ScienceHigh Energy Accelerator Research Organization (KEK)TsukubaJapan
- Center for Digital Green‐Innovation (CDG)Nara Institute of Science and Technology (NAIST)IkomaJapan
| | - Yuki Okizaki
- Department of Protein Structure, Function, and Design, Graduate School of Pharmaceutical ScienceKyushu UniversityFukuokaJapan
| | - Jose M. M. Caaveiro
- Department of Global Healthcare, Graduate School of Pharmaceutical ScienceKyushu UniversityFukuokaJapan
| | - Tadashi Ueda
- Department of Protein Structure, Function, and Design, Graduate School of Pharmaceutical ScienceKyushu UniversityFukuokaJapan
| | - Atsushi Shimada
- Division of Structural Biology, Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of ScienceChiba UniversityChibaJapan
| | - Nobutaka Shimizu
- Photon Factory, Institute of Materials Structure ScienceHigh Energy Accelerator Research Organization (KEK)TsukubaJapan
| |
Collapse
|
13
|
Cooke M, Kazanietz MG. Overarching roles of diacylglycerol signaling in cancer development and antitumor immunity. Sci Signal 2022; 15:eabo0264. [PMID: 35412850 DOI: 10.1126/scisignal.abo0264] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Diacylglycerol (DAG) is a lipid second messenger that is generated in response to extracellular stimuli and channels intracellular signals that affect mammalian cell proliferation, survival, and motility. DAG exerts a myriad of biological functions through protein kinase C (PKC) and other effectors, such as protein kinase D (PKD) isozymes and small GTPase-regulating proteins (such as RasGRPs). Imbalances in the fine-tuned homeostasis between DAG generation by phospholipase C (PLC) enzymes and termination by DAG kinases (DGKs), as well as dysregulation in the activity or abundance of DAG effectors, have been widely associated with tumor initiation, progression, and metastasis. DAG is also a key orchestrator of T cell function and thus plays a major role in tumor immunosurveillance. In addition, DAG pathways shape the tumor ecosystem by arbitrating the complex, dynamic interaction between cancer cells and the immune landscape, hence representing powerful modifiers of immune checkpoint and adoptive T cell-directed immunotherapy. Exploiting the wide spectrum of DAG signals from an integrated perspective could underscore meaningful advances in targeted cancer therapy.
Collapse
Affiliation(s)
- Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Medicine, Einstein Medical Center Philadelphia, Philadelphia, PA 19141, USA
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Okada N, Sugiyama K, Shichi S, Shirai Y, Goto K, Sakane F, Kitamura H, Taketomi A. Combination therapy for hepatocellular carcinoma with diacylglycerol kinase alpha inhibition and anti-programmed cell death-1 ligand blockade. Cancer Immunol Immunother 2022; 71:889-903. [PMID: 34482409 PMCID: PMC10991887 DOI: 10.1007/s00262-021-03041-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/22/2021] [Indexed: 12/13/2022]
Abstract
Activation of diacylglycerol kinase alpha (DGKα) augments proliferation and suppresses apoptosis of cancer cells and induces T lymphocyte anergy. We investigated the dual effects of DGKα inhibition on tumorigenesis and anti-tumor immunity with the aim of establishing a novel therapeutic strategy for cancer. We examined the effects of a DGKα inhibitor (DGKAI) on liver cancer cell proliferation and cytokine production by immune cells in vitro and on tumorigenesis and host immunity in a hepatocellular carcinoma (HCC) mouse model. Oral DGKAI significantly suppressed tumor growth and prolonged survival in model mice. Tumor infiltration of T cells and dendritic cells was also enhanced in mice treated with DGKAI, and the production of cytokines and cytotoxic molecules by CD4+ and CD8+ T cells was increased. Depletion of CD8+ T cells reduced the effect of DGKAI. Furthermore, interferon-γ stimulation augmented the expression of programmed cell death-1 ligand (PD-L1) on cancer cells, and DGKAI plus an anti-PD-L1 antibody strongly suppressed the tumor growth. These results suggest that DGKα inhibition may be a promising new treatment strategy for HCC.
Collapse
Affiliation(s)
- Naoki Okada
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Ko Sugiyama
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shunsuke Shichi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Yasuhito Shirai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Science, Faculty of Agriculture, Kobe University, Kobe, Japan
| | - Kaoru Goto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Yamagata, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Hidemitsu Kitamura
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, N15 W7 Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
15
|
Pakiet A, Sikora K, Kobiela J, Rostkowska O, Mika A, Sledzinski T. Alterations in complex lipids in tumor tissue of patients with colorectal cancer. Lipids Health Dis 2021; 20:85. [PMID: 34348720 PMCID: PMC8340484 DOI: 10.1186/s12944-021-01512-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022] Open
Abstract
Background Accumulating evidence indicates alterations in lipid metabolism and lipid composition in neoplastic tissue. Earlier nuclear magnetic resonance studies showed that the contents of major lipid groups, such as triacylglycerols, phospholipids and cholesterol, are changed in colon cancer tissue. Methods In this study, a more detailed analysis of lipids in cancer and tumor adjacent tissues from colorectal cancer patients, using liquid chromatography–mass spectrometry, allowed for comparison of 199 different lipids between cancer tissue and tumor adjacent tissue using principal component analysis. Results Significant differences were found in 67 lipid compounds between the two types of tissue; many of these lipid compounds are bioactive lipids such as ceramides, lysophospholipids or sterols and can influence the development of cancer. Additionally, increased levels of phospholipids and sphingolipids were present, which are major components of the cell membrane, and increases in these lipids can lead to changes in cell membrane properties. Conclusions This study showed that many complex lipids are significantly increased or decreased in colon cancer tissue, reflecting significant alterations in lipid metabolism. This knowledge can be used for the selection of potential molecular targets of novel anticancer strategies based on the modulation of lipid metabolism and the composition of the cell membrane in colorectal cancer cells. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01512-x.
Collapse
Affiliation(s)
- Alicja Pakiet
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Kinga Sikora
- Physics-Chemistry Workshops, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Jarek Kobiela
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Smoluchowskiego 17, 80-214, Gdansk, Poland
| | - Olga Rostkowska
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, Smoluchowskiego 17, 80-214, Gdansk, Poland
| | - Adriana Mika
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
16
|
Bozelli JC, Yune J, Takahashi D, Sakane F, Epand RM. Membrane morphology determines diacylglycerol kinase α substrate acyl chain specificity. FASEB J 2021; 35:e21602. [PMID: 33977628 DOI: 10.1096/fj.202100264r] [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: 02/10/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023]
Abstract
Diacylglycerol kinases catalyze the ATP-dependent phosphorylation of diacylglycerol (DAG) to produce phosphatidic acid (PA). In humans, the alpha isoform (DGKα) has emerged as a potential target in the treatment of cancer due to its anti-tumor and pro-immune responses. However, its mechanism of action at a molecular level is not fully understood. In this work, a systematic investigation of the role played by the membrane in the regulation of the enzymatic properties of human DGKα is presented. By using a cell-free system with purified DGKα and model membranes of variable physical and chemical properties, it is shown that membrane physical properties determine human DGKα substrate acyl chain specificity. In model membranes with a flat morphology; DGKα presents high enzymatic activity, but it is not able to differentiate DAG molecular species. Furthermore, DGKα enzymatic properties are insensitive to membrane intrinsic curvature. However, in the presence of model membranes with altered morphology, specifically the presence of physically curved membrane structures, DGKα bears substrate acyl chain specificity for palmitic acid-containing DAG. The present results identify changes in membrane morphology as one possible mechanism for the depletion of specific pools of DAG as well as the production of specific pools of PA by DGKα, adding an extra layer of regulation on the interconversion of these two potent lipid-signaling molecules. It is proposed that the interplay between membrane physical (shape) and chemical (lipid composition) properties guarantee a fine-tuned signal transduction system dependent on the levels and molecular species of DAG and PA.
