1
|
Johnson B, Guo Q, Chaludiya K, Kim S. The Proimmunomodulatory and Anti-immunomodulatory Effects of Radiotherapy in Oncologic Care. Hematol Oncol Clin North Am 2025; 39:399-411. [PMID: 39827043 PMCID: PMC11932133 DOI: 10.1016/j.hoc.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The abscopal effect in radiotherapy (RT) refers to the phenomenon where localized radiation treatment causes regression of distant, nonirradiated tumors. Although rare, recent research shows that combining radiation with immunotherapies, such as immune checkpoint inhibitors, can enhance this effect. The interaction between radiation-induced cell death, immune responses, and the tumor microenvironment manifests in competing biologic mechanisms resulting in complex immunologic outcomes. In order to maximize the therapeutic advantages of the immunogenic effect of RT in the future, further studies are needed to fully understand its biologic underpinnings.
Collapse
Affiliation(s)
- Bryan Johnson
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Qianyu Guo
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA; Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Keyur Chaludiya
- Department of Laboratory Medicine, Mayo Clinic Minnesota, 150 3rd Street SW, Rochester, MN 55902, USA
| | - Sungjune Kim
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA.
| |
Collapse
|
2
|
Kurowski K, Prozmann SN, Cabrita Figueiredo AE, Heyer J, Kind F, Schröder KM, Passlick B, Werner M, Bronsert P, Schmid S. The Ectonucleotidases CD39 and CD73 and the Purinergic Receptor P2X4 Serve as Prognostic Markers in Non-Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1142. [PMID: 40227655 PMCID: PMC11987875 DOI: 10.3390/cancers17071142] [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: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Purinergic signaling, which involves extracellular ATP (eATP), its metabolites, purinergic receptors and ectonucleotidases, plays a pivotal role in the tumor microenvironment (TME), impacting tumor progression and the antineoplastic immune response. In this study, the CD39, CD73, P2X4, and P2X7 expression in NSCLC tumor cells and the surrounding stroma of 139 resected patients was examined. METHODS The study included tissue samples from 139 NSCLC patients. Tissue microarrays (TMAs) were constructed using 1.0 mm cores from annotated tumor regions. Immunohistochemical staining for CD39, CD73, P2X4, and P2X4 was performed on 2 µm sections. TMA slides were digitized and analyzed with QuPath, where staining intensity was evaluated using a semi-quantitative H-score. Statistical analysis, including survival analysis, was performed using R, to assess the impact of biomarker expression on patient outcomes. RESULTS High CD39 expression in both tumor and stromal cells was significantly associated with prolonged PFS (respectively: p = 0.0058 and p = 0.0067), particularly in adenocarcinoma (ADC) patients (respectively: p = 0.01 and p = 0.023). In the multivariable Cox model, low CD73 expression in tumor cells correlated with longer PFS (HR: 0.47; 95% CI: [0.28, 0.8], p = 0.005), while low CD73 expression in stromal cells was linked to increased progression risk (HR: 4.81; 95% CI: [1.61, 14.4], p = 0.001). Neither P2X7 nor P2X4 demonstrated a consistent effect on PFS in univariable analyses; however, multivariable analyses suggested that P2X4 might play a prognostic role in NSCLCs (HR: 0.37; 95% CI: [0.19, 0.73], p = 0.003). CONCLUSIONS These findings underscore the importance of purinergic signaling in NSCLC prognosis and highlight the role of the ectonucleotidases CD39 and CD73 as potential therapeutic targets to enhance antineoplastic immune responses.
Collapse
Affiliation(s)
- Konrad Kurowski
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Sophie Nicole Prozmann
- Department of Thoracic Surgery, University Medical Center Freiburg, 79106 Freiburg, Germany
| | | | - Jannis Heyer
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Felix Kind
- Department of Nuclear Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Karl-Moritz Schröder
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Bernward Passlick
- Department of Thoracic Surgery, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Martin Werner
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Peter Bronsert
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Severin Schmid
- Department of Thoracic Surgery, University Medical Center Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
3
|
Chi ZC. Relationship between purinergic P2X7 receptor and colorectal cancer: Research progress and future prospect. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2025; 33:169-177. [DOI: https:/dx.doi.org/10.11569/wcjd.v33.i3.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
|
4
|
Chi ZC. Relationship between purinergic P2X7 receptor and colorectal cancer: Research progress and future prospect. Shijie Huaren Xiaohua Zazhi 2025; 33:169-177. [DOI: 10.11569/wcjd.v33.i3.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
Purinergic P2X7 receptor (P2X7R) is a cellular transmembrane protein. Its activation leads to the release of cytokines, causing the migration and invasion of cancer cells. The expression of P2X7R is associated with tumor inflammation, survival, proliferation, angiogenesis, and metastasis in colorectal cancer (CRC). Evidence suggests that P2X7R expression appears to be epigenetically regulated by DNA methylation and miRNA regulation. With the in-depth study of P2X7R, the application of P2X7R agonists and antagonists has been discussed in the treatment of CRC. This article reviews the relationship between P2X7R and CRC, focusing on the research progress and future prospects of P2X7R in CRC diagnosis and treat-ment.
