1
|
Zhou YQ, Cheng XX, He S, Liu SQ, Li YQ, Wei PP, Luo CL, Bei JX. A positive feedback loop between PLD1 and NF-κB signaling promotes tumorigenesis of nasopharyngeal carcinoma. J Genet Genomics 2024; 51:997-1006. [PMID: 38885836 DOI: 10.1016/j.jgg.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Phospholipase D (PLD) lipid-signaling enzyme superfamily has been widely implicated in various human malignancies, but its role and underlying mechanism remain unclear in nasopharyngeal carcinoma (NPC). Here, we analyze the expressions of 6 PLD family members between 87 NPC and 10 control samples through transcriptome analysis. Our findings reveal a notable upregulation of PLD1 in both NPC tumors and cell lines, correlating with worse disease-free and overall survival in NPC patients. Functional assays further elucidate the oncogenic role of PLD1, demonstrating its pivotal promotion of critical tumorigenic processes such as cell proliferation and migration in vitro, as well as tumor growth in vivo. Notably, our study uncovers a positive feedback loop between PLD1 and the NF-κB signaling pathway to render NPC progression. Specifically, PLD1 enhances NF-κB activity by facilitating the phosphorylation and nuclear translocation of RELA, which in turn binds to the promoter of PLD1, augmenting its expression. Moreover, RELA overexpression markedly rescues the inhibitory effects in PLD1-depleted NPC cells. Importantly, the application of the PLD1 inhibitor, VU0155069, substantially inhibits NPC tumorigenesis in a patient-derived xenograft model. Together, our findings identify PLD1/NF-κB signaling as a positive feedback loop with promising therapeutic and prognostic potential in NPC.
Collapse
Affiliation(s)
- Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China; Department of Medical Oncology, National Cancer Centre of Singapore, Singapore.
| |
Collapse
|
2
|
Li MY, Chen P, Pan MX, Hu HL, Jiang YJ. Palladium-catalyzed amidation of carbazole derivatives via hydroamination of isocyanates. Org Biomol Chem 2024. [PMID: 39005158 DOI: 10.1039/d4ob00771a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The first amidation of carbazoles at the N9 position via palladium-catalyzed hydroamination of isocyanates is demonstrated. This simple, general and efficient method could deliver a wide range of carbazole-N-carboxamides in up to 99% yield. The salient features of this transformation include simple conditions with no need for a strong base, high chemo- and regio-selectivities and good functional group tolerance. In particular, this work-up-free and chromatography-free protocol is time-saving, cost-effective and user-friendly.
Collapse
Affiliation(s)
- Meng-Yuan Li
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Peng Chen
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Ming-Xia Pan
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Hao-Lan Hu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Yi-Jun Jiang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
3
|
Lim SH, Lee H, Lee HJ, Kim K, Choi J, Han JM, Min DS. PLD1 is a key player in cancer stemness and chemoresistance: Therapeutic targeting of cross-talk between the PI3K/Akt and Wnt/β-catenin pathways. Exp Mol Med 2024; 56:1479-1487. [PMID: 38945955 PMCID: PMC11297275 DOI: 10.1038/s12276-024-01260-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 07/02/2024] Open
Abstract
The development of chemoresistance is a major challenge in the treatment of several types of cancers in clinical settings. Stemness and chemoresistance are the chief causes of poor clinical outcomes. In this context, we hypothesized that understanding the signaling pathways responsible for chemoresistance in cancers is crucial for the development of novel targeted therapies to overcome drug resistance. Among the aberrantly activated pathways, the PI3K-Akt/Wnt/β-catenin signaling pathway is clinically implicated in malignancies such as colorectal cancer (CRC) and glioblastoma multiforme (GBM). Aberrant dysregulation of phospholipase D (PLD) has been implicated in several malignancies, and oncogenic activation of this pathway facilitates tumor proliferation, stemness, and chemoresistance. Crosstalk involving the PLD and Wnt/β-catenin pathways promotes the progression of CRC and GBM and reduces the sensitivity of cancer cells to standard therapies. Notably, both pathways are tightly regulated and connected at multiple levels by upstream and downstream effectors. Thus, gaining deeper insights into the interactions between these pathways would help researchers discover unique therapeutic targets for the management of drug-resistant cancers. Here, we review the molecular mechanisms by which PLD signaling stimulates stemness and chemoresistance in CRC and GBM. Thus, the current review aims to address the importance of PLD as a central player coordinating cross-talk between the PI3K/Akt and Wnt/β-catenin pathways and proposes the possibility of targeting these pathways to improve cancer therapy and overcome drug resistance.
Collapse
Affiliation(s)
- Seong Hun Lim
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Hyesung Lee
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Hyun Ji Lee
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Kuglae Kim
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Junjeong Choi
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Jung Min Han
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- POSTECH Biotech Center, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Do Sik Min
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
| |
Collapse
|
4
|
Han K, He Z, Liu Y, Chen H. Identification of EDNRA as the Key Biomarker for Hypercholesterolemia and Colorectal Cancer. TOHOKU J EXP MED 2024; 262:181-189. [PMID: 38123303 DOI: 10.1620/tjem.2023.j101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Some studies have investigated the role of cholesterol in the progression of colorectal cancer (CRC). However, the underlying mechanism of action is not clear. In this study, we used bioinformatics tools to elucidate the molecular mechanisms involved. We initially obtained CRC datasets from the Gene Expression Omnibus (GEO) database and hypercholesterolemia data from GeneCards and DisGeNE. Common differentially expressed genes (DEGs) were determined by using Venn diagram web tools. Next, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses using the Database for Annotation, Visualization, and Integrated Discovery (DAVID). The hub gene was identified through common expression pattern analysis and survival analysis. Finally, we conducted an immune regulatory point analysis and predicted target drugs based on the hub gene. The results of our analysis revealed 13 common DEGs, with endothelin receptor type A (EDNRA) identified as the hub gene linking hypercholesterolemia and CRC. The results of the GO analysis showed that the common DEGs were primarily associated with the G-protein coupled receptor signaling pathway, extracellular space, and receptor binding. The results of the KEGG pathway enrichment analysis indicated enrichment in pathways related to cancer and the phospholipase D signaling pathway. Additionally, we identified potential target drugs, including Podocarpus montanus, Diospyros kaki, Herba Salviae japoniae, sitaxentan, and ambrisentan. We found that EDNRA might be an underlying biomarker for both hypercholesterolemia and CRC. The predicted target drugs provide new strategies for treating CRC.
Collapse
Affiliation(s)
- Kedong Han
- Department of Cardiology, Maoming People's Hospital
| | - Zhijiang He
- Department of Oncology, Maoming People's Hospital
| | - Yunjun Liu
- Department of Oncology, Maoming People's Hospital
| | - Hua Chen
- Department of Oncology, Maoming People's Hospital
| |
Collapse
|
5
|
Zhao H, Brånalt J, Perry M, Tyrchan C. The Role of Allylic Strain for Conformational Control in Medicinal Chemistry. J Med Chem 2023. [PMID: 37285219 DOI: 10.1021/acs.jmedchem.3c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
It is axiomatic in medicinal chemistry that optimization of the potency of a small molecule at a macromolecular target requires complementarity between the ligand and target. In order to minimize the conformational penalty on binding, both enthalpically and entropically, it is therefore preferred to have the ligand preorganized in the bound conformation. In this Perspective, we highlight the role of allylic strain in controlling conformational preferences. Allylic strain was originally described for carbon-based allylic systems, but the same principles apply to other types of structure with sp2 or pseudo-sp2 arrangements. These systems include benzylic (including heteroaryl methyl) positions, amides, N-aryl groups, aryl ethers, and nucleotides. We have derived torsion profiles from small molecule X-ray structures for these systems. Through multiple examples, we show how these effects have been applied in drug discovery and how they can be used prospectively to influence conformation in the design process.
Collapse
Affiliation(s)
- Hongtao Zhao
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Jonas Brånalt
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Matthew Perry
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Christian Tyrchan
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| |
Collapse
|
6
|
Wolf A, Tanguy E, Wang Q, Gasman S, Vitale N. Phospholipase D and cancer metastasis: A focus on exosomes. Adv Biol Regul 2023; 87:100924. [PMID: 36272918 DOI: 10.1016/j.jbior.2022.100924] [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: 09/19/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 03/01/2023]
Abstract
In mammals, phospholipase D (PLD) enzymes involve 6 isoforms, of which only three have established lipase activity to produce the signaling lipid phosphatidic acid (PA). This phospholipase activity has been postulated to contribute to cancer progression for over three decades now, but the exact mechanisms involved have yet to be uncovered. Indeed, using various models, an altered PLD activity has been proposed altogether to increase cell survival rate, promote angiogenesis, boost rapamycin resistance, and favor metastasis. Although for some part, the molecular pathways by which this increase in PA is pro-oncogenic are partially known, the pleiotropic functions of PA make it quite difficult to distinguish which among these simple signaling pathways is responsible for each of these PLD facets. In this review, we will describe an additional potential contribution of PA generated by PLD1 and PLD2 in the biogenesis, secretion, and uptake of exosomes. Those extracellular vesicles are now viewed as membrane vehicles that carry informative molecules able to modify the fate of receiving cells at distance from the original tumor to favor homing of metastasis. The perspectives for a better understanding of these complex role of PLDs will be discussed.
Collapse
Affiliation(s)
- Alexander Wolf
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Emeline Tanguy
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Qili Wang
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Stéphane Gasman
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Nicolas Vitale
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.
| |
Collapse
|
7
|
Peng T, Xu T, Liu X. Research progress of the engagement of inorganic nanomaterials in cancer immunotherapy. Drug Deliv 2022; 29:1914-1932. [PMID: 35748543 PMCID: PMC9246104 DOI: 10.1080/10717544.2022.2086940] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer has attracted widespread attention from scientists for its high morbidity and mortality, posing great threats to people’s health. Cancer immunotherapy with high specificity, low toxicity as well as triggering systemic anti-tumor response has gradually become common in clinical cancer treatment. However, due to the insufficient immunogenicity of tumor antigens peptides, weak ability to precisely target tumor sites, and the formation of tumor immunosuppressive microenvironment, the efficacy of immunotherapy is often limited. In recent years, the emergence of inorganic nanomaterials makes it possible for overcoming the limitations mentioned above. With self-adjuvant properties, high targeting ability, and good biocompatibility, the inorganic nanomaterials have been integrated with cancer immunotherapy and significantly improved the therapeutic effects.