Collapse
Affiliation(s)
- José Carlos Bozelli
- Department of Biochemistry and Biomedical Sciences, McMaster University, Health Sciences Centre, Hamilton, ON, Canada
| | - Jenny Yune
- Department of Biochemistry and Biomedical Sciences, McMaster University, Health Sciences Centre, Hamilton, ON, Canada
| | - Daisuke Takahashi
- Department of Pharmaceutical Health Care and Sciences, Kyushu University, Fukuoka, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Health Sciences Centre, Hamilton, ON, Canada
| |
Collapse
|
17
|
Abstract
Viral infections are a major health problem; therefore, there is an urgent need for novel therapeutic strategies. Antivirals used to target proteins encoded by the viral genome usually enhance drug resistance generated by the virus. A potential solution may be drugs acting at host-based targets since viruses are dependent on numerous cellular proteins and phosphorylation events that are crucial during their life cycle. Repurposing existing kinase inhibitors as antiviral agents would help in the cost and effectiveness of the process, but this strategy usually does not provide much improvement, and specific medicinal chemistry programs are needed in the field. Anyway, extensive use of FDA-approved kinase inhibitors has been quite useful in deciphering the role of host kinases in viral infection. The present perspective aims to review the state of the art of kinase inhibitors that target viral infections in different development stages.
Collapse
Affiliation(s)
- Javier García-Cárceles
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Elena Caballero
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
18
|
Liu K, Xue B, Bai G, Zhang W. Downregulation of Diacylglycerol kinase zeta (DGKZ) suppresses tumorigenesis and progression of cervical cancer by facilitating cell apoptosis and cell cycle arrest. Bioengineered 2021; 12:1517-1529. [PMID: 33926342 PMCID: PMC8806244 DOI: 10.1080/21655979.2021.1918505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Diacylglycerol kinase zeta (DGKZ) participates in cancer progression. Here, the current work aims to identify the functional role of DGKZ in cervical cancer (CC). DGKZ expression in cervical cancer tissues and paired adjacent normal cervical tissues was assessed using Immunohistochemistry assay. SiHa and HeLa cells were transfected with lentivirus plasmids (sh-DGKZ or sh-NC) to evaluate the effects of DGKZ knockdown on cell proliferation, apoptosis and cell cycle distribution in vitro. Furthermore, BALB/c nude mice were injected subcutaneously with Lentivirus-sh-DGKZ-SiHa cells or Lentivirus-sh-NC-SiHa cells to analyze the influence of DGKZ silencing on tumor growth of CC in vivo. Moreover, the potential molecular mechanisms were predicted by GO and KEGG analysis and preliminarily explored through PathScan Analysis. Elevated DGKZ expression in cervical tumor was observed. Downregulation of DGKZ repressed proliferation and boosted apoptosis of SiHa and HeLa cells and induced cell cycle arrest at G0/G1 phase. In addition, Knockdown of DGKZ restrained tumor growth in tumor xenograft mice. Importantly, GO and KEGG analysis displayed that differentially expressed proteins induced by silence of DGKZ were mostly enriched in autophagy or mitophagy, indicating that the functions of DGKZ on cell proliferation and tumor growth may be associated with autophagy or mitophagy. PathScan analysis presented that PI3K-AKT and TAK1-NF-κB signaling pathways were prominently inhibited in SiHa cells transfected with sh-DGKZ. In summary, downregulation of DGKZ impeded cell proliferation, boosted cell apoptosis and induced cell cycle arrest to suppress tumorigenesis and progression of cervical cancer.
Collapse
Affiliation(s)
- Keying Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.,Department of Gynecology and Obstetrics, Xi'an North Hospital, Xi'an, Shaanxi Province, China
| | - Biyun Xue
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Guiqin Bai
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Wentao Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
19
|
Asami M, Suzuki Y, Sakane F. Dopamine and the phosphorylated dopamine transporter are increased in the diacylglycerol kinase η-knockout mouse brain. FEBS Lett 2021; 595:1313-1321. [PMID: 33599293 DOI: 10.1002/1873-3468.14059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms generating the mania-like abnormal behaviors caused by diacylglycerol (DG) kinase (DGK) η deficiency remain unclear. Here, we found that DGKη knockout markedly increased dopamine (DA) levels in the midbrain (DA-producing region, 2.8-fold) and cerebral cortex (DA projection region, 1.2-fold). Moreover, DGKη deficiency significantly augmented phosphorylated DA transporter (DAT) levels (1.4-fold increase), which induce DA efflux to the synaptic cleft, in the cerebral cortex. Moreover, phosphorylation levels of protein kinase C-β, which is activated by DG and involved in DAT phosphorylation, were also increased. DAT expressed in Neuro-2a cells recruited DGKη to the plasma membrane and colocalized with it. These results strongly suggest that dopaminergic hyperfunction caused by DGKη deficiency in the brain leads to mania-like behaviors.
Collapse
Affiliation(s)
- Maho Asami
- Department of Chemistry, Graduate School of Science, Chiba University, Japan
| | - Yuji Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Japan
| |
Collapse
|
20
|
Takao S, Akiyama R, Sakane F. Combined inhibition/silencing of diacylglycerol kinase α and ζ simultaneously and synergistically enhances interleukin-2 production in T cells and induces cell death of melanoma cells. J Cell Biochem 2021; 122:494-506. [PMID: 33399248 DOI: 10.1002/jcb.29876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022]
Abstract
The α-isozyme of diacylglycerol kinase (DGK) enhances cancer cell proliferation and, conversely, it promotes the nonresponsive immune state known as T-cell anergy. Moreover, a DGKα-selective inhibitor, CU-3, induced cell death in cancer-derived cells and simultaneously enhanced T-cell interleukin-2 production. In addition to DGKα, DGKζ is also known to induce T-cell anergy. In the present study, we examined whether combined inhibition/silencing of DGKα and DGKζ synergistically enhanced T-cell activity. Combined treatment with CU-3 or DGKα-small interfering RNA (siRNA) and DGKζ-siRNA more potently enhanced T-cell receptor-crosslink-dependent interleukin-2 production in Jurkat T cells than treatment with either alone. Intriguingly, in addition to activating T cells, dual inhibition/silencing of DGKα and DGKζ synergistically reduced viability and increased caspase 3/7 activity in AKI melanoma cells. Taken together, these results indicate that combined inhibition/silencing of DGKα and DGKζ simultaneously and synergistically enhances interleukin-2 production in T cells and induces cell death in melanoma. Therefore, dual inhibition/silencing of these DGK isozymes represents an ideal therapy that potently attenuates cancer cell proliferation and simultaneously enhances immune responses that impact anticancer immunity.
Collapse
Affiliation(s)
- Saki Takao
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Rino Akiyama
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| |
Collapse
|
21
|
Beyond Lipid Signaling: Pleiotropic Effects of Diacylglycerol Kinases in Cellular Signaling. Int J Mol Sci 2020; 21:ijms21186861. [PMID: 32962151 PMCID: PMC7554708 DOI: 10.3390/ijms21186861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
The diacylglycerol kinase family, which can attenuate diacylglycerol signaling and activate phosphatidic acid signaling, regulates various signaling transductions in the mammalian cells. Studies on the regulation of diacylglycerol and phosphatidic acid levels by various enzymes, the identification and characterization of various diacylglycerol and phosphatidic acid-regulated proteins, and the overlap of different diacylglycerol and phosphatidic acid metabolic and signaling processes have revealed the complex and non-redundant roles of diacylglycerol kinases in regulating multiple biochemical and biological networks. In this review article, we summarized recent progress in the complex and non-redundant roles of diacylglycerol kinases, which is expected to aid in restoring dysregulated biochemical and biological networks in various pathological conditions at the bed side.
Collapse
|
22
|
New Era of Diacylglycerol Kinase, Phosphatidic Acid and Phosphatidic Acid-Binding Protein. Int J Mol Sci 2020; 21:ijms21186794. [PMID: 32947951 PMCID: PMC7555651 DOI: 10.3390/ijms21186794] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Diacylglycerol kinase (DGK) phosphorylates diacylglycerol (DG) to generate phosphatidic acid (PA). Mammalian DGK consists of ten isozymes (α–κ) and governs a wide range of physiological and pathological events, including immune responses, neuronal networking, bipolar disorder, obsessive-compulsive disorder, fragile X syndrome, cancer, and type 2 diabetes. DG and PA comprise diverse molecular species that have different acyl chains at the sn-1 and sn-2 positions. Because the DGK activity is essential for phosphatidylinositol turnover, which exclusively produces 1-stearoyl-2-arachidonoyl-DG, it has been generally thought that all DGK isozymes utilize the DG species derived from the turnover. However, it was recently revealed that DGK isozymes, except for DGKε, phosphorylate diverse DG species, which are not derived from phosphatidylinositol turnover. In addition, various PA-binding proteins (PABPs), which have different selectivities for PA species, were recently found. These results suggest that DGK–PA–PABP axes can potentially construct a large and complex signaling network and play physiologically and pathologically important roles in addition to DGK-dependent attenuation of DG–DG-binding protein axes. For example, 1-stearoyl-2-docosahexaenoyl-PA produced by DGKδ interacts with and activates Praja-1, the E3 ubiquitin ligase acting on the serotonin transporter, which is a target of drugs for obsessive-compulsive and major depressive disorders, in the brain. This article reviews recent research progress on PA species produced by DGK isozymes, the selective binding of PABPs to PA species and a phosphatidylinositol turnover-independent DG supply pathway.
