1
|
Sakane F, Murakami C, Sakai H. Upstream and downstream pathways of diacylglycerol kinase : Novel phosphatidylinositol turnover-independent signal transduction pathways. Adv Biol Regul 2024:101054. [PMID: 39368888 DOI: 10.1016/j.jbior.2024.101054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Diacylglycerol kinase (DGK) phosphorylates diacylglycerol (DG) to produce phosphatidic acid (PA). Mammalian DGK comprise ten isozymes (α-κ) that regulate a wide variety of physiological and pathological events. Recently, we revealed that DGK isozymes use saturated fatty acid (SFA)/monosaturated fatty acid (MUFA)-containing and docosahexaenoic acid (22:6)-containing DG species, but not phosphatidylinositol (PI) turnover-derived 18:0/20:4-DG. For example, DGKδ, which is involved in the pathogenesis of type 2 diabetes, preferentially uses SFA/MUFA-containing DG species, such as 16:0/16:0- and 16:0/18:1-DG species, in high glucose-stimulated skeletal muscle cells. Moreover, DGKδ, which destabilizes the serotonin transporter (SERT) and regulates the serotonergic system in the brain, primarily generates 18:0/22:6-PA. Furthermore, 16:0/16:0-PA is produced by DGKζ in Neuro-2a cells during neuronal differentiation. We searched for SFA/MUFA-PA- and 18:0/22:6-PA-selective binding proteins (candidate downstream targets of DGKδ) and found that SFA/MUFA-PA binds to and activates the creatine kinase muscle type, an energy-metabolizing enzyme, and that 18:0/22:6-PA interacts with and activates Praja-1, an E3 ubiquitin ligase acting on SERT, and synaptojanin-1, a key player in the synaptic vesicle cycle. Next, we searched for SFA/MUFA-DG-generating enzymes upstream of DGKδ. We found that sphingomyelin synthase (SMS)1, SMS2, and SMS-related protein (SMSr) commonly act as phosphatidylcholine (PC)-phospholipase C (PLC) and phosphatidylethanolamine (PE)-PLC, generating SFA/MUFA-DG species, in addition to SMS and ceramide phosphoethanolamine synthase. Moreover, the orphan phosphatase PHOSPHO1 showed PC- and PE-PLC activities that produced SFA/MUFA-DG. Although PC- and PE-PLC activities were first described 70-35 years ago, their proteins and genes were not identified for a long time. We found that DGKδ interacts with SMSr and PHOSPHO1, and that DGKζ binds to SMS1 and SMSr. Taken together, these results strongly suggest that there are previously unrecognized signal transduction pathways that include DGK isozymes and generate and utilize SFA/MUFA-DG/PA or 18:0/22:6-DG/PA but not PI-turnover-derived 18:0/20:4-DG/PA.
Collapse
Affiliation(s)
- Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan.
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan
| | - Hiromichi Sakai
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| |
Collapse
|
2
|
Cheng HS, Chong YK, Lim EKY, Lee XY, Pang QY, Novera W, Marvalim C, Lee JXT, Ang BT, Tang C, Tan NS. Dual p38MAPK and MEK inhibition disrupts adaptive chemoresistance in mesenchymal glioblastoma to temozolomide. Neuro Oncol 2024; 26:1247-1261. [PMID: 38366847 PMCID: PMC11226874 DOI: 10.1093/neuonc/noae028] [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: 09/17/2022] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Precision treatment of glioblastoma is increasingly focused on molecular subtyping, with the mesenchymal subtype particularly resistant to temozolomide. Here, we aim to develop a targeted therapy for temozolomide resensitization in the mesenchymal subtype. METHODS We integrated kinomic profiles and kinase inhibitor screens from patient-derived proneural and mesenchymal glioma-propagating cells and public clinical datasets to identify key protein kinases implicated in temozolomide resistance. RNAseq, apoptosis assays, and comet assays were used to examine the role of p38MAPK signaling and adaptive chemoresistance in mesenchymal cells. The efficacy of dual p38MAPK and MEK/ERK inhibition using ralimetinib (selective orally active p38MAPK inhibitor; phase I/II for glioblastoma) and binimetinib (approved MEK1/2 inhibitor for melanoma; phase II for high-grade glioma) in primary and recurrent mesenchymal tumors was evaluated using an intracranial patient-derived tumor xenograft model, focusing on survival analysis. RESULTS Our transcriptomic-kinomic integrative analysis revealed p38MAPK as the prime target whose gene signature enables patient stratification based on their molecular subtypes and provides prognostic value. Repurposed p38MAPK inhibitors synergize favorably with temozolomide to promote intracellular retention of temozolomide and exacerbate DNA damage. Mesenchymal cells exhibit adaptive chemoresistance to p38MAPK inhibition through a pH-/calcium-mediated MEK/ERK pathway. Dual p38MAPK and MEK inhibition effectively maintain temozolomide sensitivity in primary and recurrent intracranial mesenchymal glioblastoma xenografts. CONCLUSIONS Temozolomide resistance in mesenchymal glioblastoma is associated with p38MAPK activation. Adaptive chemoresistance in p38MAPK-resistant cells is mediated by MEK/ERK signaling. Adjuvant therapy with dual p38MAPK and MEK inhibition prolongs temozolomide sensitivity, which can be developed into a precision therapy for the mesenchymal subtype.
Collapse
Affiliation(s)
- Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Yuk Kien Chong
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Eldeen Kai Yi Lim
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Xin Yi Lee
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Qing You Pang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Wisna Novera
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Charlie Marvalim
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Jeannie Xue Ting Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Beng Ti Ang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Carol Tang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Hart WS, Myers PJ, Purow BW, Lazzara MJ. Divergent transcriptomic signatures from putative mesenchymal stimuli in glioblastoma cells. Cancer Gene Ther 2024; 31:851-860. [PMID: 38337036 PMCID: PMC11192628 DOI: 10.1038/s41417-023-00724-w] [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: 04/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 02/12/2024]
Abstract
In glioblastoma, a mesenchymal phenotype is associated with especially poor patient outcomes. Various glioblastoma microenvironmental factors and therapeutic interventions are purported drivers of the mesenchymal transition, but the degree to which these cues promote the same mesenchymal transitions and the uniformity of those transitions, as defined by molecular subtyping systems, is unknown. Here, we investigate this question by analyzing publicly available patient data, surveying commonly measured transcripts for mesenchymal transitions in glioma-initiating cells (GIC), and performing next-generation RNA sequencing of GICs. Analysis of patient tumor data reveals that TGFβ, TNFα, and hypoxia signaling correlate with the mesenchymal subtype more than the proneural subtype. In cultured GICs, the microenvironment-relevant growth factors TGFβ and TNFα and the chemotherapeutic temozolomide promote expression of commonly measured mesenchymal transcripts. However, next-generation RNA sequencing reveals that growth factors and temozolomide broadly promote expression of both mesenchymal and proneural transcripts, in some cases with equal frequency. These results suggest that glioblastoma mesenchymal transitions do not occur as distinctly as in epithelial-derived cancers, at least as determined using common subtyping ontologies and measuring response to growth factors or chemotherapeutics. Further understanding of these issues may identify improved methods for pharmacologically targeting the mesenchymal phenotype in glioblastoma.
Collapse
Affiliation(s)
- William S Hart
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Paul J Myers
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Benjamin W Purow
- Department of Neurology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, USA.
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
4
|
Lee JY, Park J, Hong D. HSPA5 and FGFR1 genes in the mesenchymal subtype of glioblastoma can improve a treatment efficacy. Anim Cells Syst (Seoul) 2024; 28:216-227. [PMID: 38770056 PMCID: PMC11104699 DOI: 10.1080/19768354.2024.2347538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have emerged as a potential treatment strategy for glioblastoma multiforme (GBM). However, their efficacy is limited by various drug resistance mechanisms. To devise more effective treatments for GBM, genetic characteristics must be considered in addition to pre-existing treatments. We performed an integrative analysis with heterogeneous GBM datasets of genomic, transcriptomic, and proteomic data from DepMap, TCGA and CPTAC. We found that poor prognosis was induced by co-upregulation of heat shock protein family A member 5 (HSPA5) and fibroblast growth factor receptor 1 (FGFR1). Co-up regulation of these two genes could regulate the PI3K/AKT pathway. GBM cell lines with co-upregulation of these two genes showed higher drug sensitivity to PI3K inhibitors. In the mesenchymal subtype, the co-upregulation of FGFR1 and HSPA5 resulted in the most malignant subtype of GBM. Furthermore, we found this newly discovered subtype was correlated with homologous recombination deficiency (HRD) In conclusion, we discovered novel druggable candidates within the group exhibiting co-upregulation of these two genes in GBM, suggest potential strategies for combination therapy.