Collapse
Affiliation(s)
- Tingwei Peng
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Pudong New Area, China
| | - Tianzhao Xu
- Shanghai Qiansu Biological Technology Co., Ltd, Pudong New Area, China.,Department of Clinical Laboratory, Gongli Hospital, School of Medicine, Shanghai University, Shanghai, China
| | - Xinghui Liu
- Department of Clinical Laboratory, Gongli Hospital, School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
8
|
Inhibition of phospholipase D1 induces immunogenic cell death and potentiates cancer immunotherapy in colorectal cancer. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1563-1576. [PMID: 36131027 PMCID: PMC9535023 DOI: 10.1038/s12276-022-00853-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 12/02/2022]
Abstract
Phospholipase D (PLD) is a potential therapeutic target against cancer. However, the contribution of PLD inhibition to the antitumor response remains unknown. We developed a potent and selective PLD1 inhibitor based on computer-aided drug design. The inhibitor enhanced apoptosis in colorectal cancer (CRC) cells but not in normal colonic cells, and in vitro cardiotoxicity was not observed. The inhibitor downregulated the Wnt/β-catenin signaling pathway and reduced the migration, invasion, and self-renewal capacity of CRC cells. In cancer, therapeutic engagement of immunogenic cell death (ICD) leads to more effective responses by eliciting the antitumor immunity of T cells. The CRC cells treated with the inhibitor showed hallmarks of ICD, including downregulation of “do not eat-me” signals (CD24, CD47, programmed cell death ligand 1 [PD-L1]), upregulation of “eat-me” signal (calreticulin), release of high-mobility group Box 1, and ATP. PLD1 inhibition subsequently enhanced the phagocytosis of cancer cells by macrophages through the surface expression of costimulatory molecules; as a result, the cancer cells were more susceptible to cytotoxic T-cell-mediated killing. Moreover, PLD1 inhibition attenuated colitis-associated CRC and orthotopically injected tumors, probably by controlling multiple pathways, including Wnt signaling, phagocytosis checkpoints, and immune signaling. Furthermore, combination therapy with a PLD1 inhibitor and an anti-PD-L1 antibody further enhanced tumor regression via immune activation in the tumor environment. Collectively, in this study, PLD1 was identified as a critical regulator of the tumor microenvironment in colorectal cancer, suggesting the potential of PLD1 inhibitors for cancer immunotherapy based on ICD and immune activation. PLD1 inhibitors may act as promising immune modulators in antitumor treatment via ICD. A novel drug that can inhibit an enzyme involved in colorectal cancer progression shows promise in trials on mouse models. The phospholipase D1 (PLD1) enzyme reinforces a critical signaling pathway that promotes cancer progression and drug resistance. Using computer-aided drug design, South Korean researchers led by Do Sik Min and Gyoonhee Han at Yonsei University in Incheon and Seoul, respectively, have developed a drug that specifically binds to and inhibits PLD1. In trials, the researchers observed downregulation of PLD1’s associated signaling pathway, and reductions in the ability of colorectal cancer cells to migrate, invade and replicate. The drug suppressed the cancer cells’ “don’t-eat-me” signals and upregulated “eat-me” signals, triggering improved responses from the immune system. The drug was even more effective when used in combination with an immunotherapy agent.
Collapse
|
9
|
Perez-Valle A, Ochoa B, Shah KN, Barreda-Gomez G, Astigarraga E, Boyano MD, Asumendi A. Upregulated phospholipase D2 expression and activity is related to the metastatic properties of melanoma. Oncol Lett 2022; 23:140. [PMID: 35340556 PMCID: PMC8931840 DOI: 10.3892/ol.2022.13260] [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: 09/30/2021] [Accepted: 02/01/2022] [Indexed: 11/05/2022] Open
Abstract
The incidence rates of melanoma have increased steadily in recent decades and nearly 25% of the patients diagnosed with early-stage melanoma will eventually develop metastasis, for which there is currently no fully effective treatment. The link between phospholipases and tumors has been studied extensively, particularly in breast and colon cancers. With the aim of finding new biomarkers and therapeutic options for melanoma, the expression of different phospholipases was assessed in 17 distinct cell lines in the present study, demonstrating that phospholipase D2 (PLD2) is upregulated in metastatic melanoma as compared to normal skin melanocytes. These results were corroborated by immunofluorescence and lipase activity assays. Upregulation of PLD2 expression and increased lipase activity were observed in metastatic melanoma relative to normal skin melanocytes. So far, the implication of PLD2 activity in melanoma malignancies has remained elusive. To the best of our knowledge, the present study was the first to demonstrate that the overexpression of PLD2 enhances lipase activity, and its effect to increase the proliferation, migration and invasion capacity of melanoma cells was assessed with XTT and Transwell assays. In addition, silencing of PLD2 in melanoma cells reduced the metastatic potential of these cells. The present study provided evidence that PLD2 is involved in melanoma malignancy and in particular, in its metastatic potential, and established a basis for future studies evaluating PLD2 blockade as a therapeutic strategy to manage this condition.
Collapse
Affiliation(s)
- Arantza Perez-Valle
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940 Bizkaia, Spain
| | - Begoña Ochoa
- Department of Physiology, School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940 Bizkaia, Spain
| | - Krushangi N. Shah
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH 45435, USA
| | | | - Egoitz Astigarraga
- IMG Pharma Biotech S.L., Bizkaia Technological Park, Zamudio, 48160 Bizkaia, Spain
| | - María Dolores Boyano
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940 Bizkaia, Spain
- Biocruces-Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, 48903 Bizkaia, Spain
| | - Aintzane Asumendi
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, 48940 Bizkaia, Spain
- Biocruces-Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, 48903 Bizkaia, Spain
| |
Collapse
|
10
|
May-Dracka TL, Gao F, Hopkins BT, Hronowski X, Chen T, Chodaparambil JV, Metrick CM, Cullivan M, Enyedy I, Kaliszczak M, Kankel MW, Marx I, Michell-Robinson MA, Murugan P, Kumar PR, Rooney M, Schuman E, Sen A, Wang T, Ye T, Peterson EA. Discovery of Phospholipase D Inhibitors with Improved Drug-like Properties and Central Nervous System Penetrance. ACS Med Chem Lett 2022; 13:665-673. [PMID: 35450377 PMCID: PMC9014516 DOI: 10.1021/acsmedchemlett.1c00682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/01/2022] [Indexed: 11/30/2022] Open
Abstract
Phospholipase D (PLD) is a phospholipase enzyme responsible for hydrolyzing phosphatidylcholine into the lipid signaling molecule, phosphatidic acid, and choline. From a therapeutic perspective, PLD has been implicated in human cancer progression as well as a target for neurodegenerative diseases, including Alzheimer's. Moreover, knockdown of PLD rescues the ALS phenotype in multiple Drosophila models of ALS (amyotrophic lateral sclerosis) and displays modest motor benefits in an SOD1 ALS mouse model. To further validate whether inhibiting PLD is beneficial for the treatment of ALS, a brain penetrant small molecule inhibitor with suitable PK properties to test in an ALS animal model is needed. Using a combination of ligand-based drug discovery and structure-based design, a dual PLD1/PLD2 inhibitor was discovered that is single digit nanomolar in the Calu-1 cell assay and has suitable PK properties for in vivo studies. To capture the in vivo measurement of PLD inhibition, a transphosphatidylation pharmacodynamic LC-MS assay was developed, in which a dual PLD1/PLD2 inhibitor was found to reduce PLD activity by 15-20-fold.
Collapse
|
11
|
Batista VF, Pinto DCGA, Silva AMS. Recent in vivo advances of spirocyclic scaffolds for drug discovery. Expert Opin Drug Discov 2022; 17:603-618. [PMID: 35333138 DOI: 10.1080/17460441.2022.2055544] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Spirocyclic scaffolds are an exceptional tool in drug design, allowing fine-tuning of a molecule's conformational and physicochemical properties. As it expands and diversifies, so does the number of therapeutics that contain this core. Several spirocyclic drugs are already marketed, and considerably more have shown promising results. AREAS COVERED This review addresses recent in vivo studies (2017-2021) on applying spirocyclic compounds to treat various diseases, mainly grouped within neurological, infectious, and metabolic diseases and cancer. An emphasis is given on the influence of the spiro-structure on activity and consequent structure-activity study. In vivo results and their significance in the future progression towards clinical trials are also presented. EXPERT OPINION Spirocyclic compounds present an exciting opportunity as an unexplored chemical space in medicinal chemistry. However, their development is hindered by their complexity and synthesis challenges. Furthermore, a clear preference is still seen for readily available spirocyclic compounds involving amine or amide bonds. Nevertheless, these are temporary as high-throughput synthesis, and computational techniques allow fast optimization studies. In our opinion, the field of spirocyclic chemistry will continue to thrive and contribute to drug development, improving activity and selectivity on emergent ailments, such as cancer, metabolic, infectious, and neurological diseases.
Collapse
Affiliation(s)
- Vasco F Batista
- Laqv-requimte & Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Diana C G A Pinto
- Laqv-requimte & Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Artur M S Silva
- Laqv-requimte & Department of Chemistry, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
12
|
Bermúdez V, Tenconi PE, Giusto NM, Mateos MV. Canonical phospholipase D isoforms in visual function and ocular response to stress. Exp Eye Res 2022; 217:108976. [DOI: 10.1016/j.exer.2022.108976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/09/2022] [Accepted: 02/01/2022] [Indexed: 01/10/2023]
|
13
|
Chang YC, Chang PMH, Li CH, Chan MH, Lee YJ, Chen MH, Hsiao M. Aldolase A and Phospholipase D1 Synergistically Resist Alkylating Agents and Radiation in Lung Cancer. Front Oncol 2022; 11:811635. [PMID: 35127525 PMCID: PMC8813753 DOI: 10.3389/fonc.2021.811635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Exposure to alkylating agents and radiation may cause damage and apoptosis in cancer cells. Meanwhile, this exposure involves resistance and leads to metabolic reprogramming to benefit cancer cells. At present, the detailed mechanism is still unclear. Based on the profiles of several transcriptomes, we found that the activity of phospholipase D (PLD) and the production of specific metabolites are related to these events. Comparing several particular inhibitors, we determined that phospholipase D1 (PLD1) plays a dominant role over other PLD members. Using the existing metabolomics platform, we demonstrated that lysophosphatidylethanolamine (LPE) and lysophosphatidylcholine (LPC) are the most critical metabolites, and are highly dependent on aldolase A (ALDOA). We further demonstrated that ALDOA could modulate total PLD enzyme activity and phosphatidic acid products. Particularly after exposure to alkylating agents and radiation, the proliferation of lung cancer cells, autophagy, and DNA repair capabilities are enhanced. The above phenotypes are closely related to the performance of the ALDOA/PLD1 axis. Moreover, we found that ALDOA inhibited PLD2 activity and enzyme function through direct protein–protein interaction (PPI) with PLD2 to enhance PLD1 and additional carcinogenic features. Most importantly, the combination of ALDOA and PLD1 can be used as an independent prognostic factor and is correlated with several clinical parameters in lung cancer. These findings indicate that, based on the PPI status between ALDOA and PLD2, a combination of radiation and/or alkylating agents with regulating ALDOA-PLD1 may be considered as a new lung cancer treatment option.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Yi-Jang Lee
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Huang Chen
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Center of Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Michael Hsiao,
| |
Collapse
|
14
|
Cai M, Wang Z, Luu TTT, Zhang D, Finke B, He J, Tay LWR, Di Paolo G, Du G. PLD1 promotes reactive oxygen species production in vascular smooth muscle cells and injury-induced neointima formation. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159062. [PMID: 34610470 DOI: 10.1016/j.bbalip.2021.159062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Phospholipase D (PLD) generates the signaling lipid phosphatidic acid (PA) and has been known to mediate proliferation signal in vascular smooth muscle cells (VSMCs). However, it remains unclear how PLD contributes to vascular diseases. VSMC proliferation directly contributes to the development and progression of cardiovascular disease, such as atherosclerosis and restenosis after angioplasty. Using the mouse carotid artery ligation model, we find that deletion of Pld1 gene inhibits neointima formation of the injuried blood vessels. PLD1 deficiency reduces the proliferation of VSMCs in both injured artery and primary cultures through the inhibition of ERK1/2 and AKT signals. Immunohistochemical staining of injured artery and flow cytometry analysis of VSMCs shows a reduction of the levels of reactive oxygen species (ROS) in Pld1-/- VSMCs. An increase of intracellular ROS by hydrogen peroxide stimulation restored the reduced activities of ERK and AKT in Pld1-/- VSMCs, whereas a reduction of ROS by N-acetyl-l-cysteine (NAC) scavenger lowered their activity in wild-type VSMCs. These results indicate that PLD1 plays a critical role in neointima, and that PLD1 mediates VSMC proliferation signal through promoting the production of ROS. Therefore, inhibition of PLD1 may be used as a therapeutic approach to suppress neointimal formation in atherosclerosis and restenosis after angioplasty.