Collapse
|
23
|
Subcellular Localization Relevance and Cancer-Associated Mechanisms of Diacylglycerol Kinases. Int J Mol Sci 2020; 21:ijms21155297. [PMID: 32722576 PMCID: PMC7432101 DOI: 10.3390/ijms21155297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
An increasing number of reports suggests a significant involvement of the phosphoinositide (PI) cycle in cancer development and progression. Diacylglycerol kinases (DGKs) are very active in the PI cycle. They are a family of ten members that convert diacylglycerol (DAG) into phosphatidic acid (PA), two-second messengers with versatile cellular functions. Notably, some DGK isoforms, such as DGKα, have been reported to possess promising therapeutic potential in cancer therapy. However, further studies are needed in order to better comprehend their involvement in cancer. In this review, we highlight that DGKs are an essential component of the PI cycle that localize within several subcellular compartments, including the nucleus and plasma membrane, together with their PI substrates and that they are involved in mediating major cancer cell mechanisms such as growth and metastasis. DGKs control cancer cell survival, proliferation, and angiogenesis by regulating Akt/mTOR and MAPK/ERK pathways. In addition, some DGKs control cancer cell migration by regulating the activities of the Rho GTPases Rac1 and RhoA.
Collapse
|
24
|
Polyunsaturated fatty acid-containing phosphatidic acids selectively interact with L-lactate dehydrogenase A and induce its secondary structural change and inactivation. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158768. [PMID: 32717303 DOI: 10.1016/j.bbalip.2020.158768] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/17/2020] [Accepted: 07/19/2020] [Indexed: 12/18/2022]
Abstract
Phosphatidic acid (PA) consists of various molecular species that have different fatty acyl chains at the sn-1 and sn-2 positions; and consequently, mammalian cells contain at least 50 structurally distinct PA molecular species. However, the different roles of each PA species are poorly understood. In the present study, we attempted to identify dipalmitoyl (16:0/16:0)-PA-binding proteins from mouse skeletal muscle using liposome precipitation and tandem mass spectrometry analysis. We identified L-lactate dehydrogenase (LDH) A, which catalyzes conversion of pyruvate to lactate and is a key checkpoint of anaerobic glycolysis critical for tumor growth, as a 16:0/16:0-PA-binding protein. LDHA did not substantially associate with other phospholipids, such as phosphatidylcholine, phosphatidylserine, phosphatidylglycerol, phosphatidylinositol, phosphoinositides and cardiolipin at physiological pH (7.4), indicating that LDHA specifically bound to PA. Interestingly, 18:0/18:0-, 18:0/20:4- and 18:0/22:6-PA also interacted with LDHA, and their binding activities were stronger than 16:0/16:0-PA at pH 7.4. Moreover, circular dichroism spectrometry showed that 18:0/20:4- and 18:0/22:6-PA, but not 16:0/16:0- or 18:0/18:0-PA, significantly reduced the α-helical structure of LDHA. Furthermore, 18:0/20:4- and 18:0/22:6-PA attenuated LDH activity. Taken together, we demonstrated for the first time that LDHA is a PA-binding protein and is a unique PA-binding protein that is structurally and functionally controlled by associating with 18:0/20:4- and 18:0/22:6-PA.
Collapse
|
25
|
Xie D, Zhang S, Chen P, Deng W, Pan Y, Xie J, Wang J, Liao B, Sleasman JW, Zhong XP. Negative control of diacylglycerol kinase ζ-mediated inhibition of T cell receptor signaling by nuclear sequestration in mice. Eur J Immunol 2020; 50:1729-1745. [PMID: 32525220 DOI: 10.1002/eji.201948442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 04/17/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
Diacylglycerol kinases (DGKs) play important roles in restraining diacylglycerol (DAG)-mediated signaling. Within the DGK family, the ζ isoform appears to be the most important isoform in T cells for controlling their development and function. DGKζ has been demonstrated to regulate T cell maturation, activation, anergy, effector/memory differentiation, defense against microbial infection, and antitumor immunity. Given its critical functions, DGKζ function should be tightly regulated to ensure proper signal transduction; however, mechanisms that control DGKζ function are still poorly understood. We report here that DGKζ dynamically translocates from the cytosol into the nuclei in T cells after TCR stimulation. In mice, DGKζ mutant defective in nuclear localization displayed enhanced ability to inhibit TCR-induced DAG-mediated signaling in primary T cells, maturation of conventional αβT and iNKT cells, and activation of peripheral T cells compared with WT DGKζ. Our study reveals for the first time nuclear sequestration of DGKζ as a negative control mechanism to spatially restrain it from terminating DAG mediated signaling in T cells. Our data suggest that manipulation of DGKζ nucleus-cytosol shuttling as a novel strategy to modulate DGKζ activity and immune responses for treatment of autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Danli Xie
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Shimeng Zhang
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Pengcheng Chen
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Wenhai Deng
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Yun Pan
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Jinhai Xie
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Jinli Wang
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Bryce Liao
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - John W Sleasman
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina
| | - Xiao-Ping Zhong
- Division of Allergy and Immunology, Department of Pediatrics-Allergy and Immunology, Duke University Medical Center, Durham, North Carolina.,Department of Immunology, Duke University Medical Center, Durham, North Carolina.,Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
26
|
Abdel-Magid AF. Cancer Immunotherapy through the Inhibition of Diacylglycerol Kinases Alpha and Zeta. ACS Med Chem Lett 2020; 11:1083-1085. [PMID: 32550982 PMCID: PMC7294543 DOI: 10.1021/acsmedchemlett.0c00118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Indexed: 11/28/2022] Open
Affiliation(s)
- Ahmed F. Abdel-Magid
- Therachem Research Medilab,
LLC. 100 Jade Park, Chelsea, Alabama 35043, United States
| |
Collapse
|
27
|
Palmitic acid- and/or palmitoleic acid-containing phosphatidic acids are generated by diacylglycerol kinase α in starved Jurkat T cells. Biochem Biophys Res Commun 2020; 525:1054-1060. [DOI: 10.1016/j.bbrc.2020.02.162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/26/2020] [Indexed: 12/11/2022]
|
28
|
Li J, Song Y, Zhang Y, Li H, Tian M, Li D, Zhang S, Cao G, Liu C. A novel compound heterozygous mutation in DGKE in a Chinese patient causes atypical hemolytic uremic syndrome. Hematology 2020; 25:101-107. [PMID: 32091318 DOI: 10.1080/16078454.2020.1731969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Objectives: DGKE mutations can lead to hemolysis and thrombus in patients with atypical hemolytic uremic syndrome (aHUS). However, the sequence variants of DGKE in Chinese patients with aHUS have not been reported, and the protein function and crystal structure of DGKE remain unresolved.Methods: Targeted exome sequencing was accomplished in one affected patient from each family using the Illumina NextSeq 500 platform. Protein modeling and functional analysis in DGKE were also performed to understand the impact of identified variants on the phenotype.Results: We report a novel compound heterozygous mutation in the DGKE gene in a Chinese consanguineous family in which a child was diagnosed with aHUS, which includes a c.231C>G missense mutation and a c.790_791delTG frameshift mutation derived from his father and mother, respectively. Our bioinformatic analysis suggested that the allelic mutations at different sites in DGKE yield abnormal crystal structures and conformations, leading to dysregulation of its downstream signaling.Conclusions: Our study further expands the spectrum of the sequence variants reported in the DGKE gene and also indicates that different races may have different DGKE variants. Moreover, the altered structures and conformations, caused by DGKE mutations, disrupt the binding of DGKE with its partners, and leading to the occurrence of aHUS.