Collapse
Affiliation(s)
- Ju Young Lee
- Department of Biomedicine and Health, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jongkeun Park
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dongwan Hong
- Department of Biomedicine and Health, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- CMC Institute for Basic Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
5
|
Liu W, Wang H, Zhao Q, Tao C, Qu W, Hou Y, Huang R, Sun Z, Zhu G, Jiang X, Fang Y, Gao J, Wu X, Yang Z, Ping R, Chen J, Yang R, Chu T, Zhou J, Fan J, Tang Z, Yang D, Shi Y. Multiomics analysis reveals metabolic subtypes and identifies diacylglycerol kinase α (DGKA) as a potential therapeutic target for intrahepatic cholangiocarcinoma. Cancer Commun (Lond) 2024; 44:226-250. [PMID: 38143235 PMCID: PMC10876206 DOI: 10.1002/cac2.12513] [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: 07/04/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (iCCA) is a highly heterogeneous and lethal hepatobiliary tumor with few therapeutic strategies. The metabolic reprogramming of tumor cells plays an essential role in the development of tumors, while the metabolic molecular classification of iCCA is largely unknown. Here, we performed an integrated multiomics analysis and metabolic classification to depict differences in metabolic characteristics of iCCA patients, hoping to provide a novel perspective to understand and treat iCCA. METHODS We performed integrated multiomics analysis in 116 iCCA samples, including whole-exome sequencing, bulk RNA-sequencing and proteome analysis. Based on the non-negative matrix factorization method and the protein abundance of metabolic genes in human genome-scale metabolic models, the metabolic subtype of iCCA was determined. Survival and prognostic gene analyses were used to compare overall survival (OS) differences between metabolic subtypes. Cell proliferation analysis, 5-ethynyl-2'-deoxyuridine (EdU) assay, colony formation assay, RNA-sequencing and Western blotting were performed to investigate the molecular mechanisms of diacylglycerol kinase α (DGKA) in iCCA cells. RESULTS Three metabolic subtypes (S1-S3) with subtype-specific biomarkers of iCCA were identified. These metabolic subtypes presented with distinct prognoses, metabolic features, immune microenvironments, and genetic alterations. The S2 subtype with the worst survival showed the activation of some special metabolic processes, immune-suppressed microenvironment and Kirsten rat sarcoma viral oncogene homolog (KRAS)/AT-rich interactive domain 1A (ARID1A) mutations. Among the S2 subtype-specific upregulated proteins, DGKA was further identified as a potential drug target for iCCA, which promoted cell proliferation by enhancing phosphatidic acid (PA) metabolism and activating mitogen-activated protein kinase (MAPK) signaling. CONCLUSION Via multiomics analyses, we identified three metabolic subtypes of iCCA, revealing that the S2 subtype exhibited the poorest survival outcomes. We further identified DGKA as a potential target for the S2 subtype.
Collapse
Affiliation(s)
- Weiren Liu
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Huqiang Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingP. R. China
| | - Qianfu Zhao
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Chenyang Tao
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Weifeng Qu
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Yushan Hou
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingP. R. China
| | - Run Huang
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Zimei Sun
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingP. R. China
| | - Guiqi Zhu
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Xifei Jiang
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Yuan Fang
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Jun Gao
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Xiaoling Wu
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Zhixiang Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingP. R. China
| | - Rongyu Ping
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingP. R. China
| | - Jiafeng Chen
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Rui Yang
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Tianhao Chu
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Jian Zhou
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Jia Fan
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Zheng Tang
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Dong Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijingP. R. China
| | - Yinghong Shi
- Department of Liver Surgery and TransplantationLiver Cancer Institute, Zhongshan HospitalFudan UniversityKey Laboratory of Carcinogenesis and Cancer Invasion of Ministry of EducationShanghaiP. R. China
- Research Unit of Liver cancer Recurrence and Metastasis, Chinese Academy of Medical SciencesBeijingP. R. China
| |
Collapse
|
6
|
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
|
7
|
Tan J, Zhong M, Hu Y, Pan G, Yao J, Tang Y, Duan H, Jiang Y, Shan W, Lin J, Liu Y, Huang J, Zheng H, Zhou Y, Fu G, Li Z, Xu B, Zha J. Ritanserin suppresses acute myeloid leukemia by inhibiting DGKα to downregulate phospholipase D and the Jak-Stat/MAPK pathway. Discov Oncol 2023; 14:118. [PMID: 37392305 DOI: 10.1007/s12672-023-00737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023] Open
Abstract
Refractory or relapsed (R/R) AML is the most challenging form of AML to treat. Due to frequent genetic mutations, therapy alternatives are limited. Here, we identified the role of ritanserin and its target DGKα in AML. Several AML cell lines and primary patient cells were treated with ritanserin and subjected to cell proliferation, apoptosis and gene analyses with CCK-8 assay, Annexin V/PI assay and Western blotting, respectively. We also evaluated the function of the ritanserin target diacylglycerol kinase alpha (DGKα) in AML by bioinformatics. In vitro experiments have revealed that ritanserin inhibits AML progression in a dose- and time-dependent manner, and it shows an anti-AML effect in xenograft mouse models. We further demonstrated that the expression of DGKα was elevated in AML and correlated with poor survival. Mechanistically, ritanserin negatively regulates SphK1 expression through PLD signaling, also inhibiting the Jak-Stat and MAPK signaling pathways via DGKα. These findings suggest that DGKα may be an available therapeutic target and provide effective preclinical evidence of ritanserin as a promising treatment for AML.
Collapse
Affiliation(s)
- Jinshui Tan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Guangchao Pan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jingwei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuanfang Tang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Hongpeng Duan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Weihang Shan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jiaqi Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Yating Liu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Jiewen Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361002, Fujian, China
| | - Huijian Zheng
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Yong Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Innovation Center for Cell Biology, Xiamen University, Xiamen, 361002, Fujian, China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China.
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, Fujian, People's Republic of China.
| |
Collapse
|
8
|
Gravina T, Boggio CMT, Gorla E, Racca L, Polidoro S, Centonze S, Ferrante D, Lunghi M, Graziani A, Corà D, Baldanzi G. Role of Diacylglycerol Kinases in Acute Myeloid Leukemia. Biomedicines 2023; 11:1877. [PMID: 37509516 PMCID: PMC10377028 DOI: 10.3390/biomedicines11071877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Diacylglycerol kinases (DGKs) play dual roles in cell transformation and immunosurveillance. According to cancer expression databases, acute myeloid leukemia (AML) exhibits significant overexpression of multiple DGK isoforms, including DGKA, DGKD and DGKG, without a precise correlation with specific AML subtypes. In the TGCA database, high DGKA expression negatively correlates with survival, while high DGKG expression is associated with a more favorable prognosis. DGKA and DGKG also feature different patterns of co-expressed genes. Conversely, the BeatAML and TARGET databases show that high DGKH expression is correlated with shorter survival. To assess the suitability of DGKs as therapeutic targets, we treated HL-60 and HEL cells with DGK inhibitors and compared cell growth and survival with those of untransformed lymphocytes. We observed a specific sensitivity to R59022 and R59949, two poorly selective inhibitors, which promoted cytotoxicity and cell accumulation in the S phase in both cell lines. Conversely, the DGKA-specific inhibitors CU-3 and AMB639752 showed poor efficacy. These findings underscore the pivotal and isoform-specific involvement of DGKs in AML, offering a promising pathway for the identification of potential therapeutic targets. Notably, the DGKA and DGKH isoforms emerge as relevant players in AML pathogenesis, albeit DGKA inhibition alone seems insufficient to impair AML cell viability.
Collapse
Affiliation(s)
- Teresa Gravina
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Chiara Maria Teresa Boggio
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Elisa Gorla
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Luisa Racca
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Silvia Polidoro
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Sara Centonze
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Ferrante
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Monia Lunghi
- Division of Hematology, Department of Translational Medicine, University of Piemonte Orientale, 28110 Novara, Italy
| | - Andrea Graziani
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC), University of Turin, 10124 Turin, Italy
| | - Davide Corà
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Gianluca Baldanzi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
9
|
Sharma G, Braga MC, Da Pieve C, Szopa W, Starzetz T, Plate KH, Kaspera W, Kramer-Marek G. Immuno-PET Imaging of Tumour PD-L1 Expression in Glioblastoma. Cancers (Basel) 2023; 15:3131. [PMID: 37370741 DOI: 10.3390/cancers15123131] [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: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
There is no established method to assess the PD-L1 expression in brain tumours. Therefore, we investigated the suitability of affibody molecule (ZPD-L1) radiolabelled with F-18 (Al18F) and Ga-68 to measure the expression of PD-L1 in xenograft mouse models of GBM. Mice bearing subcutaneous and orthotopic tumours were imaged 1 h post-radioconjugate administration. Ex vivo biodistribution studies and immunohistochemistry (IHC) staining were performed. Tumoural PD-L1 expression and CD4+/CD8+ tumour-infiltrating lymphocytes were evaluated in human GBM specimens. ZPD-L1 was radiolabelled with radiochemical yields of 32.2 ± 4.4% (F-18) and 73.3 ± 1.8% (Ga-68). The cell-associated radioactivity in vitro was consistent with PD-L1 expression levels assessed with flow cytometry. In vivo imaging demonstrated that 18F-AlF-NOTA-ZPD-L1 can distinguish between PD-L1 high-expressing tumours (U87-MGvIII) and PD-L1-negative ones (H292PD-L1Ko). The radioconjugate was quickly cleared from the blood and normal tissues, allowing for high-contrast images of brain tumours as early as 1 h post-injection. 68Ga-NOTA-ZPD-L1 showed heterogeneous and diffuse accumulation that corresponded to the extensively infiltrating GCGR-E55 tumours involving contiguous lobes of the brain. Lastly, 39% of analysed GBM patient samples showed PD-L1+ staining of tumour cells that was associated with elevated levels of CD4+ and CD8+ lymphocytes. Our results suggest that the investigated radioconjugates are very promising agents with the potential to facilitate the future design of treatment regimens for GBM patients.