Collapse
Affiliation(s)
- Ming Cai
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Ziqing Wang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Thi Thu Trang Luu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Biochemistry and Cell Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Dakai Zhang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brian Finke
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jingquan He
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Li Wei Rachel Tay
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
15
|
Logozzi M, Di Raimo R, Mizzoni D, Fais S. What we know on the potential use of exosomes for nanodelivery. Semin Cancer Biol 2021; 86:13-25. [PMID: 34517111 DOI: 10.1016/j.semcancer.2021.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022]
Abstract
Antitumor therapy is taking into consideration the possibility to use natural nanovesicles, called exosomes, as an ideal delivery for both old and new anti-cancer molecules. This with the attempt to improve the efficacy, at the same time reducing the systemic toxicity of physical, chemical, and biological molecules. Exosomes may in fact increase the level of biomimetism, through simulating what really occurs in nature. Although extracellularly released vesicles include both microvesicles (MVs) and exosomes, only exosomes have the size that may be considered suitable for potential use to this purpose, also by analogy with the diffusely used artificial nanoparticles, such as lyposomes. In fact, recent reports have shown that exosomes are able to interact with target cells within an organ or at a distance using different mechanisms. Much is yet to be understood about exosomes, and currently, we are looking at the visible top of an iceberg, with most of what we have to understand on these nanovesicles still under the sea. In fact, we know that exosomes released by normal cells always trigger positive effects, while those released by cells in pathological condition, such as tumors may induce undesired, dangerous, and mostly unknown effects. To date we have many pre-clinical data available and possibly useful to think about a strategic use of exosomes as a delivery nanodevice in cancer treatment. However, this review wants to critically emphasize two important points actually hampering further discussion in the field : (i) the clinical data are virtually absent at the moment ; (ii) the best cellular source of exosomes to be used to deliver drugs is really far to be defined. Facing off these two points may well facilitate the attempt to figure out this very important issue for improving at the best future anti-cancer treatments.
Collapse
Affiliation(s)
- Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Rossella Di Raimo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Davide Mizzoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
16
|
Phospholipase Signaling in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33983572 DOI: 10.1007/978-981-32-9620-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Breast cancer progression results from subversion of multiple intra- or intercellular signaling pathways in normal mammary tissues and their microenvironment, which have an impact on cell differentiation, proliferation, migration, and angiogenesis. Phospholipases (PLC, PLD and PLA) are essential mediators of intra- and intercellular signaling. They hydrolyze phospholipids, which are major components of cell membrane that can generate many bioactive lipid mediators, such as diacylglycerol, phosphatidic acid, lysophosphatidic acid, and arachidonic acid. Enzymatic processing of phospholipids by phospholipases converts these molecules into lipid mediators that regulate multiple cellular processes, which in turn can promote breast cancer progression. Thus, dysregulation of phospholipases contributes to a number of human diseases, including cancer. This review describes how phospholipases regulate multiple cancer-associated cellular processes, and the interplay among different phospholipases in breast cancer. A thorough understanding of the breast cancer-associated signaling networks of phospholipases is necessary to determine whether these enzymes are potential targets for innovative therapeutic strategies.
Collapse
|
17
|
Barr JL, Zhao P, Brailoiu GC, Brailoiu E. Choline-Sigma-1R as an Additional Mechanism for Potentiation of Orexin by Cocaine. Int J Mol Sci 2021; 22:5160. [PMID: 34068146 PMCID: PMC8152999 DOI: 10.3390/ijms22105160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/27/2022] Open
Abstract
Orexin A, an endogenous peptide involved in several functions including reward, acts via activation of orexin receptors OX1 and OX2, Gq-coupled GPCRs. We examined the effect of a selective OX1 agonist, OXA (17-33) on cytosolic calcium concentration, [Ca2+]i, in neurons of nucleus accumbens, an important area in the reward circuit. OXA (17-33) increased [Ca2+]i in a dose-dependent manner; the effect was prevented by SB-334867, a selective OX1 receptors antagonist. In Ca2+-free saline, the OXA (17-33)-induced increase in [Ca2+]i was not affected by pretreatment with bafilomycin A1, an endo-lysosomal calcium disrupter, but was blocked by 2-APB and xestospongin C, antagonists of inositol-1,4,5-trisphosphate (IP3) receptors. Pretreatment with VU0155056, PLD inhibitor, or BD-1047 and NE-100, Sigma-1R antagonists, reduced the [Ca2+]i response elicited by OXA (17-33). Cocaine potentiated the increase in [Ca2+]i by OXA (17-33); the potentiation was abolished by Sigma-1R antagonists. Our results support an additional signaling mechanism for orexin A-OX1 via choline-Sigma-1R and a critical role for Sigma-1R in the cocaine-orexin A interaction in nucleus accumbens neurons.
Collapse
Affiliation(s)
- Jeffrey L. Barr
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (J.L.B.); (P.Z.)
| | - Pingwei Zhao
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (J.L.B.); (P.Z.)
| | - G. Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (J.L.B.); (P.Z.)
| |
Collapse
|
18
|
Bowling FZ, Frohman MA, Airola MV. Structure and regulation of human phospholipase D. Adv Biol Regul 2021; 79:100783. [PMID: 33495125 DOI: 10.1016/j.jbior.2020.100783] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
Mammalian phospholipase D (PLD) generates phosphatidic acid, a dynamic lipid secondary messenger involved with a broad spectrum of cellular functions including but not limited to metabolism, migration, and exocytosis. As a promising pharmaceutical target, the biochemical properties of PLD have been well characterized. This has led to the recent crystal structures of human PLD1 and PLD2, the development of PLD specific pharmacological inhibitors, and the identification of cellular regulators of PLD. In this review, we discuss the PLD1 and PLD2 structures, PLD inhibition by small molecules, and the regulation of PLD activity by effector proteins and lipids.
Collapse
Affiliation(s)
- Forrest Z Bowling
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Michael A Frohman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Michael V Airola
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
19
|
Auclair N, Sané AT, Delvin E, Spahis S, Levy E. Phospholipase D as a Potential Modulator of Metabolic Syndrome: Impact of Functional Foods. Antioxid Redox Signal 2021; 34:252-278. [PMID: 32586106 DOI: 10.1089/ars.2020.8081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Cardiometabolic disorders (CMD) are composed of a plethora of metabolic dysfunctions such as dyslipidemia, nonalcoholic fatty liver disease, insulin resistance, and hypertension. The development of these disorders is highly linked to inflammation and oxidative stress (OxS), two metabolic states closely related to physiological and pathological conditions. Given the drastically rising CMD prevalence, the discovery of new therapeutic targets/novel nutritional approaches is of utmost importance. Recent Advances: The tremendous progress in methods/technologies and animal modeling has allowed the clarification of phospholipase D (PLD) critical roles in multiple cellular processes, whether directly or indirectly via phosphatidic acid, the lipid product mediating signaling functions. In view of its multiple features and implications in various diseases, PLD has emerged as a drug target. Critical Issues: Although insulin stimulates PLD activity and, in turn, PLD regulates insulin signaling, the impact of the two important PLD isoforms on the metabolic syndrome components remains vague. Therefore, after outlining PLD1/PLD2 characteristics and functions, their role in inflammation, OxS, and CMD has been analyzed and critically reported in the present exhaustive review. The influence of functional foods and nutrients in the regulation of PLD has also been examined. Future Directions: Available evidence supports the implication of PLD in CMD, but only few studies emphasize its mechanisms of action and specific regulation by nutraceutical compounds. Therefore, additional investigations are first needed to clarify the functional role of nutraceutics and, second, to elucidate whether targeting PLDs with food compounds represents an appropriate therapeutic strategy to treat CMD. Antioxid. Redox Signal. 34, 252-278.
Collapse
Affiliation(s)
- Nickolas Auclair
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology & Physiology and Université de Montréal, Montreal, Quebec, Canada
| | - Alain T Sané
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Edgard Delvin
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Schohraya Spahis
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology & Physiology and Université de Montréal, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
20
|
Ghoroghi S, Mary B, Larnicol A, Asokan N, Klein A, Osmani N, Busnelli I, Delalande F, Paul N, Halary S, Gros F, Fouillen L, Haeberle AM, Royer C, Spiegelhalter C, André-Grégoire G, Mittelheisser V, Detappe A, Murphy K, Timpson P, Carapito R, Blot-Chabaud M, Gavard J, Carapito C, Vitale N, Lefebvre O, Goetz JG, Hyenne V. Ral GTPases promote breast cancer metastasis by controlling biogenesis and organ targeting of exosomes. eLife 2021; 10:61539. [PMID: 33404012 PMCID: PMC7822591 DOI: 10.7554/elife.61539] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer extracellular vesicles (EVs) shuttle at distance and fertilize pre-metastatic niches facilitating subsequent seeding by tumor cells. However, the link between EV secretion mechanisms and their capacity to form pre-metastatic niches remains obscure. Using mouse models, we show that GTPases of the Ral family control, through the phospholipase D1, multi-vesicular bodies homeostasis and tune the biogenesis and secretion of pro-metastatic EVs. Importantly, EVs from RalA or RalB depleted cells have limited organotropic capacities in vivoand are less efficient in promoting metastasis. RalA and RalB reduce the EV levels of the adhesion molecule MCAM/CD146, which favors EV-mediated metastasis by allowing EVs targeting to the lungs. Finally, RalA, RalB, and MCAM/CD146, are factors of poor prognosis in breast cancer patients. Altogether, our study identifies RalGTPases as central molecules linking the mechanisms of EVs secretion and cargo loading to their capacity to disseminate and induce pre-metastatic niches in a CD146-dependent manner.