Collapse
Affiliation(s)
- Jitong Li
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yinsen Song
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yaodong Zhang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Hongjiang Li
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Ming Tian
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Di Li
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Shufeng Zhang
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Guanghai Cao
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Cuihua Liu
- Department of Nephrology and Rheumatology, Affiliated Children's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
29
|
Arranz-Nicolás J, Mérida I. Biological regulation of diacylglycerol kinases in normal and neoplastic tissues: New opportunities for cancer immunotherapy. Adv Biol Regul 2020; 75:100663. [PMID: 31706704 DOI: 10.1016/j.jbior.2019.100663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 06/10/2023]
Abstract
In the recent years, the arsenal of anti-cancer therapies has evolved to target T lymphocytes and restore their capacity to destroy tumor cells. However, the clinical success is limited, with a large number of patients that never responds and others that ultimately develop resistances. Overcoming the hypofunctional state imposed by solid tumors to T cells has revealed critical but challenging due to the complex strategies that tumors employ to evade the immune system. The Diacylglycerol kinases (DGK) limit DAG-dependent functions in T lymphocytes and their upregulation in tumor-infiltrating T lymphocytes contribute to limit T cell cytotoxic potential. DGK blockade could reinstate T cell attack on tumors, limiting at the same time tumor cell growth, thanks to the DGK positive input into several oncogenic pathways. In this review we summarize the latest findings regarding the regulation of specific DGK isoforms in healthy and anergic T lymphocytes, as well as their contribution to oncogenic phenotypes. We will also revise the latest advances in the search for pharmacological inhibitors and their potential as anti-cancer agents, either alone or in combination with immunomodulatory agents.
Collapse
Affiliation(s)
- Javier Arranz-Nicolás
- Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Darwin 3, UAM Campus de Cantoblanco, 28049, Madrid, Spain
| | - Isabel Mérida
- Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Darwin 3, UAM Campus de Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
30
|
Beyond the Cell Surface: Targeting Intracellular Negative Regulators to Enhance T cell Anti-Tumor Activity. Int J Mol Sci 2019; 20:ijms20235821. [PMID: 31756921 PMCID: PMC6929154 DOI: 10.3390/ijms20235821] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
It is well established that extracellular proteins that negatively regulate T cell function, such as Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA-4) and Programmed Cell Death protein 1 (PD-1), can be effectively targeted to enhance cancer immunotherapies and Chimeric Antigen Receptor T cells (CAR-T cells). Intracellular proteins that inhibit T cell receptor (TCR) signal transduction, though less well studied, are also potentially useful therapeutic targets to enhance T cell activity against tumor. Four major classes of enzymes that attenuate TCR signaling include E3 ubiquitin kinases such as the Casitas B-lineage lymphoma proteins (Cbl-b and c-Cbl), and Itchy (Itch), inhibitory tyrosine phosphatases, such as Src homology region 2 domain-containing phosphatases (SHP-1 and SHP-2), inhibitory protein kinases, such as C-terminal Src kinase (Csk), and inhibitory lipid kinases such as Src homology 2 (SH2) domain-containing inositol polyphosphate 5-phosphatase (SHIP) and Diacylglycerol kinases (DGKs). This review describes the mechanism of action of eighteen intracellular inhibitory regulatory proteins in T cells within these four classes, and assesses their potential value as clinical targets to enhance the anti-tumor activity of endogenous T cells and CAR-T cells.
Collapse
|
31
|
Honda S, Murakami C, Yamada H, Murakami Y, Ishizaki A, Sakane F. Analytical Method for Diacylglycerol Kinase ζ Activity in Cells Using Protein Myristoylation and Liquid Chromatography-Tandem Mass Spectrometry. Lipids 2019; 54:763-771. [PMID: 31736090 DOI: 10.1002/lipd.12201] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/06/2019] [Accepted: 10/24/2019] [Indexed: 01/05/2023]
Abstract
Specific inhibitors of diacylglycerol kinase (DGK) ζ can be promising anticancer medications via the activation of cancer immunity. Although the detection of cellular activities of target enzymes is essential for drug screening in addition to in vitro assays, it is difficult to detect the activity of DGKζ in cells. In the present study, we generated AcGFP-DGKζ cDNA with a consensus N-myristoylation sequence at the 5' end (Myr-AcGFP-DGKζ) to target DGKζ to membranes. Using liquid chromatography (LC)-tandem mass spectrometry (MS/MS) (LC-MS/MS), we showed that Myr-AcGFP-DGKζ, but not AcGFP-DGKζ without the myristoylation sequence, substantially augmented the levels of several phosphatidic acid (PtdOH) species. In contrast to Myr-AcGFP-DGKζ, its inactive mutant did not exhibit an increase in PtdOH production, indicating that the increase in PtdOH production was DGK activity-dependent. This method will be useful in chemical compound selection for the development of drugs targeting DGKζ and can be applicable to various soluble (nonmembrane bound) lipid-metabolizing enzymes, including other DGK isozymes.
Collapse
Affiliation(s)
- Shotaro Honda
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Haruka Yamada
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Yuki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Ayuka Ishizaki
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| |
Collapse
|
32
|
Nakano T, Topham MK, Goto K. Mice lacking DGKε show increased beige adipogenesis in visceral white adipose tissue after long-term high fat diet in a COX-2- dependent manner. Adv Biol Regul 2019; 75:100659. [PMID: 31607681 DOI: 10.1016/j.jbior.2019.100659] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/27/2022]
Abstract
Adipose tissue is a central site for energy storage in the form of triglyceride (TG). Under excess energy conditions, TG is synthesized by acylation of diacylglycerol (DG), whereas TG is broken down into DG and free fatty acid, which provide energy for mitochondrial lipid oxidation when needed. In this regard, DG is not merely an intermediate metabolite for TG metabolism; it also serves as a signaling molecule. DG kinase (DGK) phosphorylates DG to produce phosphatidic acid (PA). Consequently, DGK plays a pivotal role in the control of lipid metabolism and signal transduction pathway. Recently, a report has described that DGKε-knockout (KO) mice show expansion of epididymal white adipose tissue (WAT) together with the impairment of glucose clearance after short-term (40 days) high fat diet (HFD) feeding, an early presymptomatic phase of obesity in wild-type animals. Nevertheless, no report describes an investigation of their phenotype under long-term HFD feeding conditions. Remarkably, results obtained during long-term HFD feeding show that WAT mass is decreased significantly and that the blood glucose profile in response to glucose challenge is improved in DGKε-KO mice compared with wild-type, which contrast sharply against the phenotype shown for short-term HFD feeding. Morphological examination reveals that cyclooxygenase-2 (COX-2) expression and clusters of uncoupling protein 1 (UCP1)-positive multilocular brown-like ("beige") adipocyte are induced in DGKε-deficient WAT after long-term HFD feeding, suggesting that beige adipocytes facilitate energy expenditure during prolonged HFD feeding. Administration of celecoxib, a selective inhibitor of COX-2, abolishes the appearance of UCP1-positive beige adipocytes in DGKε-KO mice. These findings suggest that DGKε deficiency promotes visceral WAT remodeling in a COX-2-dependent manner under long-term HFD feeding conditions.
Collapse
Affiliation(s)
- Tomoyuki Nakano
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata City, Yamagata, 9909585, Japan.
| | - Matthew K Topham
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Kaoru Goto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata City, Yamagata, 9909585, Japan
| |
Collapse
|
33
|
Gu X, Wan G, Chen N, Li J, Chen B, Tang Y, Gu W, Jin C, Meng J, Zhang P, Liu L, Yang Z, Lu C. DGKζ Plays Crucial Roles in the Proliferation and Tumorigenicity of Human glioblastoma. Int J Biol Sci 2019; 15:1872-1881. [PMID: 31523189 PMCID: PMC6743304 DOI: 10.7150/ijbs.35193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/23/2019] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma is one of the most malignant brain cancers in adults, and it is a fatal disease because of its untimely pathogenetic location detection, infiltrative growth, and unfavorable prognosis. Unfortunately, multimodal treatment with maximal safe resection, chemotherapy and radiation has not increased the survival rate of patients with glioblastoma. Gene- and molecular-targeted therapy is considered to be a promising anticancer strategy for glioblastoma. The identification of novel potential targets in glioblastoma is of high importance. In this study, we found that both the mRNA and protein levels of diacylglycerol kinase ζ (DGKζ) were significantly higher in glioblastoma tissues than in precancerous lesions. The silencing of DGKζ by lentivirus-delivered shRNA reduced glioblastoma cell proliferation and induced G0/G1 phase arrest. Moreover, knockdown of DGKζ expression in U251 cells markedly reduced in vitro colony formation and in vivo tumorigenic capability. Further study showed that DGKζ inhibition resulted in decreases in cyclin D1, p-AKT and p-mTOR. Moreover, the rescue or overexpression of DGKζ in glioblastoma cells demonstrated the oncogenic function of DGKζ. In conclusion, these studies suggest that the suppression of DGKζ may inhibit the tumor growth of glioblastoma cells with high DGKζ expression. Thus, DGKζ might be a potential therapeutic target in malignant glioblastoma.