Collapse
Affiliation(s)
- Gitanjali Sharma
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London SW7 3RP, UK
| | - Marta C Braga
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London SW7 3RP, UK
| | - Chiara Da Pieve
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London SW7 3RP, UK
| | - Wojciech Szopa
- Department of Neurosurgery, Medical University of Silesia, 41-200 Sosnowiec, Poland
| | - Tatjana Starzetz
- Edinger Institute, Institute of Neurology, 60528 Frankfurt, Germany
- German Consortium for Translational Cancer Research, DKTK, 69120 Heidelberg, Germany
| | - Karl H Plate
- Edinger Institute, Institute of Neurology, 60528 Frankfurt, Germany
- German Consortium for Translational Cancer Research, DKTK, 69120 Heidelberg, Germany
| | - Wojciech Kaspera
- Department of Neurosurgery, Medical University of Silesia, 41-200 Sosnowiec, Poland
| | - Gabriela Kramer-Marek
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London SW7 3RP, UK
| |
Collapse
|
10
|
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
|
11
|
Cammarata FP, Torrisi F, Vicario N, Bravatà V, Stefano A, Salvatorelli L, D'Aprile S, Giustetto P, Forte GI, Minafra L, Calvaruso M, Richiusa S, Cirrone GAP, Petringa G, Broggi G, Cosentino S, Scopelliti F, Magro G, Porro D, Libra M, Ippolito M, Russo G, Parenti R, Cuttone G. Proton boron capture therapy (PBCT) induces cell death and mitophagy in a heterotopic glioblastoma model. Commun Biol 2023; 6:388. [PMID: 37031346 PMCID: PMC10082834 DOI: 10.1038/s42003-023-04770-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 03/28/2023] [Indexed: 04/10/2023] Open
Abstract
Despite aggressive therapeutic regimens, glioblastoma (GBM) represents a deadly brain tumor with significant aggressiveness, radioresistance and chemoresistance, leading to dismal prognosis. Hypoxic microenvironment, which characterizes GBM, is associated with reduced therapeutic effectiveness. Moreover, current irradiation approaches are limited by uncertain tumor delineation and severe side effects that comprehensively lead to unsuccessful treatment and to a worsening of the quality of life of GBM patients. Proton beam offers the opportunity of reduced side effects and a depth-dose profile, which, unfortunately, are coupled with low relative biological effectiveness (RBE). The use of radiosensitizing agents, such as boron-containing molecules, enhances proton RBE and increases the effectiveness on proton beam-hit targets. We report a first preclinical evaluation of proton boron capture therapy (PBCT) in a preclinical model of GBM analyzed via μ-positron emission tomography/computed tomography (μPET-CT) assisted live imaging, finding a significant increased therapeutic effectiveness of PBCT versus proton coupled with an increased cell death and mitophagy. Our work supports PBCT and radiosensitizing agents as a scalable strategy to treat GBM exploiting ballistic advances of proton beam and increasing therapeutic effectiveness and quality of life in GBM patients.
Collapse
Affiliation(s)
- Francesco Paolo Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, Catania, Italy
| | - Filippo Torrisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Molecular Preclinical and Translational Imaging Research Center - IMPRonTe, University of Catania, Catania, Italy
| | - Valentina Bravatà
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | - Lucia Salvatorelli
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele" Anatomic Pathology, University of Catania, Catania, Italy
| | - Simona D'Aprile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Pierangela Giustetto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giusi Irma Forte
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | - Luigi Minafra
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | - Marco Calvaruso
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | - Selene Richiusa
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | | | - Giada Petringa
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, Catania, Italy
| | - Giuseppe Broggi
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele" Anatomic Pathology, University of Catania, Catania, Italy
| | | | - Fabrizio Scopelliti
- Radiopharmacy Laboratory Nuclear Medicine Department, Cannizzaro Hospital, Catania, Italy
| | - Gaetano Magro
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele" Anatomic Pathology, University of Catania, Catania, Italy
| | - Danilo Porro
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Massimo Ippolito
- Nuclear Medicine Department, Cannizzaro Hospital, Catania, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, Cefalù, Italy.
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, Catania, Italy.
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
- Molecular Preclinical and Translational Imaging Research Center - IMPRonTe, University of Catania, Catania, Italy.
| | - Giacomo Cuttone
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, Catania, Italy
| |
Collapse
|
12
|
Zhou D, Liu T, Rao X, Jie X, Chen Y, Wu Z, Deng H, Zhang D, Wang J, Wu G. Targeting diacylglycerol kinase α impairs lung tumorigenesis by inhibiting cyclin D3. Thorac Cancer 2023; 14:1179-1191. [PMID: 36965165 PMCID: PMC10151139 DOI: 10.1111/1759-7714.14851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Diacylglycerol kinase α (DGKA) is the first member discovered from the diacylglycerol kinase family, and it has been linked to the progression of various types of tumors. However, it is unclear whether DGKA is linked to the development of lung cancer. METHODS We investigated the levels of DGKA in the lung cancer tissues. Cell growth assay, colony formation assay and EdU assay were used to examine the effects of DGKA-targeted siRNAs/shRNAs/drugs on the proliferation of lung cancer cells in vitro. Xenograft mouse model was used to investigate the role of DGKA inhibitor ritanserin on the proliferation of lung cancer cells in vivo. The downstream target of DGKA in lung tumorigenesis was identified by RNA sequencing. RESULTS DGKA is upregulated in the lung cancer cells. Functional assays and xenograft mouse model indicated that the proliferation ability of lung cancer cells was impaired after inhibiting DGKA. And cyclin D3(CCND3) is the downstream target of DGKA promoting lung cancer. CONCLUSIONS Our study demonstrated that DGKA promotes lung tumorigenesis by regulating the CCND3 expression and hence it can be considered as a potential molecular biomarker to evaluate the prognosis of lung cancer patients. What's more, we also demonstrated the efficacy of ritanserin as a promising new medication for treating lung cancer.
Collapse
Affiliation(s)
- Dong Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinrui Rao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Jie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunshang Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huilin Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
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
|
14
|
Duan J, Zhang Z, Chen Y, Zhao Y, Sun Q, Wang W, Zheng H, Liang D, Cheng J, Yan J, Li ZC. Imaging phenotypes from MRI for the prediction of glioma immune subtypes from RNA sequencing: A multicenter study. Mol Oncol 2023; 17:629-646. [PMID: 36688633 PMCID: PMC10061289 DOI: 10.1002/1878-0261.13380] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/23/2022] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Tumor subtyping based on its immune landscape may guide precision immunotherapy. The aims of this study were to identify immune subtypes of adult diffuse gliomas with RNA sequencing data, and to noninvasively predict this subtype using a biologically interpretable radiomic signature from MRI. A subtype discovery dataset (n = 210) from a public database and two radiogenomic datasets (n = 130 and 55, respectively) from two local hospitals were included. Brain tumor microenvironment-specific signatures were constructed from RNA sequencing to identify the immune types. A radiomic signature was built from MRI to predict the identified immune subtypes. The pathways underlying the radiomic signature were identified to annotate their biological meanings. The reproducibility of the findings was verified externally in multicenter datasets. Three distinctive immune subtypes were identified, including an inflamed subtype marked by elevated hypoxia-induced immunosuppression, a "cold" subtype that exhibited scarce immune infiltration with downregulated antigen presentation, and an intermediate subtype that showed medium immune infiltration. A 10-feature radiomic signature was developed to predict immune subtypes, achieving an AUC of 0.924 in the validation dataset. The radiomic features correlated with biological functions underpinning immune suppression, which substantiated the hypothesis that molecular changes can be reflected by radiomic features. The immune subtypes, predictive radiomic signature, and radiomics-correlated biological pathways were validated externally. Our data suggest that adult-type diffuse gliomas harbor three distinctive immune subtypes that can be predicted by MRI radiomic features with clear biological significance. The immune subtypes, radiomic signature, and radiogenomic links can be replicated externally.
Collapse
Affiliation(s)
- Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hairong Zheng
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong Liang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,National Innovation Center for Advanced Medical Devices, Shenzhen, China.,Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, China
| |
Collapse
|
15
|
Li J, Wang X, Chen L, Zhang J, Zhang Y, Ren X, Sun J, Fan X, Fan J, Li T, Tong L, Yi L, Chen L, Liu J, Shang G, Ren X, Zhang H, Yu S, Ming H, Huang Q, Dong J, Zhang C, Yang X. TMEM158 promotes the proliferation and migration of glioma cells via STAT3 signaling in glioblastomas. Cancer Gene Ther 2022; 29:1117-1129. [PMID: 34992215 PMCID: PMC9395270 DOI: 10.1038/s41417-021-00414-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/20/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022]
Abstract
Glioblastoma is the most common primary intracranial malignant tumor in adults and has high morbidity and high mortality. TMEM158 has been reported to promote the progression of solid tumors. However, its potential role in glioma is still unclear. Here, we found that TMEM158 expression in human glioma cells in the tumor core was significantly higher than that in noncancerous cells at the tumor edge using bioinformatics analysis. Cancer cells in patients with primary GBMs harbored significantly higher expression of TMEM158 than those in patients with WHO grade II or III gliomas. Interestingly, regardless of tumor grading, human glioma samples that were IDH1-wild-type (IDH1-WT) exhibited higher expression of TMEM158 than those with IDH1-mutant (IDH1-Mut). We also illustrated that TMEM158 mRNA expression was correlated with poor overall survival in glioma patients. Furthermore, we demonstrated that silencing TMEM158 inhibited the proliferation of glioma cells and that TMEM158 overexpression promoted the migration and invasion of glioma cells by stimulating the EMT process. We found that the underlying mechanism involves STAT3 activation mediating TMEM158-driven glioma progression. In vivo results further confirmed the inhibitory effect of the TMEM158 downregulation on glioma growth. Collectively, these findings further our understanding of the oncogenic function of TMEM158 in gliomas, which represents a potential therapeutic target, especially for GBMs.
Collapse
Affiliation(s)
- Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xuya Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Lulu Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jinhao Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Yiming Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xiao Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jinzhang Sun
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xiaoguang Fan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jikang Fan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Luqing Tong
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Yi
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jie Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Guanjie Shang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Xiude Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Hao Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Haolang Ming
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China.
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China.
| |
Collapse
|
16
|
Li D, Liang J, Yang W, Guo W, Song W, Zhang W, Wu X, He B. A distinct lipid metabolism signature of acute myeloid leukemia with prognostic value. Front Oncol 2022; 12:876981. [PMID: 35957912 PMCID: PMC9359125 DOI: 10.3389/fonc.2022.876981] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy characterized by extensive genetic abnormalities that might affect the prognosis and provide potential drug targets for treatment. Reprogramming of lipid metabolism plays important roles in tumorigenesis and progression and has been newly recognized a new hallmark of malignancy, and some related molecules in the signal pathways could be prognostic biomarkers and potential therapeutic targets for cancer treatment. However, the clinical value of lipid metabolism reprogramming in AML has not been systematically explored. In this study, we aim to explore the clinical value of lipid metabolism reprogramming and develop a prognostic risk signature for AML. Methods We implemented univariate Cox regression analysis to identify the prognosis-related lipid metabolism genes, and then performed LASSO analysis to develop the risk signature with six lipid metabolism-related genes (LDLRAP1, PNPLA6, DGKA, PLA2G4A, CBR1, and EBP). The risk scores of samples were calculated and divided into low- and high-risk groups by the median risk score. Results Survival analysis showed the high-risk group hold the significantly poorer outcomes than the low-risk group. The signature was validated in the GEO datasets and displayed a robust prognostic value in the stratification analysis. Multivariate analysis revealed the signature was an independent prognostic factor for AML patients and could serve as a potential prognostic biomarker in clinical evaluation. Furthermore, the risk signature was also found to be closely related to immune landscape and immunotherapy response in AML. Conclusions Overall, we conducted a comprehensive analysis of lipid metabolism in AML and constructed a risk signature with six genes related to lipid metabolism for the malignancy, prognosis, and immune landscape of AML, and our study might contribute to better understanding in the use of metabolites and metabolic pathways as the potential prognostic biomarkers and therapeutic targets for AML.