Collapse
Affiliation(s)
- Shima Ghoroghi
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Benjamin Mary
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Annabel Larnicol
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Nandini Asokan
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Annick Klein
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Naël Osmani
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ignacio Busnelli
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - François Delalande
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC UMR 7178, CNRS, Université de Strasbourg, Strasbourg, France
| | - Nicodème Paul
- Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,INSERM UMR_S1109, Genomax, Strasbourg, France
| | - Sébastien Halary
- CNRS, UMR 7245 MCAM, Muséum National d'Histoire Naturelle de Paris, Paris, France
| | - Frédéric Gros
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Laetitia Fouillen
- Université de Bordeaux, CNRS, Laboratoire de Biogenèse Membranaire, UMR 5200, Villenave d'Ornon, France
| | - Anne-Marie Haeberle
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Cathy Royer
- Plateforme Imagerie In Vitro, CNRS UPS 3156, Strasbourg, France
| | - Coralie Spiegelhalter
- IGBMC Imaging Center CNRS (UMR7104)/ INSERM (U1258)/ Université de Strasbourg, Illkirch, France
| | - Gwennan André-Grégoire
- Team SOAP, CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,Integrated Center for Oncology, ICO, St-Herblain, France
| | - Vincent Mittelheisser
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Nanotranslational laboratory, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Alexandre Detappe
- Nanotranslational laboratory, Institut de Cancérologie Strasbourg Europe, Strasbourg, France.,Équipe de synthèse pour l'analyse (SynPA), Institut Pluridisciplinaire Hubert Curien (IPHC), UMR7178, CNRS/Université de Strasbourg, Strasbourg, France
| | - Kendelle Murphy
- Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Paul Timpson
- Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Raphaël Carapito
- Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,INSERM UMR_S1109, Genomax, Strasbourg, France
| | | | - Julie Gavard
- Team SOAP, CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,Integrated Center for Oncology, ICO, St-Herblain, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC UMR 7178, CNRS, Université de Strasbourg, Strasbourg, France
| | - Nicolas Vitale
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Olivier Lefebvre
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Vincent Hyenne
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,CNRS SNC5055, Strasbourg, France
| |
Collapse
|
21
|
Krzystanek M, Krzystanek E, Skałacka K, Pałasz A. Enhancement in Phospholipase D Activity as a New Proposed Molecular Mechanism of Haloperidol-Induced Neurotoxicity. Int J Mol Sci 2020; 21:ijms21239265. [PMID: 33291692 PMCID: PMC7730321 DOI: 10.3390/ijms21239265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 11/25/2022] Open
Abstract
Membrane phospholipase D (PLD) is associated with numerous neuronal functions, such as axonal growth, synaptogenesis, formation of secretory vesicles, neurodegeneration, and apoptosis. PLD acts mainly on phosphatidylcholine, from which phosphatidic acid (PA) and choline are formed. In turn, PA is a key element of the PLD-dependent secondary messenger system. Changes in PLD activity are associated with the mechanism of action of olanzapine, an atypical antipsychotic. The aim of the present study was to assess the effect of short-term administration of the first-generation antipsychotic drugs haloperidol, chlorpromazine, and fluphenazine on membrane PLD activity in the rat brain. Animals were sacrificed for a time equal to the half-life of the antipsychotic drug in the brain, then the membranes in which PLD activity was determined were isolated from the tissue. The results indicate that only haloperidol in a higher dose increases the activity of phospholipase D. Such a mechanism of action of haloperidol has not been described previously. Induction of PLD activity by haloperidol may be related to its mechanism of cytotoxicity. The finding could justify the use of PLD inhibitors as protective drugs against the cytotoxicity of first-generation antipsychotic drugs like haloperidol.
Collapse
Affiliation(s)
- Marek Krzystanek
- Department and Clinic of Psychiatric Rehabilitation, Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences, Medical School of Silesia in Katowice, Ziołowa 45/47, 40-635 Katowice, Poland
- Correspondence: or
| | - Ewa Krzystanek
- Department of Neurology, Faculty of Medical Sciences, Medical School of Silesia in Katowice, Medyków 14, 40-772 Katowice, Poland;
| | - Katarzyna Skałacka
- Institute of Psychology, University of Opole, Kopernika 11A Street, 45-040 Opole, Poland;
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences, Medical School of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland;
| |
Collapse
|
22
|
Arhab Y, Bessaa K, Abla H, Aydin M, Rahier R, Comte A, Brizuela L, Mebarek S, Perret F, Cherrier MV, Abousalham A, Noiriel A. Phospholipase D inhibitors screening: Probing and evaluation of ancient and novel molecules. Int J Biol Macromol 2020; 166:1131-1140. [PMID: 33161081 DOI: 10.1016/j.ijbiomac.2020.10.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/03/2020] [Accepted: 10/31/2020] [Indexed: 11/26/2022]
Abstract
Phospholipase D (PLD) is a ubiquitous enzyme that cleaves the distal phosphoester bond of phospholipids generating phosphatidic acid (PA). In plants, PA is involved in numerous cell responses triggered by stress. Similarly, in mammals, PA is also a second messenger involved in tumorigenesis. PLD is nowadays considered as a therapeutic target and blocking its activity with specific inhibitors constitutes a promising strategy to treat cancers. Starting from already described PLD inhibitors, this study aims to investigate the effect of their structural modifications on the enzyme's activity, as well as identifying new potent inhibitors of eukaryotic PLDs. Being able to purify the plant PLD from Vigna unguiculata (VuPLD), we obtained a SAXS model of its structure. We then used a fluorescence-based test suitable for high-throughput screening to review the effect of eukaryotic PLD inhibitors described in the literature. In this regard, we found that only few molecules were in fact able to inhibit VuPLD and we confirmed that vanadate is the most potent of all with an IC50 around 58 μM. Moreover, the small-scale screening of a chemical library of 3120 compounds allowed us to optimize the different screening's steps and paved the way towards the discovery of new potent inhibitors.
Collapse
Affiliation(s)
- Yani Arhab
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Karim Bessaa
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Houda Abla
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Meryem Aydin
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Renaud Rahier
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Arnaud Comte
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Chimiothèque, Bât Lederer, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Leyre Brizuela
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Saïda Mebarek
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Florent Perret
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Chimie Supramoléculaire Appliquée (CSAp), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Mickaël V Cherrier
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins, F-38000 Grenoble, France
| | - Abdelkarim Abousalham
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France
| | - Alexandre Noiriel
- Univ Lyon, Université Lyon 1, Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, Métabolisme, Enzymes et Mécanismes Moléculaires (MEM(2)), Bât Raulin, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne cedex, France.
| |
Collapse
|
23
|
Hwang WC, Kang DW, Kang Y, Jang Y, Kim JA, Min DS. Inhibition of phospholipase D2 augments histone deacetylase inhibitor-induced cell death in breast cancer cells. Biol Res 2020; 53:34. [PMID: 32998768 PMCID: PMC7528251 DOI: 10.1186/s40659-020-00294-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/10/2020] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Histone deacetylase (HDAC) inhibitors are promising anticancer drugs but their effect on tumor treatment has been disappointing mainly due to the acquisition of HDAC inhibitor resistance. However, the mechanisms underlying such resistance remain unclear. METHODS In this study, we performed Western blot, q-PCR, and promoter assay to examine the expression of HDAC inhibitor-induced phospholipase D2 (PLD2) in MDA-MB231and MDA-MB435 breast cancer cells. Apoptosis and proliferation were analyzed by flow cytometry. In addition to invasion and migration assay, angiogenesis was further measured using in vitro tube formation and chick embryo chorioallantoic membrane model. RESULTS HDAC inhibitors including suberoylanilide hydroxamic acid (SAHA), trichostatin, and apicidin, induce expression of PLD2 in a transcriptional level. SAHA upregulates expression of PLD2 via protein kinase C-ζ in breast cancer cells and increases the enzymatic activity of PLD. The combination treatment of SAHA with PLD2 inhibitor significantly enhances cell death in breast cancer cells. Phosphatidic acid, a product of PLD activity, prevented apoptosis promoted by cotreatment with SAHA and PLD2 inhibitor, suggesting that SAHA-induced PLD2 expression and subsequent activation of PLD2 might confers resistance of breast cancer cells to HDAC inhibitor. The combinational treatment of the drugs significantly suppressed invasion, migration, and angiogenesis, compared with that of either treatment. CONCLUSION These findings provide further insight into elucidating the advantages of combination therapy with HDAC and PLD2 inhibitors over single-agent strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 609-735, South Korea.,College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, 609-735, South Korea
| | - Youra Kang
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Younghoon Jang
- Department of Biology and Chemistry, Changwon National University, Changwon, South Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea.
| |
Collapse
|
24
|
Armistead J, Hatzold J, van Roye A, Fahle E, Hammerschmidt M. Entosis and apical cell extrusion constitute a tumor-suppressive mechanism downstream of Matriptase. J Cell Biol 2020; 219:132730. [PMID: 31819976 PMCID: PMC7041680 DOI: 10.1083/jcb.201905190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/02/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Armistead et al. show that in a bilayered epithelium in vivo, apical cell extrusion of basal cells is achieved via their engulfment by surface cells. In zebrafish hai1a mutants, this constitutes a tumor-suppressive mechanism, revealing a double face of Matriptase. The type II transmembrane serine protease Matriptase 1 (ST14) is commonly known as an oncogene, yet it also plays an understudied role in suppressing carcinogenesis. This double face is evident in the embryonic epidermis of zebrafish loss-of-function mutants in the cognate Matriptase inhibitor Hai1a (Spint1a). Mutant embryos display epidermal hyperplasia, but also apical cell extrusions, during which extruding outer keratinocytes carry out an entosis-like engulfment and entrainment of underlying basal cells, constituting a tumor-suppressive effect. These counteracting Matriptase effects depend on EGFR and the newly identified mediator phospholipase D (PLD), which promotes both mTORC1-dependent cell proliferation and sphingosine-1-phosphate (S1P)–dependent entosis and apical cell extrusion. Accordingly, hypomorphic hai1a mutants heal spontaneously, while otherwise lethal hai1a amorphs are efficiently rescued upon cotreatment with PLD inhibitors and S1P. Together, our data elucidate the mechanisms underlying the double face of Matriptase function in vivo and reveal the potential use of combinatorial carcinoma treatments when such double-face mechanisms are involved.