Collapse
Affiliation(s)
- Xuefeng Gu
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,College of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Guoqing Wan
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Nianhong Chen
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Jinhong Li
- Department of Neurosurgery, Jiangmen Central Hospital, Jiangmen, Guangdong, PR China
| | - Bing Chen
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Yeling Tang
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Wangxian Gu
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Cuiting Jin
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Jihong Meng
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Peng Zhang
- College of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Li Liu
- College of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Zhifang Yang
- College of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| | - Changlian Lu
- Collaborative Research Center, Shanghai University of Medicine & Health Sciences, Shanghai, PR China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, PR China
| |
Collapse
|
34
|
Diacylglycerol kinase control of protein kinase C. Biochem J 2019; 476:1205-1219. [DOI: 10.1042/bcj20180620] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/27/2022]
Abstract
Abstract
The diacylglycerol kinases (DGK) are lipid kinases that transform diacylglycerol (DAG) into phosphatidic acid (PA) in a reaction that terminates DAG-based signals. DGK provide negative regulation to conventional and novel protein kinase C (PKC) enzymes, limiting local DAG availability in a tissue- and subcellular-restricted manner. Defects in the expression/activity of certain DGK isoforms contribute substantially to cognitive impairment and mental disorders. Abnormal DGK overexpression in tumors facilitates invasion and resistance to chemotherapy preventing tumor immune destruction by tumor-infiltrating lymphocytes. Effective translation of these findings into therapeutic approaches demands a better knowledge of the physical and functional interactions between the DGK and PKC families. DGKζ is abundantly expressed in the nervous and immune system, where physically and functionally interacts with PKCα. The latest discoveries suggest that PDZ-mediated interaction facilitates spatial restriction of PKCα by DGKζ at the cell–cell contact sites in a mechanism where the two enzymes regulate each other. In T lymphocytes, DGKζ interaction with Sorting Nexin 27 (SNX27) guarantees the basal control of PKCα activation. SNX27 is a trafficking component required for normal brain function whose deficit has been linked to Alzheimer's disease (AD) pathogenesis. The enhanced PKCα activation as the result of SNX27 silencing in T lymphocytes aligns with the recent correlation found between gain-of-function PKCα mutations and AD and suggests that disruption of the mechanisms that provides a correct spatial organization of DGKζ and PKCα may lie at the basis of immune and neuronal synapse impairment.
Collapse
|
35
|
Saito T, Takahashi D, Sakane F. Expression, Purification, and Characterization of Human Diacylglycerol Kinase ζ. ACS OMEGA 2019; 4:5540-5546. [PMID: 31893253 PMCID: PMC6935893 DOI: 10.1021/acsomega.9b00079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/08/2019] [Indexed: 05/14/2023]
Abstract
Diacylglycerol kinase ζ (DGKζ) phosphorylates diacylglycerol (DG) to generate phosphatidic acid. The dysfunction of DGKζ has been linked to several diseases, such as cardiac hypertrophy, ischemia, and seizures. Moreover, much attention has been paid to DGKζ, together with DGKα, as a potential target for cancer immunotherapy. However, DGKζ has never been purified and, thus, neither its enzymatic properties nor its structure has yet been reported, hindering our understanding of the catalytic mechanism of DGKζ and the development of a reasonable structure-based drug design. In the present study, we generated a full-length DGKζ using a baculovirus-insect cell expression system for enzymological and structural studies. Full-length DGKζ remained soluble and was purified to near homogeneity as a monomer with yields suitable for protein crystallization (0.63 mg/1 L culture). Enzymatic characterization showed that the purified DGKζ is in a fully functional state. The K m values for adenosine triphosphate (ATP) and DG were 0.05 mM and 1.5 mol %, respectively, and the EC50 for the activator phosphatidylserine was 8.6 mol %, indicating that its affinity for ATP is moderately higher than those of DGKα and DGKε, and its affinities for DG and phosphatidylserine are comparable to those of DGKα/DGKε. We further confirmed that the purified enzyme could be concentrated without any significant aggregation. Circular dichroism revealed that DGKζ is comprised of 25% α-helices and 18% β-strands. This is the first successful purification and characterization of the enzymatic and conformational properties of DGKζ. The purification of DGKζ allows detailed analyses of this important enzyme and will advance our understanding of DGKζ-related diseases and therapies.
Collapse
Affiliation(s)
- Takumi Saito
- Department
of Chemistry, Graduate School of Science, Chiba University, 1-33
Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Daisuke Takahashi
- Department
of Pharmaceutical Health Care and Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Fumio Sakane
- Department
of Chemistry, Graduate School of Science, Chiba University, 1-33
Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
- E-mail: . Tel/Fax: +81-43-290-3695
| |
Collapse
|
36
|
Takahashi D, Suzuki K, Sakamoto T, Iwamoto T, Murata T, Sakane F. Crystal structure and calcium-induced conformational changes of diacylglycerol kinase α EF-hand domains. Protein Sci 2019; 28:694-706. [PMID: 30653270 DOI: 10.1002/pro.3572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 01/07/2023]
Abstract
Diacylglycerol kinases (DGKs) are multi-domain lipid kinases that phosphorylate diacylglycerol into phosphatidic acid, modulating the levels of these key signaling lipids. Recently, increasing attention has been paid to DGKα isozyme as a potential target for cancer immunotherapy. We have previously shown that DGKα is positively regulated by Ca2+ binding to its N-terminal EF-hand domains (DGKα-EF). However, little progress has been made for the structural biology of mammalian DGKs and the molecular mechanism underlying the Ca2+ -triggered activation remains unclear. Here we report the first crystal structure of Ca2+ -bound DGKα-EF and analyze the structural changes upon binding to Ca2+ . DGKα-EF adopts a canonical EF-hand fold, but unexpectedly, has an additional α-helix (often called a ligand mimic [LM] helix), which is packed into the hydrophobic core. Biophysical and biochemical analyses reveal that DGKα-EF adopts a protease-susceptible "open" conformation without Ca2+ that tends to form a dimer. Cooperative binding of two Ca2+ ions dissociates the dimer into a well-folded monomer, which resists to proteolysis. Taken together, our results provide experimental evidence that Ca2+ binding induces substantial conformational changes in DGKα-EF, which likely regulates intra-molecular interactions responsible for the activation of DGKα and suggest a possible role of the LM helix for the Ca2+ -induced conformational changes. SIGNIFICANCE STATEMENT: Diacylglycerol kinases (DGKs), which modulates the levels of two lipid second messengers, diacylglycerol and phosphatidic acid, is still structurally enigmatic enzymes since its first identification in 1959. We here present the first crystal structure of EF-hand domains of diacylglycerol kinase α in its Ca2+ bound form and characterize Ca2+ -induced conformational changes, which likely regulates intra-molecular interactions. Our study paves the way for future studies to understand the structural basis of DGK isozymes.