Collapse
Affiliation(s)
- Ding Li
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiaming Liang
- Department of Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Yang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenbin Guo
- Department of Pathology, Pingtan Comprehensive Experimental Area Hospital, Fuzhou, China
| | - Wenping Song
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xuan Wu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- *Correspondence: Baoxia He, ; Xuan Wu,
| | - Baoxia He
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Baoxia He, ; Xuan Wu,
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Granade ME, Manigat LC, Lemke MC, Purow BW, Harris TE. Identification of ritanserin analogs that display DGK isoform specificity. Biochem Pharmacol 2022; 197:114908. [PMID: 34999054 PMCID: PMC8858877 DOI: 10.1016/j.bcp.2022.114908] [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: 11/08/2021] [Revised: 12/28/2021] [Accepted: 01/01/2022] [Indexed: 11/15/2022]
Abstract
The diacylglycerol kinase (DGK) family of lipid enzymes catalyzes the conversion of diacylglycerol (DAG) to phosphatidic acid (PA). Both DAG and PA are lipid signaling molecules that are of notable importance in regulating cell processes such as proliferation, apoptosis, and migration. There are ten mammalian DGK enzymes that appear to have distinct biological functions. DGKα has emerged as a promising therapeutic target in numerous cancers including glioblastoma (GBM) and melanoma as treatment with small molecule DGKα inhibitors results in reduced tumor sizes and prolonged survival. Importantly, DGKα has also been identified as an immune checkpoint due to its promotion of T cell anergy, and its inhibition has been shown to improve T cell activation. There are few small molecule DGKα inhibitors currently available, and the application of existing compounds to clinical settings is hindered by species-dependent variability in potency, as well as concerns regarding isotype specificity particularly amongst other type I DGKs. In order to resolve these issues, we have screened a library of compounds structurally analogous to the DGKα inhibitor, ritanserin, in an effort to identify more potent and specific alternatives. We identified two compounds that more potently and selectively inhibit DGKα, one of which (JNJ-3790339) demonstrates similar cytotoxicity in GBM and melanoma cells as ritanserin. Consistent with its inhibitor profile towards DGKα, JNJ-3790339 also demonstrated improved activation of T cells compared with ritanserin. Together our data support efforts to identify DGK isoform-selective inhibitors as a mechanism to produce pharmacologically relevant cancer therapies.
Collapse
Affiliation(s)
- Mitchell E Granade
- University of Virginia, School of Medicine, Department of Pharmacology, Charlottesville, VA, United States
| | - Laryssa C Manigat
- University of Virginia, School of Medicine, Department of Pathology, Charlottesville, VA, United States
| | - Michael C Lemke
- University of Virginia, School of Medicine, Department of Pharmacology, Charlottesville, VA, United States
| | - Benjamin W Purow
- University of Virginia, Department of Neurology, Division of Neuro-Oncology, Charlottesville, VA, United States.
| | - Thurl E Harris
- University of Virginia, School of Medicine, Department of Pharmacology, Charlottesville, VA, United States.
| |
Collapse
|
20
|
Purow B. Delivering Glioblastoma a Kick-DGKα Inhibition as a Promising Therapeutic Strategy for GBM. Cancers (Basel) 2022; 14:cancers14051269. [PMID: 35267577 PMCID: PMC8909282 DOI: 10.3390/cancers14051269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 11/16/2022] Open
Abstract
Diacylglycerol kinase α (DGKα) inhibition may be particularly relevant for the treatment of glioblastoma (GBM), a relatively common brain malignancy incurable with current therapies. Prior reports have shown that DGKα inhibition has multiple direct activities against GBM cells, including suppressing the oncogenic pathways mTOR and HIF-1α. It also inhibits pathways associated with the normally treatment-resistant mesenchymal phenotype, yielding preferential activity against mesenchymal GBM; this suggests possible utility in combining DGKα inhibition with radiation and other therapies for which the mesenchymal phenotype promotes resistance. The potential for DGKα inhibition to block or reverse T cell anergy also suggests the potential of DGKα inhibition to boost immunotherapy against GBM, which is generally considered an immunologically "cold" tumor. A recent report indicates that DGKα deficiency increases responsiveness of macrophages, indicating that DGKα inhibition could also have the potential to boost macrophage and microglia activity against GBM-which could be a particularly promising approach given the heavy infiltration of these cells in GBM. DGKα inhibition may therefore offer a promising multi-pronged attack on GBM, with multiple direct anti-GBM activities and also the ability to boost both adaptive and innate immune responses against GBM. However, both the direct and indirect benefits of DGKα inhibition for GBM will likely require combinations with other therapies to achieve meaningful efficacy. Furthermore, GBM offers other challenges for the application of DGKα inhibitors, including decreased accessibility from the blood-brain barrier (BBB). The ideal DGKα inhibitor for GBM will combine potency, specificity, and BBB penetrability. No existing inhibitor is known to meet all these criteria, but the strong potential of DGKα inhibition against this lethal brain cancer should help drive development and testing of agents to bring this promising strategy to the clinic for patients with GBM.
Collapse
Affiliation(s)
- Benjamin Purow
- Neurology Department, University of Virginia, Charlottesville, VA 22904, USA
| |
Collapse
|
21
|
Lv SQ, Fu Z, Yang L, Li QR, Zhu J, Gai QJ, Mao M, He J, Qin Y, Yao XX, Lan X, Wang YX, Lu HM, Xiang Y, Zhang ZX, Huang GH, Yang W, Kang P, Sun Z, Shi Y, Yao XH, Bian XW, Wang Y. Comprehensive omics analyses profile genesets related with tumor heterogeneity of multifocal glioblastomas and reveal LIF/CCL2 as biomarkers for mesenchymal subtype. Theranostics 2022; 12:459-473. [PMID: 34987659 PMCID: PMC8690928 DOI: 10.7150/thno.65739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/27/2021] [Indexed: 01/22/2023] Open
Abstract
Rationale: Around 10%-20% patients with glioblastoma (GBM) are diagnosed with more than one tumor lesions or multifocal GBM (mGBM). However, the understanding on genetic, DNA methylomic, and transcriptomic characteristics of mGBM is still limited. Methods: In this study, we collected nine tumor foci from three mGBM patients followed by whole genome sequencing, whole genome bisulfite sequencing, RNA sequencing, and immunohistochemistry. The data were further examined using public GBM databases and GBM cell line. Results: Analysis on genetic data confirmed common features of GBM, including gain of chr.7 and loss of chr.10, loss of critical tumor suppressors, high frequency of PDGFA and EGFR amplification. Through profiling DNA methylome of individual tumor foci, we found that promoter methylation status of genes involved in detection of chemical stimulus, immune response, and Hippo/YAP1 pathway was significantly changed in mGBM. Although both CNV and promoter methylation alteration were involved in heterogeneity of different tumor foci from same patients, more CNV events than promoter hypomethylation events were shared by different tumor foci, implying CNV were relatively earlier than promoter methylation alteration during evolution of different tumor foci from same mGBM. Moreover, different tumor foci from same mGBM assumed different molecular subtypes and mesenchymal subtype was prevalent in mGBM, which might explain the worse prognosis of mGBM than single GBM. Interestingly, we noticed that LIF and CCL2 was tightly correlated with mesenchymal subtype tumor focus in mGBM and predicted poor survival of GBM patients. Treatment with LIF and CCL2 produced mesenchymal-like transcriptome in GBM cells. Conclusions: Together, our work herein comprehensively profiled multi-omics features of mGBM and emphasized that components of extracellular microenvironment, such as LIF and CCL2, contributed to the evolution and prognosis of tumor foci in mGBM patients.
Collapse
|
22
|
Sakane F, Hoshino F, Ebina M, Sakai H, Takahashi D. The Roles of Diacylglycerol Kinase α in Cancer Cell Proliferation and Apoptosis. Cancers (Basel) 2021; 13:cancers13205190. [PMID: 34680338 PMCID: PMC8534027 DOI: 10.3390/cancers13205190] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary Diacylglycerol (DG) kinase (DGK) phosphorylates DG to generate phosphatidic acid (PA). DGKα is highly expressed in several refractory cancer cells, including melanoma, hepatocellular carcinoma, and glioblastoma cells, attenuates apoptosis, and promotes proliferation. In cancer cells, PA produced by DGKα plays an important role in proliferation/antiapoptosis. In addition to cancer cells, DGKα is highly abundant in T cells and induces a nonresponsive state (anergy), representing the main mechanism by which advanced cancers avoid immune action. In T cells, DGKα induces anergy through DG consumption. Therefore, a DGKα-specific inhibitor is expected to be a dual effective anticancer treatment that inhibits cancer cell proliferation and simultaneously activates T cell function. Moreover, the inhibition of DGKα synergistically enhances the anticancer effects of programmed cell death-1/programmed cell death ligand 1 blockade. Taken together, DGKα inhibition provides a promising new treatment strategy for refractory cancers. Abstract Diacylglycerol (DG) kinase (DGK) phosphorylates DG to generate phosphatidic acid (PA). The α isozyme is activated by Ca2+ through its EF-hand motifs and tyrosine phosphorylation. DGKα is highly expressed in several refractory cancer cells including melanoma, hepatocellular carcinoma, and glioblastoma cells. In melanoma cells, DGKα is an antiapoptotic factor that activates nuclear factor-κB (NF-κB) through the atypical protein kinase C (PKC) ζ-mediated phosphorylation of NF-κB. DGKα acts as an enhancer of proliferative activity through the Raf–MEK–ERK pathway and consequently exacerbates hepatocellular carcinoma progression. In glioblastoma and melanoma cells, DGKα attenuates apoptosis by enhancing the phosphodiesterase (PDE)-4A1–mammalian target of the rapamycin pathway. As PA activates PKCζ, Raf, and PDE, it is likely that PA generated by DGKα plays an important role in the proliferation/antiapoptosis of cancer cells. In addition to cancer cells, DGKα is highly abundant in T cells and induces a nonresponsive state (anergy), which represents the main mechanism by which advanced cancers escape immune action. In T cells, DGKα attenuates the activity of Ras-guanyl nucleotide-releasing protein, which is activated by DG and avoids anergy through DG consumption. Therefore, a DGKα-specific inhibitor is expected to be a dual effective anticancer treatment that inhibits cancer cell proliferation and simultaneously enhances T cell functions. Moreover, the inhibition of DGKα synergistically enhances the anticancer effects of programmed cell death-1/programmed cell death ligand 1 blockade. Taken together, DGKα inhibition provides a promising new treatment strategy for refractory cancers.