Collapse
Affiliation(s)
- Joy Armistead
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Julia Hatzold
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Anna van Roye
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Evelin Fahle
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
25
|
Kang DW, Hwang WC, Noh YN, Park KS, Min DS. Phospholipase D1 inhibition sensitizes glioblastoma to temozolomide and suppresses its tumorigenicity. J Pathol 2020; 252:304-316. [PMID: 32725633 PMCID: PMC7693208 DOI: 10.1002/path.5519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/12/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
Resistance of glioblastoma to the chemotherapeutic compound temozolomide is associated with the presence of glioblastoma stem cells in glioblastoma and is a key obstacle for the poor prognosis of glioblastoma. Here, we show that phospholipase D1 is elevated in CD44High glioblastoma stem cells and in glioblastoma, especially recurring glioblastoma. Phospholipase D1 elevation positively correlated with the level of CD44 and poor prognosis in glioblastoma patients. Temozolomide significantly upregulated the expression of phospholipase D1 in the low and moderate CD44 populations of glioblastoma stem cells, but not in the CD44High population in which phospholipase D1 is highly expressed. Phospholipase D1 conferred resistance to temozolomide in CD44High glioblastoma stem cells and increased their self‐renewal capacity and maintenance. Phospholipase D1 expression significantly correlated with levels of temozolomide resistance factors, which were suppressed by microRNA‐320a and ‐4496 induced by phospholipase D1 inhibition. Genetic and pharmacological targeting of phospholipase D1 attenuated glioblastoma stem cell‐derived intracranial tumors of glioblastoma using the microRNAs, and improved survival. Treatment solely with temozolomide produced no benefits on the glioblastoma, whereas in combination, phospholipase D1 inhibition sensitized glioblastoma stem cells to temozolomide and reduced glioblastoma tumorigenesis. Together, these findings indicate that phospholipase D1 inhibition might overcome resistance to temozolomide and represents a potential treatment strategy for glioblastoma. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Yu Na Noh
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Kang Seo Park
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea.,Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| |
Collapse
|
26
|
Belkacemi L, Darmani NA. Dopamine receptors in emesis: Molecular mechanisms and potential therapeutic function. Pharmacol Res 2020; 161:105124. [PMID: 32814171 DOI: 10.1016/j.phrs.2020.105124] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/20/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
Dopamine is a member of the catecholamine family and is associated with multiple physiological functions. Together with its five receptor subtypes, dopamine is closely linked to neurological disorders such as schizophrenia, Parkinson's disease, depression, attention deficit-hyperactivity, and restless leg syndrome. Unfortunately, several dopamine receptor-based agonists used to treat some of these diseases cause nausea and vomiting as impending side-effects. The high degree of cross interactions of dopamine receptor ligands with many other targets including G-protein coupled receptors, transporters, enzymes, and ion-channels, add to the complexity of discovering new targets for the treatment of nausea and vomiting. Using activation status of signaling cascades as mechanism-based biomarkers to foresee drug sensitivity combined with the development of dopamine receptor-based biased agonists may hold great promise and seems as the next step in drug development for the treatment of such multifactorial diseases. In this review, we update the present knowledge on dopamine and dopamine receptors and their potential roles in nausea and vomiting. The pre- and clinical evidence provided in this review supports the implication of both dopamine and dopamine receptor agonists in the incidence of emesis. Besides the conventional dopaminergic antiemetic drugs, potential novel antiemetic targeting emetic protein signaling cascades may offer superior selectivity profile and potency.
Collapse
Affiliation(s)
- Louiza Belkacemi
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Nissar A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766, USA.
| |
Collapse
|
27
|
Mono- and Poly-unsaturated Phosphatidic Acid Regulate Distinct Steps of Regulated Exocytosis in Neuroendocrine Cells. Cell Rep 2020; 32:108026. [DOI: 10.1016/j.celrep.2020.108026] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/16/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
|
28
|
Borel M, Cuvillier O, Magne D, Mebarek S, Brizuela L. Increased phospholipase D activity contributes to tumorigenesis in prostate cancer cell models. Mol Cell Biochem 2020; 473:263-279. [PMID: 32661773 DOI: 10.1007/s11010-020-03827-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 07/04/2020] [Indexed: 12/30/2022]
Abstract
Prostate cancer (PCa) is the most frequent cancer among men and the first cause of death over 65. Approximately 90% of patients with advanced disease will develop bone metastasis, which dramatically reduces long-term survival. Therefore, effective therapies need to be developed, especially when disease is still well-localized. Phospholipase D (PLD), an enzyme that hydrolyzes phosphatidylcholine to yield phosphatidic acid, regulates several cellular functions as proliferation, survival, migration or vesicular trafficking. PLD is implicated in numerous diseases such as neurodegenerative, cardiovascular, autoimmune disorders or cancer. Indeed, PLD controls different aspects of oncogenesis including tumor progression and resistance to targeted therapies such as radiotherapy. PLD1 and PLD2 are the only isoforms with catalytic activity involved in cancer. Surprisingly, studies deciphering the role of PLD in the pathophysiology of PCa are scarce. Here we describe the correlation between PLD activity and PLD1 and PLD2 expression in PCa bone metastasis-derived cell lines C4-2B and PC-3. Next, by using PLD pharmacological inhibitors and RNA interference strategy, we validate the implication of PLD1 and PLD2 in cell viability, clonogenicity and proliferation of C4-2B and PC-3 cells and in migration capacity of PC-3 cells. Last, we show an increase in PLD activity as well as PLD2 protein expression during controlled starvation of PC-3 cells, concomitant with an augmentation of its migration capacity. Specifically, upregulation of PLD activity appears to be PKC-independent. Taken together, our results indicate that PLD, and in particular PLD2, could be considered as a potential therapeutic target for the treatment of PCa-derived bone metastasis.
Collapse
Affiliation(s)
- Mathieu Borel
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, 69622, Lyon, France
| | - Olivier Cuvillier
- Université de Toulouse, UPS, CNRS UMR 5089, Institut de Pharmacologie et de Biologie Structurale, IPBS, 31077, Toulouse Cedex, France
| | - David Magne
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, 69622, Lyon, France
| | - Saida Mebarek
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, 69622, Lyon, France
| | - Leyre Brizuela
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, ICBMS, 69622, Lyon, France.
| |
Collapse
|
29
|
Barisano D, Frohman MA. Roles for Phospholipase D1 in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:77-87. [PMID: 32578172 DOI: 10.1007/978-3-030-43093-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The lipid-modifying signal transduction enzyme phospholipase D (PLD) has been proposed to have roles in oncogenic processes for well-on 30 years, with most of the early literature focused on potential functions for PLD in the biology of the tumor cells themselves. While such roles remain under investigation, evidence has also now been generated to support additional roles for PLD, in particular PLD1, in the tumor microenvironment, including effects on neoangiogenesis, the supply of nutrients, interactions of platelets with circulating cancer cells, the response of the immune system, and exosome biology. Here, we review these lines of investigation, accompanied by a discussion of the limitations of the existing studies and some cautionary notes regarding the study and interpretation of PLD function using model systems.
Collapse
Affiliation(s)
- Daniela Barisano
- Center for Developmental Genetics and the Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Michael A Frohman
- Center for Developmental Genetics and the Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, NY, USA.
| |
Collapse
|
30
|
Lingelem ABD, Kavaliauskiene S, Halsne R, Klokk TI, Surma MA, Klose C, Skotland T, Sandvig K. Diacylglycerol kinase and phospholipase D inhibitors alter the cellular lipidome and endosomal sorting towards the Golgi apparatus. Cell Mol Life Sci 2020; 78:985-1009. [PMID: 32447426 PMCID: PMC7897626 DOI: 10.1007/s00018-020-03551-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022]
Abstract
The membrane lipids diacylglycerol (DAG) and phosphatidic acid (PA) are important second messengers that can regulate membrane transport by recruiting proteins to the membrane and by altering biophysical membrane properties. DAG and PA are involved in the transport from the Golgi apparatus to endosomes, and we have here investigated whether changes in these lipids might be important for regulation of transport to the Golgi using the protein toxin ricin. Modulation of DAG and PA levels using DAG kinase (DGK) and phospholipase D (PLD) inhibitors gave a strong increase in retrograde ricin transport, but had little impact on ricin recycling or degradation. Inhibitor treatment strongly affected the endosome morphology, increasing endosomal tubulation and size. Furthermore, ricin was present in these tubular structures together with proteins known to regulate retrograde transport. Using siRNA to knock down different isoforms of PLD and DGK, we found that several isoforms of PLD and DGK are involved in regulating ricin transport to the Golgi. Finally, by performing lipidomic analysis we found that the DGK inhibitor gave a weak, but expected, increase in DAG levels, while the PLD inhibitor gave a strong and unexpected increase in DAG levels, showing that it is important to perform lipidomic analysis when using inhibitors of lipid metabolism.
Collapse
Affiliation(s)
- Anne Berit Dyve Lingelem
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Forensic Biology, Oslo University Hospital, Oslo, Norway
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ruth Halsne
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Forensic Biology, Oslo University Hospital, Oslo, Norway
| | - Tove Irene Klokk
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Regional Committees for Medical and Health Research Ethics, University of Oslo, Oslo, Norway
| | | | | | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway. .,Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
31
|
Yoo HJ, Hwang WC, Min DS. Targeting of Phospholipase D1 Ameliorates Collagen-Induced Arthritis via Modulation of Treg and Th17 Cell Imbalance and Suppression of Osteoclastogenesis. Int J Mol Sci 2020; 21:ijms21093230. [PMID: 32370217 PMCID: PMC7247592 DOI: 10.3390/ijms21093230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Phospholipase D1 (PLD1) plays a crucial role in various inflammatory and autoimmune diseases. Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease. However, the role of PLD1 in the pathogenesis of RA remains unknown. Here, we first investigated the role and effects of PLD1 in collagen-induced arthritis (CIA) and found that genetic and pharmacological inhibition of PLD1 in DBA1/J mice with CIA reduced the incidence of CIA, decreased the clinical score, and abrogated disease symptoms including infiltration of leukocytes, synovial inflammation, bone erosion, and cartilage destruction. Moreover, ablation and inhibition of PLD1 suppressed the production of type II collagen-specific IgG2a autoantibody and proinflammatory cytokines, accompanied by an increase in the regulatory T (Treg) cell population and a decrease in the Th17 cell population in CIA mice. The PLD1 inhibitor also promoted differentiation of Treg cells and suppressed differentiation of Th17 cells in vitro. Furthermore, the PLD1 inhibitor attenuated pathologic bone destruction in CIA mice by suppressing osteoclastogenesis and bone resorption. Thus, our findings indicate that the targeting of PLD1 can ameliorate CIA by modulating the imbalance of Treg and Th17 cells and suppressing osteoclastogenesis, which might be a novel strategy to treat autoimmune diseases, such as RA.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/prevention & control
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/prevention & control
- Benzimidazoles/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Cytokines/blood
- Disease Models, Animal
- Knee Joint/drug effects
- Knee Joint/metabolism
- Knee Joint/pathology
- Male
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Phospholipase D/antagonists & inhibitors
- Phospholipase D/genetics
- Phospholipase D/metabolism
- Piperidines/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- X-Ray Microtomography
Collapse
Affiliation(s)
- Hyun Jung Yoo
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46241, Korea; (H.J.Y.); (W.C.H.)
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46241, Korea; (H.J.Y.); (W.C.H.)