Collapse
Affiliation(s)
- Daisuke Takahashi
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Kano Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Taiichi Sakamoto
- Department of Life Science, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba, Japan
| | - Takeo Iwamoto
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan.,Molecular Chirality Research Center, Chiba University, Chiba, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| |
Collapse
|
37
|
Ma Q, Gabelli SB, Raben DM. Diacylglycerol kinases: Relationship to other lipid kinases. Adv Biol Regul 2019; 71:104-110. [PMID: 30348515 PMCID: PMC6347529 DOI: 10.1016/j.jbior.2018.09.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 04/17/2023]
Abstract
Lipid kinases regulate a wide variety of cellular functions and have emerged as one the most promising targets for drug design. Diacylglycerol kinases (DGKs) are a family of enzymes that catalyze the ATP-dependent phosphorylation of diacylglycerol (DAG) to phosphatidic acid (PtdOH). Despite the critical role in lipid biosynthesis, both DAG and PtdOH have been shown as bioactive lipids mediating a number of signaling pathways. Although there is increasing recognition of their role in signaling systems, our understanding of the key enzyme which regulate the balance of these two lipid messages remain limited. Solved structures provide a wealth of information for understanding the function and regulation of these enzymes. Solving the structures of mammalian DGKs by traditional NMR and X-ray crystallography approaches have been challenging and so far, there are still no three-dimensional structures of these DGKs. Despite this, some insights may be gained by examining the similarities and differences between prokaryotic DGKs and other mammalian lipid kinases. This review focuses on summarizing and comparing the structure of prokaryotic and mammalian DGKs as well as two other lipid kinases: sphingosine kinase and phosphatidylinositol-3-kinase. How these known lipid kinases structures relate to mammalian DGKs will also be discussed.
Collapse
Affiliation(s)
- Qianqian Ma
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sandra B Gabelli
- The Department of Biophysics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel M Raben
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
38
|
Ávila-Flores A, Arranz-Nicolás J, Mérida I. Transcriptional Activity of FOXO Transcription Factors Measured by Luciferase Assays. Methods Mol Biol 2019; 1890:91-102. [PMID: 30414147 DOI: 10.1007/978-1-4939-8900-3_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Forkhead box O (FOXO) family of transcription factors translates environmental cues into gene expression. FOXO factors are crucial for the maintenance of cell homeostasis, with important roles in cell fate decisions and differentiation. Identification of FOXO target genes requires strict validation by several methods. Luciferase-based reporters are a valuable starting point for determining the transcription-promoting capacity of potential FOXO-binding sites in candidate genes. Luciferase, an enzyme found in bioluminescent organisms catalyzes oxidation of luciferin to produce oxyluciferin together with light, which can be easily detected and measured with a luminometer. Due to their many advantages, transcriptional assays based on luciferase activity are widely used; they are easy, highly reproducible, and very sensitive. Continued improvements in luciferase-based vectors and measurement reagents confer considerable versatility. Luciferase-based reporters are also a reliable approach in the search for unknown components in the signaling pathways that control FOXO factor activity.We previously reported that FOXO transcription factors control expression of the enzyme diacylglycerol kinase α (DGKα) in T cells. DGKα consumes diacylglycerol, a lipid that activates several mitogenic pathways. Here, we describe the use of a luciferase-based promoter bearing the FOXO-binding sites of the DGKα gene to explore the relationship between the expression of this enzyme and stress conditions in NIH3T3 mouse fibroblasts. Our data support a role for FOXO factors in promoting high DGKα levels in conditions of growth factor deprivation. DGKα regulation by FOXO factors correlates with the reported alterations in DGKα expression during cell transformation and cancer progression.
Collapse
Affiliation(s)
- Antonia Ávila-Flores
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| | - Javier Arranz-Nicolás
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Isabel Mérida
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
39
|
Abrams SL, Follo MY, Steelman LS, Lertpiriyapong K, Cocco L, Ratti S, Martelli AM, Candido S, Libra M, Murata RM, Rosalen PL, Montalto G, Cervello M, Gizak A, Rakus D, Mao W, Lombardi P, McCubrey JA. Abilities of berberine and chemically modified berberines to inhibit proliferation of pancreatic cancer cells. Adv Biol Regul 2019; 71:172-182. [PMID: 30361003 DOI: 10.1016/j.jbior.2018.10.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 06/08/2023]
Abstract
Berberine (BBR) is a common nutraceutical consumed by millions worldwide. BBR has many different effects on human health, e.g., diabetes, diarrhea, inflammation and now more recently it has been proposed to have potent anti-cancer effects. BBR has been shown to suppress the growth of cancer cells more than normal cells. BBR has been proposed to exert its growth-inhibitory effects by many different biochemical mechanisms including: suppression of cell cycle progression, induction of reactive oxygen species, induction of apoptosis and autophagy and interactions with DNA potentially leading to DNA damage, and altered gene expression. Pancreatic cancer is a leading cancer worldwide associated with a poor prognosis. As our population ages, pancreatic cancer has an increasing incidence and will likely become the second leading cause of death from cancer. There are few truly-effective therapeutic options for pancreatic cancer. Surgery and certain chemotherapeutic drugs are used to treat pancreatic cancer patients. Novel approaches to treat pancreatic cancer patients are direly needed as they usually survive for less than a year after being diagnosed. In the following manuscript, we discuss the abilities of BBR and certain chemically-modified BBRs (NAX compounds) to suppress growth of pancreatic cancer cells.
Collapse
Affiliation(s)
- Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Matilde Y Follo
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA; Center of Comparative Medicine and Pathology, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medicine and the Hospital for Special Surgery, New York City, NY, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Ramiro M Murata
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Weifeng Mao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe di Vittorio 70, Novate Milanese, 20026, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA.
| |
Collapse
|
40
|
Merida I, Arranz-Nicolás J, Torres-Ayuso P, Ávila-Flores A. Diacylglycerol Kinase Malfunction in Human Disease and the Search for Specific Inhibitors. Handb Exp Pharmacol 2019; 259:133-162. [PMID: 31227890 DOI: 10.1007/164_2019_221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The diacylglycerol kinases (DGKs) are master regulator kinases that control the switch from diacylglycerol (DAG) to phosphatidic acid (PA), two lipids with important structural and signaling properties. Mammalian DGKs distribute into five subfamilies that regulate local availability of DAG and PA pools in a tissue- and subcellular-restricted manner. Pharmacological manipulation of DGK activity holds great promise, given the critical contribution of specific DGK subtypes to the control of membrane structure, signaling complexes, and cell-cell communication. The latest advances in the DGK field have unveiled the differential contribution of selected isoforms to human disease. Defects in the expression/activity of individual DGK isoforms contribute substantially to cognitive impairment, mental disorders, insulin resistance, and vascular pathologies. Abnormal DGK overexpression, on the other hand, confers the acquisition of malignant traits including invasion, chemotherapy resistance, and inhibition of immune attack on tumors. Translation of these findings into therapeutic approaches will require development of methods to pharmacologically modulate DGK functions. In particular, inhibitors that target the DGKα isoform hold particular promise in the fight against cancer, on their own or in combination with immune-targeting therapies.
Collapse
Affiliation(s)
- Isabel Merida
- Department of Immunology and Oncology, National Center of Biotechnology (CNB-CSIC), Madrid, Spain.
| | - Javier Arranz-Nicolás
- Department of Immunology and Oncology, National Center of Biotechnology (CNB-CSIC), Madrid, Spain
| | - Pedro Torres-Ayuso
- Laboratory of Cell and Developmental Signaling, National Cancer Institute (NCI-NIH), Frederick, MD, USA
| | - Antonia Ávila-Flores
- Department of Immunology and Oncology, National Center of Biotechnology (CNB-CSIC), Madrid, Spain
| |
Collapse
|
41
|
Adamo A, Dal Collo G, Bazzoni R, Krampera M. Role of mesenchymal stromal cell-derived extracellular vesicles in tumour microenvironment. Biochim Biophys Acta Rev Cancer 2018; 1871:192-198. [PMID: 30599188 DOI: 10.1016/j.bbcan.2018.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/22/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Stromal cells, deriving from mesenchymal stromal cells (MSCs), are crucial component of tumour microenvironment and represent key regulators of tumour processes. MSCs can be recruited to the tumour environment and interact with many cellular elements, thus influencing tumour biology. Cell-to-cell communication is in part mediated by the release of extracellular vesicle (EVs). EVs can induce significant molecular changes in recipient cells, delivering bioactive molecules. In this review, we describe the MSC-derived EVs content and discuss their role in different processes related to cancer biology. Furthermore, we summarize chemical or biological EVs modifications aiming to develop more efficient antitumor therapies.