Collapse
Affiliation(s)
- Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan; (F.H.); (M.E.)
- Correspondence: ; Tel.: +81-43-290-3695
| | - Fumi Hoshino
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan; (F.H.); (M.E.)
| | - Masayuki Ebina
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan; (F.H.); (M.E.)
| | - Hiromichi Sakai
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo 693-8501, Japan;
| | - Daisuke Takahashi
- Department of Pharmaceutical Health Care and Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| |
Collapse
|
23
|
Giakoumettis D, Sgouros S. Nanotechnology in neurosurgery: a systematic review. Childs Nerv Syst 2021; 37:1045-1054. [PMID: 33462733 DOI: 10.1007/s00381-020-05008-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND The application of nanotechnology in medicine encompasses an interdisciplinary field of sciences for the diagnosis, treatment, and monitoring of medical conditions. This study aims to systematically review and summarize the advances of nanotechnology applicable to neurosurgery. METHODS We performed a PubMed advanced search of reports exploring the advances of nanotechnology and nanomedicine relating to diagnosis, treatment, or both, in neurosurgery, for the last decade. The search was performed according to PRISMA guidelines, and the following data were extracted from each paper: title; authors; article type; PMID; DOI; year of publication; in vitro, in vivo model; nanomedical, nanotechnological material; nanofield; neurosurgical field; the application of the system; and main conclusions of the study. RESULTS A total of 78 original studies were included in this review. The results were organized into the following categories: functional neurosurgery, head trauma, neurodegenerative diseases, neuro-oncology, spinal surgery and peripheral nerves, vascular neurosurgery, and studies that apply to more than one field. A further categorization applied in terms of nanomedical field such as neuroimaging, neuro-nanotechnology, neuroregeneration, theranostics, and neuro-nanotherapy. CONCLUSION In reviewing the literature, significant advances in imaging and treatment of central nervous system diseases are underway and are expected to reach clinical practice in the next decade by the application of the rapidly evolving nanotechnology techniques.
Collapse
Affiliation(s)
- Dimitrios Giakoumettis
- Department of Neurosurgery, "Evangelismos" Hospital, Athens, Greece
- Department of Neurosurgery, Medical School, National and "Kapodistrian" University of Athens, Athens, Greece
| | - Spyros Sgouros
- Department of Neurosurgery, Medical School, National and "Kapodistrian" University of Athens, Athens, Greece.
- Department of Pediatric Neurosurgery, "Iaso" Children's Hospital, Kifisias Avenue 37-39, 151 23 Marousi, Athens, Greece.
| |
Collapse
|
24
|
Kovalenko A, Sanin A, Kosmas K, Zhang L, Wang J, Akl EW, Giannikou K, Probst CK, Hougard TR, Rue RW, Krymskaya VP, Asara JM, Lam HC, Kwiatkowski DJ, Henske EP, Filippakis H. Therapeutic Targeting of DGKA-Mediated Macropinocytosis Leads to Phospholipid Reprogramming in Tuberous Sclerosis Complex. Cancer Res 2021; 81:2086-2100. [PMID: 33593821 DOI: 10.1158/0008-5472.can-20-2218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/16/2020] [Accepted: 02/10/2021] [Indexed: 11/16/2022]
Abstract
Lymphangioleiomyomatosis is a rare destructive lung disease affecting primarily women and is the primary lung manifestation of tuberous sclerosis complex (TSC). In lymphangioleiomyomatosis, biallelic loss of TSC1/2 leads to hyperactivation of mTORC1 and inhibition of autophagy. To determine how the metabolic vulnerabilities of TSC2-deficient cells can be targeted, we performed a high-throughput screen utilizing the "Repurposing" library at the Broad Institute of MIT and Harvard (Cambridge, MA), with or without the autophagy inhibitor chloroquine. Ritanserin, an inhibitor of diacylglycerol kinase alpha (DGKA), was identified as a selective inhibitor of proliferation of Tsc2-/- mouse embryonic fibroblasts (MEF), with no impact on Tsc2+/+ MEFs. DGKA is a lipid kinase that metabolizes diacylglycerol to phosphatidic acid, a key component of plasma membranes. Phosphatidic acid levels were increased 5-fold in Tsc2-/- MEFs compared with Tsc2+/+ MEFs, and treatment of Tsc2-/- MEFs with ritanserin led to depletion of phosphatidic acid as well as rewiring of phospholipid metabolism. Macropinocytosis is known to be upregulated in TSC2-deficient cells. Ritanserin decreased macropinocytic uptake of albumin, limited the number of lysosomes, and reduced lysosomal activity in Tsc2-/- MEFs. In a mouse model of TSC, ritanserin treatment decreased cyst frequency and volume, and in a mouse model of lymphangioleiomyomatosis, genetic downregulation of DGKA prevented alveolar destruction and airspace enlargement. Collectively, these data indicate that DGKA supports macropinocytosis in TSC2-deficient cells to maintain phospholipid homeostasis and promote proliferation. Targeting macropinocytosis with ritanserin may represent a novel therapeutic approach for the treatment of TSC and lymphangioleiomyomatosis. SIGNIFICANCE: This study identifies macropinocytosis and phospholipid metabolism as novel mechanisms of metabolic homeostasis in mTORC1-hyperactive cells and suggest ritanserin as a novel therapeutic strategy for use in mTORC1-hyperactive tumors, including pancreatic cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2086/F1.large.jpg.
Collapse
Affiliation(s)
- Andrii Kovalenko
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andres Sanin
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kosmas Kosmas
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Long Zhang
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ji Wang
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elie W Akl
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Krinio Giannikou
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Clemens K Probst
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas R Hougard
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ryan W Rue
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vera P Krymskaya
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hilaire C Lam
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David J Kwiatkowski
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Harilaos Filippakis
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
25
|
Potential role of diacylglycerol kinases in immune-mediated diseases. Clin Sci (Lond) 2021; 134:1637-1658. [PMID: 32608491 DOI: 10.1042/cs20200389] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
The mechanism promoting exacerbated immune responses in allergy and autoimmunity as well as those blunting the immune control of cancer cells are of primary interest in medicine. Diacylglycerol kinases (DGKs) are key modulators of signal transduction, which blunt diacylglycerol (DAG) signals and produce phosphatidic acid (PA). By modulating lipid second messengers, DGK modulate the activity of downstream signaling proteins, vesicle trafficking and membrane shape. The biological role of the DGK α and ζ isoforms in immune cells differentiation and effector function was subjected to in deep investigations. DGK α and ζ resulted in negatively regulating synergistic way basal and receptor induced DAG signals in T cells as well as leukocytes. In this way, they contributed to keep under control the immune response but also downmodulate immune response against tumors. Alteration in DGKα activity is also implicated in the pathogenesis of genetic perturbations of the immune function such as the X-linked lymphoproliferative disease 1 and localized juvenile periodontitis. These findings suggested a participation of DGK to the pathogenetic mechanisms underlying several immune-mediated diseases and prompted several researches aiming to target DGK with pharmacologic and molecular strategies. Those findings are discussed inhere together with experimental applications in tumors as well as in other immune-mediated diseases such as asthma.
Collapse
|
26
|
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
|
27
|
Azam Z, TO ST, Tannous BA. Mesenchymal Transformation: The Rosetta Stone of Glioblastoma Pathogenesis and Therapy Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002015. [PMID: 33240762 PMCID: PMC7675056 DOI: 10.1002/advs.202002015] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/23/2020] [Indexed: 05/06/2023]
Abstract
Despite decades of research, glioblastoma (GBM) remains invariably fatal among all forms of cancers. The high level of inter- and intratumoral heterogeneity along with its biological location, the brain, are major barriers against effective treatment. Molecular and single cell analysis identifies different molecular subtypes with varying prognosis, while multiple subtypes can reside in the same tumor. Cellular plasticity among different subtypes in response to therapies or during recurrence adds another hurdle in the treatment of GBM. This phenotypic shift is induced and sustained by activation of several pathways within the tumor itself, or microenvironmental factors. In this review, the dynamic nature of cellular shifts in GBM and how the tumor (immune) microenvironment shapes this process leading to therapeutic resistance, while highlighting emerging tools and approaches to study this dynamic double-edged sword are discussed.