- College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Correspondence: ; Tel.: +82-32-749-4522
| |
Collapse
|
32
|
Hou L, Tian C, Yan Y, Zhang L, Zhang H, Zhang Z. Manganese-Based Nanoactivator Optimizes Cancer Immunotherapy via Enhancing Innate Immunity. ACS NANO 2020; 14:3927-3940. [PMID: 32298077 DOI: 10.1021/acsnano.9b06111] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are essential components of the innate immune sensors to cytosolic DNA and elicit type I interferon (IFN). Recent studies have revealed that manganese (Mn) can enhance cGAS and STING activation to viral infection. However, the role of Mn in antitumor immunity has not been explored. Here, we designed a nanoactivator, which can induce the presence of DNA in cytoplasm and simultaneously elevate Mn2+ accumulation within tumor cells. In detail, amorphous porous manganese phosphate (APMP) NPs that are highly responsive to tumor microenvironment were employed to construct doxorubicin (DOX)-loaded and phospholipid (PL)-coated hybrid nanoparticles (PL/APMP-DOX NPs). PL/APMP-DOX NPs were stably maintained during systemic circulation, but triggered to release DOX for inducing DNA damage and Mn2+ to augment cGAS/STING activity. We found that PL/APMP-DOX NPs with superior tumor-targeting capacity boosted dendritic cell maturation and increased cytotoxic T lymphocyte infiltration as well as natural killer cell recruitment into the tumor site. Furthermore, the NPs increased production of type I IFN and secretion of pro-inflammatory cytokines (for example, TNF-α and IL-6). Consequently, PL/APMP-DOX NPs exhibited excellent antitumor efficacy and prolonged the lifespan of the tumor-bearing mice. Collectively, we developed a PL-decorated Mn-based hybrid nanoactivator to intensify immune activation and that might provide therapeutic potential for caner immunotherapy.
Collapse
Affiliation(s)
- Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, and Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Chunyu Tian
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
- Modern Analysis and Computer Center of Zhengzhou University, Zhengzhou 450001, China
| | - Yingshan Yan
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
- Modern Analysis and Computer Center of Zhengzhou University, Zhengzhou 450001, China
| | - Lewen Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, and Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Huijuan Zhang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
33
|
Lin D, Yao H, Jia L, Tan J, Xu Z, Zheng W, Xue H. Phospholipase D-derived phosphatidic acid promotes root hair development under phosphorus deficiency by suppressing vacuolar degradation of PIN-FORMED2. THE NEW PHYTOLOGIST 2020; 226:142-155. [PMID: 31745997 PMCID: PMC7065129 DOI: 10.1111/nph.16330] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/10/2019] [Indexed: 05/03/2023]
Abstract
Root hair development is crucial for phosphate absorption, but how phosphorus deficiency affects root hair initiation and elongation remains unclear. We demonstrated the roles of auxin efflux carrier PIN-FORMED2 (PIN2) and phospholipase D (PLD)-derived phosphatidic acid (PA), a key signaling molecule, in promoting root hair development in Arabidopsis thaliana under a low phosphate (LP) condition. Root hair elongation under LP conditions was greatly suppressed in pin2 mutant or under treatment with a PLDζ2-specific inhibitor, revealing that PIN2 and polar auxin transport and PLDζ2-PA are crucial in LP responses. PIN2 was accumulated and degraded in the vacuole under a normal phosphate (NP) condition, whereas its vacuolar accumulation was suppressed under the LP or NP plus PA conditions. Vacuolar accumulation of PIN2 was increased in pldζ2 mutants under LP conditions. Increased or decreased PIN2 vacuolar accumulation is not observed in sorting nexin1 (snx1) mutant, indicating that vacuolar accumulation of PIN2 is mediated by SNX1 and the relevant trafficking process. PA binds to SNX1 and promotes its accumulation at the plasma membrane, especially under LP conditions, and hence promotes root hair development by suppressing the vacuolar degradation of PIN2. We uncovered a link between PLD-derived PA and SNX1-dependent vacuolar degradation of PIN2 in regulating root hair development under phosphorus deficiency.
Collapse
Affiliation(s)
- De‐Li Lin
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop ScienceCollege of Life SciencesHenan Agricultural University450002ZhengzhouChina
| | - Hong‐Yan Yao
- National Key Laboratory of Plant Molecular GeneticsCAS Center for Excellence in Molecular Plant SciencesShanghai Institute of Plant Physiology and EcologyChinese academy of Sciences200032ShanghaiChina
| | - Li‐Hua Jia
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop ScienceCollege of Life SciencesHenan Agricultural University450002ZhengzhouChina
| | - Jin‐Fang Tan
- College of Resource and EnvironmentHenan Agricultural University450002ZhengzhouChina
| | - Zhi‐Hong Xu
- National Key Laboratory of Plant Molecular GeneticsCAS Center for Excellence in Molecular Plant SciencesShanghai Institute of Plant Physiology and EcologyChinese academy of Sciences200032ShanghaiChina
| | - Wen‐Ming Zheng
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop ScienceCollege of Life SciencesHenan Agricultural University450002ZhengzhouChina
| | - Hong‐Wei Xue
- National Key Laboratory of Plant Molecular GeneticsCAS Center for Excellence in Molecular Plant SciencesShanghai Institute of Plant Physiology and EcologyChinese academy of Sciences200032ShanghaiChina
- Joint Center for Single Cell BiologySchool of Agriculture and BiologyShanghai Jiao Tong University200240ShanghaiChina
| |
Collapse
|
34
|
Crystal structure of human PLD1 provides insight into activation by PI(4,5)P 2 and RhoA. Nat Chem Biol 2020; 16:400-407. [PMID: 32198492 PMCID: PMC7117805 DOI: 10.1038/s41589-020-0499-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 02/10/2020] [Indexed: 11/08/2022]
Abstract
The signal transduction enzyme phospholipase D1 (PLD1) hydrolyzes phosphatidylcholine to generate the lipid second-messenger phosphatidic acid, which plays roles in disease processes such as thrombosis and cancer. PLD1 is directly and synergistically regulated by protein kinase C, Arf and Rho GTPases, and the membrane lipid phosphatidylinositol-4,5-bisphosphate (PIP2). Here, we present a 1.8 Å-resolution crystal structure of the human PLD1 catalytic domain, which is characterized by a globular fold with a funnel-shaped hydrophobic cavity leading to the active site. Adjacent is a PIP2-binding polybasic pocket at the membrane interface that is essential for activity. The C terminus folds into and contributes part of the catalytic pocket, which harbors a phosphohistidine that mimics an intermediate stage of the catalytic cycle. Mapping of PLD1 mutations that disrupt RhoA activation identifies the RhoA-PLD1 binding interface. This structure sheds light on PLD1 regulation by lipid and protein effectors, enabling rationale inhibitor design for this well-studied therapeutic target.
Collapse
|
35
|
Tei R, Baskin JM. Spatiotemporal control of phosphatidic acid signaling with optogenetic, engineered phospholipase Ds. J Cell Biol 2020; 219:e201907013. [PMID: 31999306 PMCID: PMC7054994 DOI: 10.1083/jcb.201907013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/09/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Phosphatidic acid (PA) is both a central phospholipid biosynthetic intermediate and a multifunctional lipid second messenger produced at several discrete subcellular locations. Organelle-specific PA pools are believed to play distinct physiological roles, but tools with high spatiotemporal control are lacking for unraveling these pleiotropic functions. Here, we present an approach to precisely generate PA on demand on specific organelle membranes. We exploited a microbial phospholipase D (PLD), which produces PA by phosphatidylcholine hydrolysis, and the CRY2-CIBN light-mediated heterodimerization system to create an optogenetic PLD (optoPLD). Directed evolution of PLD using yeast membrane display and IMPACT, a chemoenzymatic method for visualizing cellular PLD activity, yielded a panel of optoPLDs whose range of catalytic activities enables mimicry of endogenous, physiological PLD signaling. Finally, we applied optoPLD to elucidate that plasma membrane, but not intracellular, pools of PA can attenuate the oncogenic Hippo signaling pathway. OptoPLD represents a powerful and precise approach for revealing spatiotemporally defined physiological functions of PA.
Collapse
Affiliation(s)
| | - Jeremy M. Baskin
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| |
Collapse
|
36
|
Yao Y, Wang X, Li H, Fan J, Qian X, Li H, Xu Y. Phospholipase D as a key modulator of cancer progression. Biol Rev Camb Philos Soc 2020; 95:911-935. [PMID: 32073216 DOI: 10.1111/brv.12592] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
The phospholipase D (PLD) family has a ubiquitous expression in cells. PLD isoforms (PLDs) and their hydrolysate phosphatidic acid (PA) have been demonstrated to engage in multiple stages of cancer progression. Aberrant expression of PLDs, especially PLD1 and PLD2, has been detected in various cancers. Inhibition or elimination of PLDs activity has been shown to reduce tumour growth and metastasis. PLDs and PA also serve as downstream effectors of various cell-surface receptors, to trigger and regulate propagation of intracellular signals in the process of tumourigenesis and metastasis. Here, we discuss recent advances in understanding the functions of PLDs and PA in discrete stages of cancer progression, including cancer cell growth, invasion and migration, and angiogenesis, with special emphasis on the tumour-associated signalling pathways mediated by PLDs and PA and the functional importance of PLDs and PA in cancer therapy.
Collapse
Affiliation(s)
- Yuanfa Yao
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyi Wang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Clinical Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hanbing Li
- Institute of Pharmacology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jiannan Fan
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Xiaohan Qian
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Respiratory Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Li
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
37
|
Metrick CM, Peterson EA, Santoro JC, Enyedy IJ, Murugan P, Chen T, Michelsen K, Cullivan M, Spilker KA, Kumar PR, May-Dracka TL, Chodaparambil JV. Human PLD structures enable drug design and characterization of isoenzyme selectivity. Nat Chem Biol 2020; 16:391-399. [PMID: 32042197 DOI: 10.1038/s41589-019-0458-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Phospholipase D enzymes (PLDs) are ubiquitous phosphodiesterases that produce phosphatidic acid (PA), a key second messenger and biosynthetic building block. Although an orthologous bacterial Streptomyces sp. strain PMF PLD structure was solved two decades ago, the molecular basis underlying the functions of the human PLD enzymes (hPLD) remained unclear based on this structure due to the low homology between these sequences. Here, we describe the first crystal structures of hPLD1 and hPLD2 catalytic domains and identify novel structural elements and functional differences between the prokaryotic and eukaryotic enzymes. Furthermore, structure-based mutation studies and structures of inhibitor-hPLD complexes allowed us to elucidate the binding modes of dual and isoform-selective inhibitors, highlight key determinants of isoenzyme selectivity and provide a basis for further structure-based drug discovery and functional characterization of this therapeutically important superfamily of enzymes.