Collapse
Affiliation(s)
- A Adamo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - G Dal Collo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - R Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - M Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
42
|
Yamaki A, Akiyama R, Murakami C, Takao S, Murakami Y, Mizuno S, Takahashi D, Kado S, Taketomi A, Shirai Y, Goto K, Sakane F. Diacylglycerol kinase α-selective inhibitors induce apoptosis and reduce viability of melanoma and several other cancer cell lines. J Cell Biochem 2018; 120:10043-10056. [PMID: 30536880 DOI: 10.1002/jcb.28288] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/24/2018] [Indexed: 02/02/2023]
Abstract
Diacylglycerol (DG) kinase (DGK), which phosphorylates DG to generate phosphatidic acid (PA), consists of ten isozymes (α-к). Recently, we identified a novel small molecule inhibitor, CU-3, that selectively inhibits the activity of the α isozyme. In addition, we newly obtained Compound A, which selectively and strongly inhibits type I DGKs (α, β, and γ). In the present study, we demonstrated that both CU-3 and Compound A induced apoptosis (caspase 3/7 activity and DNA fragmentation) and viability reduction of AKI melanoma cells. Liquid chromatography-mass spectrometry revealed that the production of 32:0- and 34:0-PA species was commonly attenuated by CU-3 and Compound A, suggesting that lower levels of these PA molecular species are involved in the apoptosis induction and viability reduction of AKI cells. We determined the effects of the DGKα inhibitors on several other cancer cell lines derived from refractory cancers. In addition to melanoma, the DGKα inhibitors enhanced caspase 3/7 activity and reduced the viability of hepatocellular carcinoma, glioblastoma, and pancreatic cancer cells, but not breast adenocarcinoma cells. Interestingly, Western blot analysis indicated that the DGKα expression levels were positively correlated with the sensitivity to the DGK inhibitors. Because both CU-3 and Compound A induced interleukin-2 production by T cells, it is believed that these two compounds can enhance cancer immunity. Taken together, our results suggest that DGKα inhibitors are promising anticancer drugs.
Collapse
Affiliation(s)
- Atsumi Yamaki
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Rino Akiyama
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Saki Takao
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Yuki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Satoru Mizuno
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Daisuke Takahashi
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Sayaka Kado
- Center for Analytical Instrumentation, Chiba University, Chiba, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yasuhito Shirai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Science, Faculty of Agriculture, Kobe University, Kobe, Japan
| | - Kaoru Goto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Yamagata, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| |
Collapse
|
43
|
Roy J, Dibaeinia P, Fan TM, Sinha S, Das A. Global analysis of osteosarcoma lipidomes reveal altered lipid profiles in metastatic versus nonmetastatic cells. J Lipid Res 2018; 60:375-387. [PMID: 30504231 DOI: 10.1194/jlr.m088559] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma (OS) is the most common form of primary bone cancer in humans. The early detection and subsequent control of metastasis has been challenging in OS. Lipids are important constituents of cells that maintain structural integrity that can be converted into lipid-signaling molecules and are reprogrammed in cancerous states. Here, we investigate the global lipidomic differences in metastatic (143B) and nonmetastatic (HOS) human OS cells as compared with normal fetal osteoblast cells (FOB) using lipidomics. We detect 15 distinct lipid classes in all three cell lines that included over 1,000 lipid species across various classes including phospholipids, sphingolipids and ceramides, glycolipids, and cholesterol. We identify a key class of lipids, diacylglycerols, which are overexpressed in metastatic OS cells as compared with their nonmetastatic or nontumorigenic counterparts. As a proof of concept, we show that blocking diacylglycerol synthesis reduces cellular viability and reduces cell migration in metastatic OS cells. Thus, the differentially regulated lipids identified in this study might aid in biomarker discovery, and the synthesis and metabolism of specific lipids could serve as future targets for therapeutic development.
Collapse
Affiliation(s)
- Jahnabi Roy
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL 61802
| | - Payam Dibaeinia
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61802
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802
| | - Saurabh Sinha
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61802.,Neuroscience Program and Department of Bioengineering, Institute of Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61802
| | - Aditi Das
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, IL 61802 .,Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61802.,Beckman Institute for Advanced Science, Division of Nutritional Sciences, Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61802
| |
Collapse
|
44
|
Takahashi D, Sakane F. Expression and purification of human diacylglycerol kinase α from baculovirus-infected insect cells for structural studies. PeerJ 2018; 6:e5449. [PMID: 30128205 PMCID: PMC6089211 DOI: 10.7717/peerj.5449] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/26/2018] [Indexed: 02/02/2023] Open
Abstract
Diacylglycerol kinases (DGKs) are lipid kinases that modulate the levels of lipid second messengers, diacylglycerol and phosphatidic acid. Recently, increasing attention has been paid to its α isozyme (DGKα) as a potential target for cancer immunotherapy. DGKα consists of the N-terminal regulatory domains including EF-hand motifs and C1 domains, and the C-terminal catalytic domain (DGKα-CD). To date, however, no structures of mammalian DGKs including their CDs have yet been reported, impeding our understanding on the catalytic mechanism of DGKs and the rational structure-based drug design. Here we attempted to produce DGKα-CD or a full-length DGKα using bacterial and baculovirus-insect cell expression system for structural studies. While several DGKα-CD constructs produced using both bacterial and insect cells formed insoluble or soluble aggregates, the full-length DGKα expressed in insect cells remained soluble and was purified to near homogeneity as a monomer with yields (1.3 mg/mL per one L cell culture) feasible for protein crystallization. Following enzymatic characterization showed that the purified DGKα is in fully functional state. We further demonstrated that the purified enzyme could be concentrated without any significant aggregation, and characterized its secondary structure by circular dichroism. Taken together, these results suggest that the presence of N-terminal regulatory domains suppress protein aggregation likely via their intramolecular interactions with DGKα-CD, and demonstrate that the baculovirus-insect cell expression of the full-length form of DGKα, not DGKα-CD alone, represents a promising approach to produce protein sample for structural studies of DGKα. Thus, our study will encourage future efforts to determine the crystal structure of DGK, which has not been determined since it was first identified in 1959.
Collapse
Affiliation(s)
- Daisuke Takahashi
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| |
Collapse
|
45
|
Chappell WH, Candido S, Abrams SL, Russo S, Ove R, Martelli AM, Cocco L, Ramazzotti G, Cervello M, Montalto G, Steelman LS, Leng X, Arlinghaus RB, Libra M, McCubrey JA. Roles of p53, NF-κB and the androgen receptor in controlling NGAL expression in prostate cancer cell lines. Adv Biol Regul 2018; 69:43-62. [PMID: 29861174 DOI: 10.1016/j.jbior.2018.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL a.k.a lipocalin 2, lnc2) is a secreted protein which can form a complex with matrix metalloproteinase-9 (MMP9). This MMP9/NGAL complex has been associated with metastasis. MMP9 and NGAL are detected in the urine of patients afflicted with many different types of cancer, including prostate cancer. The effects of p53, NF-κB and the androgen receptor (AR) on the expression of NGAL was examined in four prostate cancer cell lines. Prostate cancer cell lines that are AR negative and expressed either mutant or no p53 (DU145 and PC3) displayed higher levels of NGAL expression compared to the prostate cancer cell lines (LNCaP and 22Rv-1) which are AR positive and express wild type (WT) p53. Introduction of WT-p53 into the PC3 prostate cancer cell line, resulted in reduction of the levels of NGAL expression. Conversely, introduction of dominant negative (DN) p53 or a retroviral construct expressing NF-κB into LNCaP cells increased NGAL expression. NGAL expression had functional effects on the ability of the cells to form colonies in soft agar. Whereas suppression of WT-53 in LNCaP cells increased NGAL expression, the introduction of WT-p53 suppressed NGAL transcription activity in PC3 prostate cells which normally express high level of NGAL. NF-κB and p53 were determined to regulate NGAL expression by positive and negative mechanisms, respectively. Our data indicate that prostate cancer growth, progression and sensitivity to chemotherapeutic drugs are regulated in part by NGAL and may involve complex interactions between NGAL, MMP9, NF-κB and p53.