Collapse
Affiliation(s)
- Zulfikar Azam
- Experimental Therapeutics and Molecular Imaging UnitDepartment of NeurologyNeuro‐Oncology DivisionMassachusetts General Hospital and Harvard Medical SchoolBostonMA02129USA
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Shing‐Shun Tony TO
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Bakhos A. Tannous
- Experimental Therapeutics and Molecular Imaging UnitDepartment of NeurologyNeuro‐Oncology DivisionMassachusetts General Hospital and Harvard Medical SchoolBostonMA02129USA
| |
Collapse
|
28
|
VEGFC negatively regulates the growth and aggressiveness of medulloblastoma cells. Commun Biol 2020; 3:579. [PMID: 33067561 PMCID: PMC7568583 DOI: 10.1038/s42003-020-01306-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/17/2020] [Indexed: 02/08/2023] Open
Abstract
Medulloblastoma (MB), the most common brain pediatric tumor, is a pathology composed of four molecular subgroups. Despite a multimodal treatment, 30% of the patients eventually relapse, with the fatal appearance of metastases within 5 years. The major actors of metastatic dissemination are the lymphatic vessel growth factor, VEGFC, and its receptors/co-receptors. Here, we show that VEGFC is inversely correlated to cell aggressiveness. Indeed, VEGFC decreases MB cell proliferation and migration, and their ability to form pseudo-vessel in vitro. Irradiation resistant-cells, which present high levels of VEGFC, lose the ability to migrate and to form vessel-like structures. Thus, irradiation reduces MB cell aggressiveness via a VEGFC-dependent process. Cells intrinsically or ectopically overexpressing VEGFC and irradiation-resistant cells form smaller experimental tumors in nude mice. Opposite to the common dogma, our results give strong arguments in favor of VEGFC as a negative regulator of MB growth. Manon Penco-Campillo, Yannick Comoglio et al. show that VEGFC decreases the proliferation and migration of medulloblastoma cells, as well as their ability to form pseudo vessels. Cells expressing high levels of VEGFC also form smaller tumors when subcutaneously injected into the flank of nude mice, thus highlighting a negative regulatory role for VEGFC on tumor growth.
Collapse
|
29
|
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
|
30
|
Zhang P, Xia Q, Liu L, Li S, Dong L. Current Opinion on Molecular Characterization for GBM Classification in Guiding Clinical Diagnosis, Prognosis, and Therapy. Front Mol Biosci 2020; 7:562798. [PMID: 33102518 PMCID: PMC7506064 DOI: 10.3389/fmolb.2020.562798] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is highly invasive and the deadliest brain tumor in adults. It is characterized by inter-tumor and intra-tumor heterogeneity, short patient survival, and lack of effective treatment. Prognosis and therapy selection is driven by molecular data from gene transcription, genetic alterations and DNA methylation. The four GBM molecular subtypes are proneural, neural, classical, and mesenchymal. More effective personalized therapy heavily depends on higher resolution molecular subtype signatures, combined with gene therapy, immunotherapy and organoid technology. In this review, we summarize the principal GBM molecular classifications that guide diagnosis, prognosis, and therapeutic recommendations.
Collapse
Affiliation(s)
- Pei Zhang
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Qin Xia
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Lei Dong
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
31
|
Fusi F, Trezza A, Sgaragli G, Spiga O, Saponara S, Bova S. Ritanserin blocks Ca V1.2 channels in rat artery smooth muscles: electrophysiological, functional, and computational studies. Acta Pharmacol Sin 2020; 41:1158-1166. [PMID: 32132658 PMCID: PMC7608335 DOI: 10.1038/s41401-020-0370-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/19/2020] [Indexed: 12/14/2022] Open
Abstract
CaV1.2 channel blockers or 5-HT2 receptor antagonists constitute effective therapy for Raynaud’s syndrome. A functional link between the inhibition of 5-HT2 receptors and CaV1.2 channel blockade in arterial smooth muscles has been hypothesized. Therefore, the effects of ritanserin, a nonselective 5-HT2 receptor antagonist, on vascular CaV1.2 channels were investigated through electrophysiological, functional, and computational studies. Ritanserin blocked CaV1.2 channel currents (ICa1.2) in a concentration-dependent manner (Kr = 3.61 µM); ICa1.2 inhibition was antagonized by Bay K 8644 and partially reverted upon washout. Conversely, the ritanserin analog ketanserin (100 µM) inhibited ICa1.2 by ~50%. Ritanserin concentration-dependently shifted the voltage dependence of the steady-state inactivation curve to more negative potentials (Ki = 1.58 µM) without affecting the slope of inactivation and the activation curve, and decreased ICa1.2 progressively during repetitive (1 Hz) step depolarizations (use-dependent block). The addition of ritanserin caused the contraction of single myocytes not yet dialyzed with the conventional method. Furthermore, in depolarized rings, ritanserin, and to a lesser extent, ketanserin, caused a concentration-dependent relaxation, which was antagonized by Bay K 8644. Ritanserin and ketanserin were docked at a region of the CaV1.2 α1C subunit nearby that of Bay K 8644; however, only ritanserin and Bay K 8644 formed a hydrogen bond with key residue Tyr-1489. In conclusion, ritanserin caused in vitro vasodilation, accomplished through the blockade of CaV1.2 channels, which was achieved preferentially in the inactivated and/or resting state of the channel. This novel activity encourages the development of ritanserin derivatives for their potential use in the treatment of Raynaud’s syndrome.
Collapse
|
32
|
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
|
33
|
Yi L, Guo G, Li J, Fan X, Li T, Tong L, Liu P, Wang X, Yuan F, Yu S, Huang Q, Yang X. IKBKE, a prognostic factor preferentially expressed in mesenchymal glioblastoma, modulates tumoral immunosuppression through the STAT3/PD‐L1 pathway. Clin Transl Med 2020. [PMCID: PMC7418810 DOI: 10.1002/ctm2.130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Li Yi
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
- Department of Oncology‐Pathology, Karolinska InstitutetKarolinska University Hospital Solna Stockholm Sweden
| | - Gaochao Guo
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
- Department of Neurosurgery, Henan Provincial People's HospitalPeople's Hospital of Zhengzhou University Zhengzhou Henan China
| | - Jiabo Li
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Xiaoguang Fan
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Tao Li
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Luqing Tong
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
- Department of NeurosurgeryJohns Hopkins University School of Medicine Baltimore MD USA
| | - Peidong Liu
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
- Department of NeurosurgeryJohns Hopkins University School of Medicine Baltimore MD USA
| | - Xuya Wang
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Feng Yuan
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Shengping Yu
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Qiang Huang
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| | - Xuejun Yang
- Department of NeurosurgeryTianjin Medical University General Hospital Tianjin China
- Laboratory of Neuro‐OncologyTianjin Neurological Institute Tianjin China
| |
Collapse
|
34
|
Liu CS, Schmezer P, Popanda O. Diacylglycerol Kinase Alpha in Radiation-Induced Fibrosis: Potential as a Predictive Marker or Therapeutic Target. Front Oncol 2020; 10:737. [PMID: 32477950 PMCID: PMC7235333 DOI: 10.3389/fonc.2020.00737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is an efficient tool in cancer treatment, but it brings along the risk of side effects such as fibrosis in the irradiated healthy tissue thus limiting tumor control and impairing quality of life of cancer survivors. Knowledge on radiation-related fibrosis risk and therapeutic options is still limited and requires further research. Recent studies demonstrated that epigenetic regulation of diacylglycerol kinase alpha (DGKA) is associated with radiation-induced fibrosis. However, the specific mechanisms are still unknown. In this review, we scrutinized the role of DGKA in the radiation response and in further cellular functions to show the potential of DGKA as a predictive marker or a novel target in fibrosis treatment. DGKA was reported to participate in immune response, lipid signaling, exosome production, and migration as well as cell proliferation, all processes which are suggested to be critical steps in fibrogenesis. Most of these functions are based on the conversion of diacylglycerol (DAG) to phosphatidic acid (PA) at plasma membranes, but DGKA might have also other, yet not well-known functions in the nucleus. Current evidence summarized here underlines that DGKA activation may play a central role in fibrosis formation post-irradiation and shows a potential of direct DGKA inhibitors or epigenetic modulators to attenuate pro-fibrotic reactions, thus providing novel therapeutic choices.
Collapse
Affiliation(s)
- Chun-Shan Liu
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schmezer
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Odilia Popanda
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
35
|
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
|
36
|
Lee HH, Lin CH, Lin HY, Kuei CH, Zheng JQ, Wang YH, Lu LS, Lee FP, Hu CJ, Wu D, Lin YF. Histone 2A Family Member J Drives Mesenchymal Transition and Temozolomide Resistance in Glioblastoma Multiforme. Cancers (Basel) 2019; 12:cancers12010098. [PMID: 31906036 PMCID: PMC7016639 DOI: 10.3390/cancers12010098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor and has a poor prognosis and is poorly sensitive to radiotherapy or temozolomide (TMZ) chemotherapy. Therefore, identifying new biomarkers to predict therapeutic responses of GBM is urgently needed. By using The Cancer Genome Atlas (TCGA) database, we found that the upregulation of histone 2A family member J (H2AFJ), but not other H2AFs, is extensively detected in the therapeutic-insensitive mesenchymal, IDH wildtype, MGMT unmethylated, or non-G-CIMP GBM and is associated with poor TMZ responsiveness independent of radiation. Similar views were also found in GBM cell lines. Whereas H2AFJ knockdown diminished TMZ resistance, H2AFJ overexpression promoted TMZ resistance in a panel of GBM cell lines. Gene set enrichment analysis (GSEA) revealed that H2AFJ upregulation accompanied by the activation of TNF-α/NF-κB and IL-6/STAT3-related pathways is highly predicted. Luciferase-based promoter activity assay further validated that the activities of NF-κB and STAT3 are causally affected by H2AFJ expression in GBM cells. Moreover, we found that therapeutic targeting HADC3 by tacedinaline or NF-κB by ML029 is likely able to overcome the TMZ resistance in GBM cells with H2AFJ upregulation. Significantly, the GBM cohorts harboring a high-level H2AFJ transcript combined with high-level expression of TNF-α/NF-κB geneset, IL-6/STAT3 geneset or HADC3 were associated with a shorter time to tumor repopulation after initial treatment with TMZ. These findings not only provide H2AFJ as a biomarker to predict TMZ therapeutic effectiveness but also suggest a new strategy to combat TMZ-insensitive GBM by targeting the interaction network constructed by TNF-α/NF-κB, IL-6/STAT3, HDAC3, and H2AFJ.