Collapse
Affiliation(s)
- Claire M Metrick
- Physical Biochemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA.,Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, USA
| | - Emily A Peterson
- Medicinal Chemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Joseph C Santoro
- Bioassays and High Throughput Screens, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Istvan J Enyedy
- Medicinal Chemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Paramasivam Murugan
- Bioassays and High Throughput Screens, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - TeYu Chen
- Medicinal Chemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Klaus Michelsen
- Physical Biochemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Michael Cullivan
- Physical Biochemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Kerri A Spilker
- Physical Biochemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - P Rajesh Kumar
- Physical Biochemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | - Tricia L May-Dracka
- Medicinal Chemistry, Biotherapeutic and Medicinal Sciences, Biogen, Cambridge, MA, USA
| | | |
Collapse
|
38
|
Frias MA, Mukhopadhyay S, Lehman E, Walasek A, Utter M, Menon D, Foster DA. Phosphatidic acid drives mTORC1 lysosomal translocation in the absence of amino acids. J Biol Chem 2020; 295:263-274. [PMID: 31767684 PMCID: PMC6952608 DOI: 10.1074/jbc.ra119.010892] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/14/2019] [Indexed: 01/30/2023] Open
Abstract
Mammalian target of rapamycin complex 1 (mTORC1) promotes cell growth and proliferation in response to nutrients and growth factors. Amino acids induce lysosomal translocation of mTORC1 via the Rag GTPases. Growth factors activate Ras homolog enriched in brain (Rheb), which in turn activates mTORC1 at the lysosome. Amino acids and growth factors also induce the phospholipase D (PLD)-phosphatidic acid (PA) pathway, required for mTORC1 signaling through mechanisms that are not fully understood. Here, using human and murine cell lines, along with immunofluorescence, confocal microscopy, endocytosis, PLD activity, and cell viability assays, we show that exogenously supplied PA vesicles deliver mTORC1 to the lysosome in the absence of amino acids, Rag GTPases, growth factors, and Rheb. Of note, pharmacological or genetic inhibition of endogenous PLD prevented mTORC1 lysosomal translocation. We observed that precancerous cells with constitutive Rheb activation through loss of tuberous sclerosis complex subunit 2 (TSC2) exploit the PLD-PA pathway and thereby sustain mTORC1 activation at the lysosome in the absence of amino acids. Our findings indicate that sequential inputs from amino acids and growth factors trigger PA production required for mTORC1 translocation and activation at the lysosome.
Collapse
Affiliation(s)
- Maria A Frias
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021; Clinical and Translational Master's Program, Clinical and Translational Science Center, Weill Cornell Medicine, New York, New York 10065.
| | - Suman Mukhopadhyay
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021
| | - Elyssa Lehman
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021
| | - Aleksandra Walasek
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021
| | - Matthew Utter
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021
| | - Deepak Menon
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021
| | - David A Foster
- Department of Biological Sciences, Hunter College of the City University of New York, New York, New York 10021; Department of Pharmacology, Weill Cornell Medicine, New York, New York 10065.
| |
Collapse
|
39
|
Moghbeli M. Genetic and Molecular Biology of Multiple Sclerosis Among Iranian Patients: An Overview. Cell Mol Neurobiol 2020; 40:65-85. [PMID: 31482432 DOI: 10.1007/s10571-019-00731-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/24/2019] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is one if the common types of autoimmune disorders in developed countries. Various environmental and genetic factors are associated with initiation and progression of MS. It is believed that the life style changes can be one of the main environmental risk factors. The environmental factors are widely studied and reported, whereas minority of reports have considered the role of genetic factors in biology of MS. Although Iran is a low-risk country in the case of MS prevalence, it has been shown that there was a dramatically rising trend of MS prevalence among Iranian population during recent decades. Therefore, it is required to assess the probable MS risk factors in Iran. In the present study, we summarized all of the reported genes until now which have been associated with MS susceptibility among Iranian patients. To clarify the probable molecular biology of MS progression, we categorized these reported genes based on their cellular functions. This review paves the way of introducing a specific population-based diagnostic panel of genetic markers among the Iranian population for the first time in the world.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
40
|
Abstract
Functions for phospholipase D1 and D2 (PLD1 and PLD2), the canonical isoforms of the PLD superfamily in mammals, have been explored using cell biological and animal disease models for two decades. PLD1 and PLD2, which are activated as a consequence of extracellular signaling events and generate the second messenger signaling lipid phosphatidic acid (PA), have been reported to play roles in settings ranging from platelet activation to the response to cardiac ischemia, viral infection, neurodegenerative disease, and cancer. Of these, the most tractable as therapeutic targets may be thrombotic disease and cancer, as will be discussed here in the context of ongoing efforts to develop small molecule PLD inhibitors.
Collapse
Affiliation(s)
- Christian Salazar
- Center for Developmental Genetics and the Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Michael A Frohman
- Center for Developmental Genetics and the Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, NY, USA.
| |
Collapse
|
41
|
McDermott MI, Wang Y, Wakelam MJO, Bankaitis VA. Mammalian phospholipase D: Function, and therapeutics. Prog Lipid Res 2019; 78:101018. [PMID: 31830503 DOI: 10.1016/j.plipres.2019.101018] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/08/2019] [Accepted: 10/14/2019] [Indexed: 01/23/2023]
Abstract
Despite being discovered over 60 years ago, the precise role of phospholipase D (PLD) is still being elucidated. PLD enzymes catalyze the hydrolysis of the phosphodiester bond of glycerophospholipids producing phosphatidic acid and the free headgroup. PLD family members are found in organisms ranging from viruses, and bacteria to plants, and mammals. They display a range of substrate specificities, are regulated by a diverse range of molecules, and have been implicated in a broad range of cellular processes including receptor signaling, cytoskeletal regulation and membrane trafficking. Recent technological advances including: the development of PLD knockout mice, isoform-specific antibodies, and specific inhibitors are finally permitting a thorough analysis of the in vivo role of mammalian PLDs. These studies are facilitating increased recognition of PLD's role in disease states including cancers and Alzheimer's disease, offering potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- M I McDermott
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America.
| | - Y Wang
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America
| | - M J O Wakelam
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - V A Bankaitis
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America; Department of Chemistry, Texas A&M University, College Station, Texas 77840, United States of America
| |
Collapse
|
42
|
Noble AR, Hogg K, Suman R, Berney DM, Bourgoin S, Maitland NJ, Rumsby MG. Phospholipase D2 in prostate cancer: protein expression changes with Gleason score. Br J Cancer 2019; 121:1016-1026. [PMID: 31673104 PMCID: PMC6964697 DOI: 10.1038/s41416-019-0610-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Phospholipases D1 and D2 (PLD1/2) are implicated in tumorigenesis through their generation of the signalling lipid phosphatidic acid and its downstream effects. Inhibition of PLD1 blocks prostate cell growth and colony formation. Here a role for PLD2 in prostate cancer (PCa), the major cancer of men in the western world, is examined. METHODS PLD2 expression was analysed by immunohistochemistry and western blotting. The effects of PLD2 inhibition on PCa cell viability and cell motility were measured using MTS, colony forming and wound-healing assays. RESULTS PLD2 protein is expressed about equally in luminal and basal prostate epithelial cells. In cells from different Gleason-scored PCa tissue PLD2 protein expression is generally higher than in non-tumorigenic cells and increases in PCa tissue scored Gleason 6-8. PLD2 protein is detected in the cytosol and nucleus and had a punctate appearance. In BPH tissue stromal cells as well as basal and luminal cells express PLD2. PLD2 protein co-expresses with chromogranin A in castrate-resistant PCa tissue. PLD2 inhibition reduces PCa cell viability, colony forming ability and directional cell movement. CONCLUSIONS PLD2 expression correlates with increasing Gleason score to GS8. PLD2 inhibition has the potential to reduce PCa progression.
Collapse
Affiliation(s)
- Amanda R Noble
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK
| | - Karen Hogg
- Technology Facility, Department of Biology, University of York, York, YO10 5DD, UK
| | - Rakesh Suman
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK
| | - Daniel M Berney
- Department of Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Sylvain Bourgoin
- Centre de Recherche du CHU de Québec, Axe des Maladies Infectieuses et Immunitaires, local T1-58, 2705 boulevard Laurier, Québec, G1V 4G2, QC, Canada
| | - Norman J Maitland
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK
| | - Martin G Rumsby
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK.
| |
Collapse
|
43
|
Baek MO, Ahn CB, Cho HJ, Choi JY, Son KH, Yoon MS. Simulated microgravity inhibits C2C12 myogenesis via phospholipase D2-induced Akt/FOXO1 regulation. Sci Rep 2019; 9:14910. [PMID: 31624287 PMCID: PMC6797799 DOI: 10.1038/s41598-019-51410-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/30/2019] [Indexed: 12/19/2022] Open
Abstract
The skeletal muscle system has evolved to maintain body posture against a constant gravitational load. Mammalian target of rapamycin (mTOR) regulates the mechanically induced increase in the skeletal muscle mass. In the present study, we investigated mTOR pathway in C2C12 myoblasts in a model of mechanical unloading by creating a simulated microgravity (SM) using 3 D clinorotation. SM decreased the phosphorylation of Akt at Ser 473, which was mediated by mTOR complex 2 (mTORC2), in C2C12 myoblasts, leading to a decrease in the cell growth rate. Subsequently, SM inhibited C2C12 myogenesis in an Akt-dependent manner. In addition, SM increased the phospholipase D (PLD) activity by enhancing PLD2 expression, resulting in the dissociation of mSIN1 from the mTORC2, followed by decrease in the phosphorylation of Akt at Ser 473, and FOXO1 at Ser 256 in C2C12 myoblasts. Exposure to SM decreased the autophagic flux of C2C12 myoblasts by regulation of mRNA level of autophagic genes in a PLD2 and FOXO1-dependent manner, subsequently, resulting in a decrease in the C2C12 myogenesis. In conclusion, by analyzing the molecular signature of C2C12 myogenesis using SM, we suggest that the regulatory axis of the PLD2 induced Akt/FOXO1, is critical for C2C12 myogenesis.
Collapse
Affiliation(s)
- Mi-Ock Baek
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Chi Bum Ahn
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Hye-Jeong Cho
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Ji-Young Choi
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon, 21565, Republic of Korea.
| | - Mee-Sup Yoon
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea. .,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea. .,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
44
|
Crystal structure of plant PLDα1 reveals catalytic and regulatory mechanisms of eukaryotic phospholipase D. Cell Res 2019; 30:61-69. [PMID: 31619765 DOI: 10.1038/s41422-019-0244-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Phospholipase D (PLD) hydrolyzes the phosphodiester bond of glycerophospholipids and produces phosphatidic acid (PA), which acts as a second messenger in many living organisms. A large number of PLDs have been identified in eukaryotes, and are viewed as promising targets for drug design because these enzymes are known to be tightly regulated and to function in the pathophysiology of many human diseases. However, the underlying molecular mechanisms of catalysis and regulation of eukaryotic PLD remain elusive. Here, we determined the crystal structure of full-length plant PLDα1 in the apo state and in complex with PA. The structure shows that the N-terminal C2 domain hydrophobically interacts with the C-terminal catalytic domain that features two HKD motifs. Our analysis reveals the catalytic site, substrate-binding mechanism, and a new Ca2+-binding site that is required for the activation of PLD. In addition, we tested several efficient small-molecule inhibitors against PLDα1, and suggested a possible competitive inhibition mechanism according to structure-based docking analysis. This study explains many long-standing questions about PLDs and provides structural insights into PLD-targeted inhibitor/drug design.