Collapse
Affiliation(s)
- William H Chappell
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA; Becton, Dickinson and Company (BD), BD Diagnostics, Franklin Lakes, NJ, USA
| | - Saverio Candido
- Department of Bio-Medical Sciences, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Suzanne Russo
- Department of Radiation Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA; Case Western Reserve University, Cleveland, OH, USA
| | - Roger Ove
- Department of Radiation Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA; Case Western Reserve University, Cleveland, OH, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Giulia Ramazzotti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Giuseppe Montalto
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy; Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Xiaohong Leng
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas Medical Center at Houston, Houston, TX, USA
| | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas Medical Center at Houston, Houston, TX, USA
| | - Massimo Libra
- Department of Bio-Medical Sciences, University of Catania, Catania, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
46
|
Candido S, Abrams SL, Steelman L, Lertpiriyapong K, Martelli AM, Cocco L, Ratti S, Follo MY, Murata RM, Rosalen PL, Lombardi P, Montalto G, Cervello M, Gizak A, Rakus D, Suh PG, Libra M, McCubrey JA. Metformin influences drug sensitivity in pancreatic cancer cells. Adv Biol Regul 2018; 68:13-30. [PMID: 29482945 DOI: 10.1016/j.jbior.2018.02.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 06/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5-10% after diagnosis and treatment. Pancreatic cancer has been associated with type II diabetes as the frequency of recently diagnosed diabetics that develop pancreatic cancer within a 10-year period of initial diagnosis of diabetes in increased in comparison to non-diabetic patients. Metformin is a very frequently prescribed drug used to treat type II diabetes. Metformin acts in part by stimulating AMP-kinase (AMPK) and results in the suppression of mTORC1 activity and the induction of autophagy. In the following studies, we have examined the effects of metformin in the presence of various chemotherapeutic drugs, signal transduction inhibitors and natural products on the growth of three different PDAC lines. Metformin, by itself, was not effective at suppressing growth of the pancreatic cancer cell lines at concentration less than 1000 nM, however, in certain PDAC lines, a suboptimal dose of metformin (250 nM) potentiated the effects of various chemotherapeutic drugs used to treat pancreatic cancer (e.g., gemcitabine, cisplatin, 5-fluorouracil) and other cancer types (e.g., doxorubicin, docetaxel). Furthermore, metformin could increase anti-proliferative effects of mTORC1 and PI3K/mTOR inhibitors as well as natural products such as berberine and the anti-malarial drug chloroquine in certain PDAC lines. Thus, metformin can enhance the effects of certain drugs and signal transduction inhibitors which are used to treat pancreatic and various other cancers.
Collapse
Affiliation(s)
- Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Linda Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Ramiro M Murata
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Pann-Gill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
47
|
McCloud RL, Franks CE, Campbell ST, Purow BW, Harris TE, Hsu KL. Deconstructing Lipid Kinase Inhibitors by Chemical Proteomics. Biochemistry 2018; 57:231-236. [PMID: 29155586 PMCID: PMC5771882 DOI: 10.1021/acs.biochem.7b00962] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Diacylglycerol kinases (DGKs) regulate lipid metabolism and cell signaling through ATP-dependent phosphorylation of diacylglycerol to biosynthesize phosphatidic acid. Selective chemical probes for studying DGKs are currently lacking and are needed to annotate isoform-specific functions of these elusive lipid kinases. Previously, we explored fragment-based approaches to discover a core fragment of DGK-α (DGKα) inhibitors responsible for selective binding to the DGKα active site. Here, we utilize quantitative chemical proteomics to deconstruct widely used DGKα inhibitors to identify structural regions mediating off-target activity. We tested the activity of a fragment (RLM001) derived from a nucleotide-like region found in the DGKα inhibitors R59022 and ritanserin and discovered that RLM001 mimics ATP in its ability to broadly compete at ATP-binding sites of DGKα as well as >60 native ATP-binding proteins (kinases and ATPases) detected in cell proteomes. Equipotent inhibition of activity-based probe labeling by RLM001 supports a contiguous ligand-binding site composed of C1, DAGKc, and DAGKa domains in the DGKα active site. Given the lack of available crystal structures of DGKs, our studies highlight the utility of chemical proteomics in revealing active-site features of lipid kinases to enable development of inhibitors with enhanced selectivity against the human proteome.
Collapse
Affiliation(s)
- Rebecca L. McCloud
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Caroline E. Franks
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Sean T. Campbell
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Benjamin W. Purow
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Thurl E. Harris
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| |
Collapse
|
48
|
McCubrey JA, Abrams SL, Lertpiriyapong K, Cocco L, Ratti S, Martelli AM, Candido S, Libra M, Murata RM, Rosalen PL, Lombardi P, Montalto G, Cervello M, Gizak A, Rakus D, Steelman LS. Effects of berberine, curcumin, resveratrol alone and in combination with chemotherapeutic drugs and signal transduction inhibitors on cancer cells-Power of nutraceuticals. Adv Biol Regul 2018; 67:190-211. [PMID: 28988970 DOI: 10.1016/j.jbior.2017.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 06/07/2023]
Abstract
Over the past fifty years, society has become aware of the importance of a healthy diet in terms of human fitness and longevity. More recently, the concept of the beneficial effects of certain components of our diet and other compounds, that are consumed often by different cultures in various parts of the world, has become apparent. These "healthy" components of our diet are often referred to as nutraceuticals and they can prevent/suppress: aging, bacterial, fungal and viral infections, diabetes, inflammation, metabolic disorders and cardiovascular diseases and have other health-enhancing effects. Moreover, they are now often being investigated because of their anti-cancer properties/potentials. Understanding the effects of various natural products on cancer cells may enhance their usage as anti-proliferative agents which may be beneficial for many health problems. In this manuscript, we discuss and demonstrate how certain nutraceuticals may enhance other anti-cancer drugs to suppress proliferation of cancer cells.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA.
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA; Center of Comparative Medicine and Pathology, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medicine and the Hospital for Special Surgery, New York City, New York, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Ramiro M Murata
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|
49
|
Ratti S, Ramazzotti G, Faenza I, Fiume R, Mongiorgi S, Billi AM, McCubrey JA, Suh PG, Manzoli L, Cocco L, Follo MY. Nuclear inositide signaling and cell cycle. Adv Biol Regul 2018; 67:1-6. [PMID: 29102395 DOI: 10.1016/j.jbior.2017.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 06/07/2023]
Abstract
Phosphatidylinositols (PIs) are responsible for several signaling pathways related to many cellular functions, such as cell cycle regulation at different check-points, cell proliferation, cell differentiation, membrane trafficking and gene expression. PI metabolism is not only present at the cytoplasmic level, but also at the nuclear one, where different signaling pathways affect essential nuclear mechanisms in eukaryotic cells. In this review we focus on nuclear inositide signaling in relation to cell cycle regulation. Many evidences underline the pivotal role of nuclear inositide signaling in cell cycle regulation and cell proliferation associated to different strategic physiopathological mechanisms in several cell systems and diseases.
Collapse
Affiliation(s)
- Stefano Ratti
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Irene Faenza
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Roberta Fiume
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Sara Mongiorgi
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Anna Maria Billi
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, MS#629, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Lucia Manzoli
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Lucio Cocco
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Matilde Y Follo
- Cellular Signalling Laboratory Department of Biomedical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
50
|
Jang HJ, Suh PG, Lee YJ, Shin KJ, Cocco L, Chae YC. PLCγ1: Potential arbitrator of cancer progression. Adv Biol Regul 2018; 67:179-189. [PMID: 29174396 DOI: 10.1016/j.jbior.2017.11.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 06/07/2023]
Abstract
Phospholipase C (PLC) is an essential mediator of cellular signaling. PLC regulates multiple cellular processes by generating bioactive molecules such as inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG). These products propagate and regulate cellular signaling via calcium (Ca2+) mobilization and activation of protein kinase C (PKC), other kinases, and ion channels. PLCγ1, one of the primary subtypes of PLC, is directly activated by membrane receptors, including receptor tyrosine kinases (RTKs), and adhesion receptors such as integrin. PLCγ1 mediates signaling through direct interactions with other signaling molecules via SH domains, as well as its lipase activity. PLCγ1 is frequently enriched and mutated in various cancers, and is involved in the processes of tumorigenesis, including proliferation, migration, and invasion. Although many studies have suggested that PLCγ functions in cell mobility rather than proliferation in cancer, questions remain as to whether PLCγ regulates mitogenesis and whether PLCγ promotes or inhibits proliferation. Moreover, how PLCγ regulates cancer-associated cellular processes and the interplay among other proteins involved in cancer progression have yet to be fully elucidated. In this review, we discuss the current understanding of the role of PLCγ1 in cancer mobility and proliferation.
Collapse
Affiliation(s)
- Hyun-Jun Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yu Jin Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Kyeong Jin Shin
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| | - Young Chan Chae
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea.
| |
Collapse
|