Collapse
Affiliation(s)
- Hsun-Hua Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Dizziness and Balance Disorder Center, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Che-Hsuan Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Otolaryngology, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Hui-Yu Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Breast Center, Department of General Surgery, Cardinal Tien Hospital, Xindian District, New Taipei City 231, Taiwan
| | - Chia-Hao Kuei
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Urology, Division of Surgery, Cardinal Tien Hospital, Xindian District, New Taipei City 231, Taiwan
| | - Jing-Quan Zheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Department of Critical Care Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Yuan-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Department of Medical Research, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Long-Sheng Lu
- Department of Radiation Oncology, TMU Hospital, Taipei Medical University, Taipei 11031, Taiwan;
| | - Fei-Peng Lee
- Department of Otolaryngology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chaur-Jong Hu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Dean Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Dizziness and Balance Disorder Center, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 23561, Taiwan
- Sleep Center, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Correspondence: (D.W.); (Y.-F.L.); Tel.: +886-2-22490088 (ext. 8112) (D.W.); +886-2-2736-1661 (ext. 3106) (Y.-F.L.); Fax: +886-2-22490088 (D.W.); +886-2-2739-0500 (Y.-F.L.)
| | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-H.L.); (H.-Y.L.); (C.-H.K.); (J.-Q.Z.); (Y.-H.W.); (C.-J.H.)
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Correspondence: (D.W.); (Y.-F.L.); Tel.: +886-2-22490088 (ext. 8112) (D.W.); +886-2-2736-1661 (ext. 3106) (Y.-F.L.); Fax: +886-2-22490088 (D.W.); +886-2-2739-0500 (Y.-F.L.)
| |
Collapse
|
37
|
Epithelial to mesenchymal plasticity and differential response to therapies in pancreatic ductal adenocarcinoma. Proc Natl Acad Sci U S A 2019; 116:26835-26845. [PMID: 31843922 PMCID: PMC6936349 DOI: 10.1073/pnas.1914915116] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PDAC epithelial (E) and quasi-mesenchymal (QM) subtypes can be modulated by different therapies demonstrating the plasticity of PDAC cells that contributes to the heterogeneity of PDAC tumors and their intrinsic resistance to a broad spectrum of therapies. Understanding and monitoring the fluidity of PDAC E and QM states are critical to the development of improved clinical trial design to target these different subpopulations. Transcriptional profiling has defined pancreatic ductal adenocarcinoma (PDAC) into distinct subtypes with the majority being classical epithelial (E) or quasi-mesenchymal (QM). Despite clear differences in clinical behavior, growing evidence indicates these subtypes exist on a continuum with features of both subtypes present and suggestive of interconverting cell states. Here, we investigated the impact of different therapies being evaluated in PDAC on the phenotypic spectrum of the E/QM state. We demonstrate using RNA-sequencing and RNA-in situ hybridization (RNA-ISH) that FOLFIRINOX combination chemotherapy induces a common shift of both E and QM PDAC toward a more QM state in cell lines and patient tumors. In contrast, Vitamin D, another drug under clinical investigation in PDAC, induces distinct transcriptional responses in each PDAC subtype, with augmentation of the baseline E and QM state. Importantly, this translates to functional changes that increase metastatic propensity in QM PDAC, but decrease dissemination in E PDAC in vivo models. These data exemplify the importance of both the initial E/QM subtype and the plasticity of E/QM states in PDAC in influencing response to therapy, which highlights their relevance in guiding clinical trials.
Collapse
|
38
|
DGKα in Neutrophil Biology and Its Implications for Respiratory Diseases. Int J Mol Sci 2019; 20:ijms20225673. [PMID: 31766109 PMCID: PMC6887790 DOI: 10.3390/ijms20225673] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022] Open
Abstract
Diacylglycerol kinases (DGKs) play a key role in phosphoinositide signaling by removing diacylglycerol and generating phosphatidic acid. Besides the well-documented role of DGKα and DGKζ as negative regulators of lymphocyte responses, a robust body of literature points to those enzymes, and specifically DGKα, as crucial regulators of leukocyte function. Upon neutrophil stimulation, DGKα activation is necessary for migration and a productive response. The role of DGKα in neutrophils is evidenced by its aberrant behavior in juvenile periodontitis patients, which express an inactive DGKα transcript. Together with in vitro experiments, this suggests that DGKs may represent potential therapeutic targets for disorders where inflammation, and neutrophils in particular, plays a major role. In this paper we focus on obstructive respiratory diseases, including asthma and chronic obstructive pulmonary disease (COPD), but also rare genetic diseases such as alpha-1-antitrypsin deficiency. Indeed, the biological role of DGKα is understudied outside the T lymphocyte field. The recent wave of research aiming to develop novel and specific inhibitors as well as KO mice will allow a better understanding of DGK's role in neutrophilic inflammation. Better knowledge and pharmacologic tools may also allow DGK to move from the laboratory bench to clinical trials.
Collapse
|
39
|
Liu Q, Guan Y, Li Z, Wang Y, Liu Y, Cui R, Wang Y. miR-504 suppresses mesenchymal phenotype of glioblastoma by directly targeting the FZD7-mediated Wnt-β-catenin pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:358. [PMID: 31419987 PMCID: PMC6697940 DOI: 10.1186/s13046-019-1370-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background MicroRNAs (miRNAs) play crucial roles in tumor initiation and development. Previously, we indicated that miR-504 is downregulated and suppresses tumor proliferation in glioblastoma (GBM). However, the regulation and relevant mechanism of miR-504 in GBM mesenchymal (ME) transition remain unclear. Methods Transcriptome and clinical data were obtained from The Cancer Genome Atlas (TCGA) database. The potential functions of miR-504 were predicted using gene ontology analysis. GBM cell migration and invasion were examined using wound healing and Transwell assays. Epithelial–mesenchymal transition (EMT) progression in GBM cell lines was detected with immunofluorescence and western blotting. The stemness activity of glioma stem-like cells (GSCs) was assessed by sphere formation assay and tumor xenograft model. miR-504 binding to the FZD7 (frizzled class receptor 7) 3′ untranslated region (3′UTR) was validated using dual luciferase reporter assay. TOP/FOP Flash assays were conducted to determine the effects of miR-504 on Wnt/β-catenin signaling. Results Analysis of TCGA transcriptomic data showed that low miR-504 expression correlated with ME subtype transition and poor survival in patients with GBM. Functional experiments showed that miR-504 overexpression suppressed malignant behaviors of GBM cells, such as migration, invasion, EMT, and stemness activity. Furthermore, miR-504 was a negative regulator of the Wnt–β-catenin pathway by directly repressing FZD7 expression, and FZD7 overexpression reversed the EMT inhibition caused by miR-504. Moreover, the low miR-504/FZD7 expression ratio was a ME subtype marker and could serve as a significant prognostic indicator and predict the clinical outcome of chemotherapy and radiotherapy for patients with GBM in TCGA dataset. Conclusions Our results suggest that miR-504 suppresses the aggressive biological processes associated with the ME phenotype of GBM and could be a potential candidate for therapeutic applications in these malignant brain tumors. Electronic supplementary material The online version of this article (10.1186/s13046-019-1370-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Yanlei Guan
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Zhenhang Li
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Yao Wang
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Yu Liu
- Department of Cardiac Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Run Cui
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Yunjie Wang
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
40
|
Wang ZL, Wang Z, Li GZ, Wang QW, Bao ZS, Zhang CB, Jiang T. Immune Cytolytic Activity Is Associated With Genetic and Clinical Properties of Glioma. Front Immunol 2019; 10:1756. [PMID: 31428092 PMCID: PMC6688525 DOI: 10.3389/fimmu.2019.01756] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 07/11/2019] [Indexed: 12/30/2022] Open
Abstract
Background: Immunotherapy provided unprecedented advances in the treatment of several previously untreated cancers. However, these immunomodulatory maneuvers showed limited response to patients with glioma in clinical trials. Our aim was to depict the immune characteristics of glioma with immune cytolytic activity at genetic and transcriptome levels. Methods: In total, 325 gliomas from CGGA dataset as training cohort and 699 gliomas from TCGA dataset as validation cohort were enrolled in our analysis. We calculated the immune cytolytic activity for 1,000 of gliomas. The characteristics of immune cytolytic activity in gliomas were interpreted by the corresponding clinical, molecular genetics and radiological information. Results: We found that immune cytolytic activity was highly associated with molecular, clinical, and edema extent. High cytolytic activity gliomas were more likely to be diagnosed as glioblastoma and might be a potential marker of mesenchymal subtype. Moreover, those gliomas exhibited significantly increased copy number alterations including recurrent focal amplifications of PDGFA and EGFR, as well as recurrent deletions of CDKN2A/B. Subsequent biological function analysis revealed that the immune response and immune checkpoints expression were significantly correlated with the cytolytic activity of gliomas. Immune cytolytic activity was significantly positively associated with the extent of peri-tumor edema and was independently correlated with reduced survival time. Conclusion: Our results highlighted the immunoregulatory mechanism heterogeneity of gliomas. Cytolytic activity, indirectly reflected by the extent of peri-tumor edema, may provide a potential index to evaluate the status of immune microenvironment and immune checkpoints in glioma, which should be fully valued for precision classification and immunotherapy.