Collapse
|
45
|
VU0155069 inhibits inflammasome activation independent of phospholipase D1 activity. Sci Rep 2019; 9:14349. [PMID: 31586128 PMCID: PMC6778193 DOI: 10.1038/s41598-019-50806-9] [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: 05/13/2019] [Accepted: 09/19/2019] [Indexed: 01/05/2023] Open
Abstract
The inflammasome is a specialized multiprotein oligomer that regulates IL-1β production. Although regulation of the inflammasome is related to crucial inflammatory disorders such as sepsis, pharmacological inhibitors that effectively inhibit inflammasome activity are limited. Here, we evaluated the effects of a phospholipase D1 (PLD1)-selective inhibitor (VU0155069) against sepsis and inflammasome activation. VU0155069 strongly enhances survival rate in cecal ligation and puncture (CLP)-induced sepsis by inhibiting lung inflammation, leukocyte apoptosis, and the production of proinflammatory cytokines, especially IL-1β. VU0155069 also significantly blocked IL-1β production, caspase-1 activation, and pyroptosis caused by several inflammasome-activating signals in the bone marrow-derived macrophages (BMDMs). However, VU0155069 did not affect LPS-induced activation of signaling molecules such as MAPK, Akt, NF-κB, and NLRP3 expression in the BMDMs. VU0155069 also failed to affect mitochondrial ROS generation and calcium increase caused by nigericin or ATP, and subsequent ASC oligomerization caused by several inflammasome-activating signals. VU0155069 indirectly inhibited caspase-1 activity caused by LPS + nigericin in BMDMs independent of PLD1 activity. We demonstrated that a PLD1 inhibitor, VU0155069, shows anti-septic activity as well as inflammasome-inhibiting effects. Our results suggest that VU0155069 can be considered a novel inflammasome inhibitor.
Collapse
|
46
|
Liang D, Wu K, Tei R, Bumpus TW, Ye J, Baskin JM. A real-time, click chemistry imaging approach reveals stimulus-specific subcellular locations of phospholipase D activity. Proc Natl Acad Sci U S A 2019; 116:15453-15462. [PMID: 31311871 PMCID: PMC6681737 DOI: 10.1073/pnas.1903949116] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The fidelity of signal transduction requires spatiotemporal control of the production of signaling agents. Phosphatidic acid (PA) is a pleiotropic lipid second messenger whose modes of action differ based on upstream stimulus, biosynthetic source, and site of production. How cells regulate the local production of PA to effect diverse signaling outcomes remains elusive. Unlike other second messengers, sites of PA biosynthesis cannot be accurately visualized with subcellular precision. Here, we describe a rapid, chemoenzymatic approach for imaging physiological PA production by phospholipase D (PLD) enzymes. Our method capitalizes on the remarkable discovery that bulky, hydrophilic trans-cyclooctene-containing primary alcohols can supplant water as the nucleophile in the PLD active site in a transphosphatidylation reaction of PLD's lipid substrate, phosphatidylcholine. The resultant trans-cyclooctene-containing lipids are tagged with a fluorogenic tetrazine reagent via a no-rinse, inverse electron-demand Diels-Alder (IEDDA) reaction, enabling their immediate visualization by confocal microscopy in real time. Strikingly, the fluorescent reporter lipids initially produced at the plasma membrane (PM) induced by phorbol ester stimulation of PLD were rapidly internalized via apparent nonvesicular pathways rather than endocytosis, suggesting applications of this activity-based imaging toolset for probing mechanisms of intracellular phospholipid transport. By instead focusing on the initial 10 s of the IEDDA reaction, we precisely pinpointed the subcellular locations of endogenous PLD activity as elicited by physiological agonists of G protein-coupled receptor and receptor tyrosine kinase signaling. These tools hold promise to shed light on both lipid trafficking pathways and physiological and pathological effects of localized PLD signaling.
Collapse
Affiliation(s)
- Dongjun Liang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Kane Wu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Reika Tei
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Timothy W Bumpus
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Johnny Ye
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Jeremy M Baskin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853;
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| |
Collapse
|
47
|
Li WQ, Luo LD, Hu ZW, Lyu TJ, Cen C, Wang Y. PLD1 promotes dendritic spine morphogenesis via activating PKD1. Mol Cell Neurosci 2019; 99:103394. [PMID: 31356881 DOI: 10.1016/j.mcn.2019.103394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 01/20/2023] Open
Abstract
Dendritic spines on the dendrites of pyramidal neurons are one of the most important components for excitatory synapses, where excitatory information exchanges and integrates. The defects of dendritic spine development have been closely connected with many nervous system diseases including autism, intellectual disability and so forth. Based on our previous studies, we here report a new functional signaling link between phospholipase D1 (PLD1) and protein kinase D1 (PKD1) in dendritic spine morphogenesis. Coimmunoprecipitation assays showed that PLD1 associates with PKD1. A series of knocking down and rescuing experiments demonstrated that PLD1 acts upstream of PKD1 in positively regulating dendritic spine morphogenesis. Using PLD1 inhibitor, we found that PLD1 activates PKD1 to promote dendritic spine morphogenesis. Thus, we further reveal the roles of the two different enzymes in neuronal development.
Collapse
Affiliation(s)
- Wen-Qi Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Li-Da Luo
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China.
| | - Zhi-Wen Hu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Tian-Jie Lyu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Cheng Cen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
48
|
Tenconi PE, Bermúdez V, Oresti GM, Giusto NM, Salvador GA, Mateos MV. High glucose-induced phospholipase D activity in retinal pigment epithelium cells: New insights into the molecular mechanisms of diabetic retinopathy. Exp Eye Res 2019; 184:243-257. [PMID: 31059692 DOI: 10.1016/j.exer.2019.04.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/05/2019] [Accepted: 04/30/2019] [Indexed: 02/02/2023]
Abstract
Chronic hyperglycemia, oxidative stress and inflammation are key players in the pathogenesis of diabetic retinopathy (DR). In this work we study the role of phospholipase D (PLD) pathway in an in vitro model of high glucose (HG)-induced damage. To this end, we exposed human retinal pigment epithelium (RPE) cell lines (ARPE-19 and D407) to HG concentrations (16.5 or 33 mM) or to normal glucose concentration (NG, 5.5 mM) for 4, 24 or 72 h. Exposure to HG increased reactive oxygen species levels and caspase-3 cleavage and reduced cell viability after 72 h of incubation. In addition, short term HG exposure (4 h) induced the activation of early events, that involve PLD and ERK1/2 signaling, nuclear factor kappa B (NFκB) nuclear translocation and IκB phosphorylation. The increment in pro-inflammatory interleukins (IL-6 and IL-8) and cyclooxygenase-2 (COX-2) mRNA levels was observed after 24 h of HG exposure. The effect of selective pharmacological PLD1 (VU0359595) and PLD2 (VU0285655-1) inhibitors demonstrated that ERK1/2 and NFκB activation were downstream events of both PLD isoforms. The increment in IL-6 and COX-2 mRNA levels induced by HG was reduced to control levels in cells pre-incubated with both PLD inhibitors. Furthermore, the inhibition of PLD1, PLD2 and MEK/ERK pathway prevented the loss of cell viability and the activation of caspase-3 induced by HG. In conclusion, our findings demonstrate that PLD1 and PLD2 mediate the inflammatory response triggered by HG in RPE cells, pointing to their potential use as a therapeutic target for DR treatment.
Collapse
Affiliation(s)
- Paula E Tenconi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 8000, Bahía, Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), 8000, Bahía, Blanca, Argentina
| | - Vicente Bermúdez
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 8000, Bahía, Blanca, Argentina
| | - Gerardo M Oresti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 8000, Bahía, Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), 8000, Bahía, Blanca, Argentina
| | - Norma M Giusto
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 8000, Bahía, Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), 8000, Bahía, Blanca, Argentina
| | - Gabriela A Salvador
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 8000, Bahía, Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), 8000, Bahía, Blanca, Argentina
| | - Melina V Mateos
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 8000, Bahía, Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), 8000, Bahía, Blanca, Argentina.
| |
Collapse
|
49
|
A phospholipase D2 inhibitor, CAY10594, ameliorates acetaminophen-induced acute liver injury by regulating the phosphorylated-GSK-3β/JNK axis. Sci Rep 2019; 9:7242. [PMID: 31076618 PMCID: PMC6510900 DOI: 10.1038/s41598-019-43673-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
We examined the role of phospholipase D2 (PLD2) on acetaminophen (APAP)-induced acute liver injury using a PLD2 inhibitor (CAY10594). 500 mg/kg of APAP challenge caused acute liver damage. CAY10594 administration markedly blocked the acute liver injury in a dose-dependent manner, showing almost complete inhibition with 8 mg/kg of CAY10594. During the pathological progress of acute liver injury, GSH levels are decreased, and this is significantly recovered upon the administration of CAY10594 at 6 hours post APAP challenge. GSK-3β (Serine 9)/JNK phosphorylation is mainly involved in APAP-induced liver injury. CAY10594 administration strongly blocked GSK-3β (Serine 9)/JNK phosphorylation in the APAP-induced acute liver injury model. Consistently, sustained JNK activation in the cytosol and mitochondria from hepatocytes were also decreased in CAY10594-treated mice. Many types of immune cells are also implicated in APAP-induced liver injury. However, neutrophil and monocyte populations were not different between vehicle- and CAY10594-administered mice which are challenged with APAP. Therapeutic administration of CAY10594 also significantly attenuated liver damage caused by the APAP challenge, eliciting an enhanced survival rate. Taken together, these results indicate that PLD2 is involved in the intrinsic response pathway of hepatocytes driving the pathogenesis of APAP-induced acute liver injury, and PLD2 may therefore represent an important therapeutic target for patients with drug-induced liver injury.
Collapse
|
50
|
Ramenskaia GV, Melnik EV, Petukhov AE. [Phospholipase D: its role in metabolism processes and disease development]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:84-93. [PMID: 29460838 DOI: 10.18097/pbmc20186401084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Phospholipase D (PLD) is one of the key enzymes that catalyzes the hydrolysis of cell membrane phospholipids. In this review current knowledge about six human PLD isoforms, their structure and role in physiological and pathological processes is summarized. Comparative analysis of PLD isoforms structure is presented. The mechanism of the hydrolysis and transphosphatidylation performed by PLD is described. The PLD1 and PLD2 role in the pathogenesis of some cancer, infectious, thrombotic and neurodegenerative diseases is analyzed. The prospects of PLD isoform-selective inhibitors development are shown in the context of the clinical usage and the already-existing inhibitors are characterized. Moreover, the formation of phosphatidylethanol (PEth), the alcohol abuse biomarker, as the result of PLD-catalyzed phospholipid transphosphatidylation is considered.
Collapse
Affiliation(s)
- G V Ramenskaia
- Sechenov First Moscow State Medical University (Sechenovskiy University), Moscow, Russia
| | - E V Melnik
- Sechenov First Moscow State Medical University (Sechenovskiy University), Moscow, Russia
| | - A E Petukhov
- Sechenov First Moscow State Medical University (Sechenovskiy University), Moscow, Russia; Moscow Research and Practical Centre for Narcology, Moscow, Russia
| |
Collapse
|