Collapse
Affiliation(s)
- Zhi-Liang Wang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guan-Zhang Li
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Qiang-Wei Wang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhao-Shi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuan-Bao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
41
|
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
|
42
|
Audia A, Bhat KP. Ritanserin, a novel agent targeting the mesenchymal subtype of glioblastomas. Neuro Oncol 2018; 20:151-152. [PMID: 29365204 DOI: 10.1093/neuonc/nox240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
| | - Krishna P Bhat
- University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
43
|
The proneural gene ASCL1 governs the transcriptional subgroup affiliation in glioblastoma stem cells by directly repressing the mesenchymal gene NDRG1. Cell Death Differ 2018; 26:1813-1831. [PMID: 30538287 PMCID: PMC6748080 DOI: 10.1038/s41418-018-0248-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 11/03/2018] [Accepted: 11/21/2018] [Indexed: 01/09/2023] Open
Abstract
Achaete-scute homolog 1 gene (ASCL1) is a gene classifier for the proneural (PN) transcriptional subgroup of glioblastoma (GBM) that has a relevant role in the neuronal-like differentiation of GBM cancer stem cells (CSCs) through the activation of a PN gene signature. Besides prototypical ASCL1 PN target genes, the molecular effectors mediating ASCL1 function in regulating GBM differentiation and, most relevantly, subgroup specification are currently unknown. Here we report that ASCL1 not only promotes the acquisition of a PN phenotype in CSCs by inducing a glial-to-neuronal lineage switch but also concomitantly represses mesenchymal (MES) features by directly downregulating the expression of N-Myc downstream-regulated gene 1 (NDRG1), which we propose as a novel gene classifier of MES GBMs. Increasing the expression of ASCL1 in PN CSCs results in suppression of self-renewal, promotion of differentiation and, most significantly, decrease in tumorigenesis, which is also reproduced by NDRG1 silencing. Conversely, both abrogation of ASCL1 expression in PN CSCs and enforcement of NDRG1 expression in either PN or MES CSCs induce proneural-to-mesenchymal transition (PMT) and enhanced mesenchymal features. Surprisingly, ASCL1 overexpression in MES CSCs increases malignant features and gives rise to a neuroendocrine-like secretory phenotype. Altogether, our results propose that the fine interplay between ASCL1 and its target NDRG1 might serve as potential subgroup-specific targetable vulnerability in GBM; enhancing ASCL1 expression in PN GBMs might reduce tumorigenesis, whereas repressing NDRG1 expression might be actionable to hamper the malignancy of GBM belonging to the MES subgroup.
Collapse
|
44
|
Campbell ST, Franks CE, Borne AL, Shin M, Zhang L, Hsu KL. Chemoproteomic Discovery of a Ritanserin-Targeted Kinase Network Mediating Apoptotic Cell Death of Lung Tumor Cells. Mol Pharmacol 2018; 94:1246-1255. [PMID: 30158316 PMCID: PMC6160665 DOI: 10.1124/mol.118.113001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
Ritanserin was tested in the clinic as a serotonin receptor inverse agonist but recently emerged as a novel kinase inhibitor with potential applications in cancer. Here, we discovered that ritanserin induced apoptotic cell death of non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) cells via a serotonin-independent mechanism. We used quantitative chemical proteomics to reveal a ritanserin-dependent kinase network that includes key mediators of lipid [diacylglycerol kinase α, phosphatidylinositol 4-kinase β] and protein [feline encephalitis virus-related kinase, rapidly accelerated fibrosarcoma (RAF)] signaling, metabolism [eukaryotic elongation factor 2 kinase, eukaryotic translation initiation factor 2-α kinase 4], and DNA damage response [tousled-like kinase 2] to broadly kill lung tumor cell types. Whereas ritanserin exhibited polypharmacology in NSCLC proteomes, this compound showed unexpected specificity for c-RAF in the SCLC subtype, with negligible activity against other kinases mediating mitogen-activated protein kinase signaling. Here we show that ritanserin blocks c-RAF but not B-RAF activation of established oncogenic signaling pathways in live cells, providing evidence in support of c-RAF as a key target mediating its anticancer activity. Given the role of c-RAF activation in RAS-mutated cancers resistant to clinical B-RAF inhibitors, our findings may have implications in overcoming resistance mechanisms associated with c-RAF biology. The unique target landscape combined with acceptable safety profiles in humans provides new opportunities for repositioning ritanserin in cancer.
Collapse
Affiliation(s)
- Sean T Campbell
- Departments of Chemistry (S.T.C., C.E.F., M.S., L.Z., K.-L.H.), Pathology (S.T.C.), and Pharmacology (A.L.B., K.-L.H.), University of Virginia Cancer Center (K.-L.H.), University of Virginia, Charlottesville, Virginia
| | - Caroline E Franks
- Departments of Chemistry (S.T.C., C.E.F., M.S., L.Z., K.-L.H.), Pathology (S.T.C.), and Pharmacology (A.L.B., K.-L.H.), University of Virginia Cancer Center (K.-L.H.), University of Virginia, Charlottesville, Virginia
| | - Adam L Borne
- Departments of Chemistry (S.T.C., C.E.F., M.S., L.Z., K.-L.H.), Pathology (S.T.C.), and Pharmacology (A.L.B., K.-L.H.), University of Virginia Cancer Center (K.-L.H.), University of Virginia, Charlottesville, Virginia
| | - Myungsun Shin
- Departments of Chemistry (S.T.C., C.E.F., M.S., L.Z., K.-L.H.), Pathology (S.T.C.), and Pharmacology (A.L.B., K.-L.H.), University of Virginia Cancer Center (K.-L.H.), University of Virginia, Charlottesville, Virginia
| | - Liuzhi Zhang
- Departments of Chemistry (S.T.C., C.E.F., M.S., L.Z., K.-L.H.), Pathology (S.T.C.), and Pharmacology (A.L.B., K.-L.H.), University of Virginia Cancer Center (K.-L.H.), University of Virginia, Charlottesville, Virginia
| | - Ku-Lung Hsu
- Departments of Chemistry (S.T.C., C.E.F., M.S., L.Z., K.-L.H.), Pathology (S.T.C.), and Pharmacology (A.L.B., K.-L.H.), University of Virginia Cancer Center (K.-L.H.), University of Virginia, Charlottesville, Virginia
| |
Collapse
|
45
|
Ratti S, Follo MY, Ramazzotti G, Faenza I, Fiume R, Suh PG, McCubrey JA, Manzoli L, Cocco L. Nuclear phospholipase C isoenzyme imbalance leads to pathologies in brain, hematologic, neuromuscular, and fertility disorders. J Lipid Res 2018; 60:312-317. [PMID: 30287524 DOI: 10.1194/jlr.r089763] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/30/2018] [Indexed: 12/31/2022] Open
Abstract
Phosphoinositide-specific phospholipases C (PI-PLCs) are involved in signaling pathways related to critical cellular functions, such as cell cycle regulation, cell differentiation, and gene expression. Nuclear PI-PLCs have been studied as key enzymes, molecular targets, and clinical prognostic/diagnostic factors in many physiopathologic processes. Here, we summarize the main studies about nuclear PI-PLCs, specifically, the imbalance of isozymes such as PI-PLCβ1 and PI-PLCζ, in cerebral, hematologic, neuromuscular, and fertility disorders. PI-PLCβ1 and PI-PLCɣ1 affect epilepsy, depression, and bipolar disorder. In the brain, PI-PLCβ1 is involved in endocannabinoid neuronal excitability and is a potentially novel signature gene for subtypes of high-grade glioma. An altered quality or quantity of PI-PLCζ contributes to sperm defects that result in infertility, and PI-PLCβ1 aberrant inositide signaling contributes to both hematologic and degenerative muscle diseases. Understanding the mechanisms behind PI-PLC involvement in human pathologies may help identify new strategies for personalized therapies of these conditions.
Collapse
Affiliation(s)
- Stefano Ratti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Matilde Y Follo
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Irene Faenza
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Roberta Fiume
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Pann-Ghill Suh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 689-798, Korea
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Lucio Cocco
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
46
|
Olmez I, Zhang Y, Manigat L, Benamar M, Brenneman B, Nakano I, Godlewski J, Bronisz A, Lee J, Abbas T, Abounader R, Purow B. Combined c-Met/Trk Inhibition Overcomes Resistance to CDK4/6 Inhibitors in Glioblastoma. Cancer Res 2018; 78:4360-4369. [PMID: 29844123 DOI: 10.1158/0008-5472.can-17-3124] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/17/2018] [Accepted: 05/22/2018] [Indexed: 01/02/2023]
Abstract
Glioblastoma (GBM) is the most common primary brain malignancy and carries an extremely poor prognosis. Recent molecular studies revealed the CDK4/6-Rb-E2F axis and receptor tyrosine kinase (RTK) signaling to be deregulated in most GBM, creating an opportunity to develop more effective therapies by targeting both pathways. Using a phospho-RTK protein array, we found that both c-Met and TrkA-B pathways were significantly activated upon CDK4/6 inhibition in GBM cells. We therefore investigated the efficacy of combined CDK4/6 and c-Met/TrkA-B inhibition against GBM. We show that both c-Met and TrkA-B pathways transactivate each other, and targeting both pathways simultaneously results in more efficient pathway suppression. Mechanistically, inhibition of CDK4/6 drove NF-κB-mediated upregulation of hepatocyte growth factor, brain-derived neurotrophic factor, and nerve growth factor that in turn activated both c-Met and TrkA-B pathways. Combining the CDK4/6 inhibitor abemaciclib with the c-Met/Trk inhibitor altiratinib or the corresponding siRNAs induced apoptosis, leading to significant synergy against GBM. Collectively, these findings demonstrate that the activation of c-Met/TrkA-B pathways is a novel mechanism involved in therapeutic resistance of GBM to CDK4/6 inhibition and that dual inhibition of c-Met/Trk with CDK4/6 should be considered in future clinical trials.Significance: CDK4/6 inhibition in glioblastoma activates the c-Met and TrkA-B pathways mediated by NF-κB and can be reversed by a dual c-Met/Trk inhibitor. Cancer Res; 78(15); 4360-9. ©2018 AACR.
Collapse
Affiliation(s)
- Inan Olmez
- Department of Neurology, University of Virginia, Charlottesville, Virginia.
| | - Ying Zhang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Laryssa Manigat
- Department of Neurology, University of Virginia, Charlottesville, Virginia
| | - Mouadh Benamar
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia.,Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - Breanna Brenneman
- Department of Neurology, University of Virginia, Charlottesville, Virginia
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama, Birmingham, Alabama
| | - Jakub Godlewski
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Agnieszka Bronisz
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Tarek Abbas
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia.,Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - Roger Abounader
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Benjamin Purow
- Department of Neurology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|