Collapse
Affiliation(s)
- Zhao-Chun Chi
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266011, Shandong Province, China
| |
Collapse
|
5
|
Pulica R, Aquib A, Varsanyi C, Gadiyar V, Wang Z, Frederick T, Calianese DC, Patel B, de Dios KV, Poalasin V, De Lorenzo MS, Kotenko SV, Wu Y, Yang A, Choudhary A, Sriram G, Birge RB. Dys-regulated phosphatidylserine externalization as a cell intrinsic immune escape mechanism in cancer. Cell Commun Signal 2025; 23:131. [PMID: 40069722 PMCID: PMC11900106 DOI: 10.1186/s12964-025-02090-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
The negatively charged aminophospholipid, phosphatidylserine (PS), is typically restricted to the inner leaflet of the plasma membrane under normal, healthy physiological conditions. PS is irreversibly externalized during apoptosis, where it serves as a signal for elimination by efferocytosis. PS is also reversibly and transiently externalized during cell activation such as platelet and immune cell activation. These events associated with physiological PS externalization are tightly controlled by the regulated activation of flippases and scramblases. Indeed, improper regulation of PS externalization results in thrombotic diseases such as Scott Syndrome, a defect in coagulation and thrombin production, and in the case of efferocytosis, can result in autoimmunity such as systemic lupus erythematosus (SLE) when PS-mediated apoptosis and efferocytosis fails. The physiological regulation of PS is also perturbed in cancer and during viral infection, whereby PS becomes persistently exposed on the surface of such stressed and diseased cells, which can lead to chronic thrombosis and chronic immune evasion. In this review, we summarize evidence for the dysregulation of PS with a main focus on cancer biology and the pathogenic mechanisms for immune evasion and signaling by PS, as well as the discussion of new therapeutic strategies aimed to target externalized PS. We posit that chronic PS externalization is a universal and agnostic marker for diseased tissues, and in cancer, likely reflects a cell intrinsic form of immune escape. The continued development of new therapeutic strategies for targeting PS also provides rationale for their co-utility as adjuvants and with immune checkpoint therapeutics.
Collapse
Affiliation(s)
- Rachael Pulica
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Ahmed Aquib
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Christopher Varsanyi
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Ziren Wang
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Trevor Frederick
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - David C Calianese
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Bhumik Patel
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Kenneth Vergel de Dios
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Victor Poalasin
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Mariana S De Lorenzo
- Department of Cell Biology and Molecular Medicine, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Sergei V Kotenko
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Yi Wu
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Aizen Yang
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Alok Choudhary
- International Center for Public Health, Public Health Research Institute, Newark, NJ, 07103, USA
| | - Ganapathy Sriram
- Department Biological, Chemical and Environmental Sciences, Wheaton College, 26 E Main St, Norton, MA, 02766, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
6
|
Szymczak B, Pegoraro A, De Marchi E, Grignolo M, Maciejewski B, Czarnecka J, Adinolfi E, Roszek K. Retinoic acid-induced alterations enhance eATP-mediated anti-cancer effects in glioma cells: Implications for P2X7 receptor variants as key players. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167611. [PMID: 39626856 DOI: 10.1016/j.bbadis.2024.167611] [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: 03/13/2024] [Revised: 11/08/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024]
Abstract
Retinoic acid (RA) is a small, lipophilic molecule that inhibits cell proliferation and induces differentiation through activation of a family of nuclear receptors (RARs). The therapeutic potential of RA in the treatment of glioma was first evaluated two decades ago, but these attempts were considered not conclusive. Based on the complexity of tumor microenvironment and the role of purinergic signals within TME, we aimed to support RA-induced alterations in glioma cells with extracellular ATP. Our experiments focused on defining the purinergic signaling dynamics of two different human glioma cell lines M059K and M059J subjected to RA-based differentiation protocol. The applied procedure caused considerable modulation in P2X7 receptor variants expression at the gene and protein level, and decrease in ecto-nucleotidase activity. Collectively, it led to the decrease in cell proliferation rate and migration, as well as boosted sensitivity to cytotoxic eATP influence. We confirmed that micromolar concentrations of ATP decreased cell viability by 40 and 20 % in RA-treated M059K and M059J cells, respectively. Moreover, the decrease in migration capability up to 60 % in the presence of 100 μM ATP was observed. Both effects were mediated by P2X7R activation and reversed in the presence of A740003 antagonist, confirming the role of P2X7 receptor. We postulate that retinoic acid-induced changes coupled with micromolar eATP could be effective as anti-cancer treatment affecting the purinergic signaling. The obtained results point out the role of P2X7R variants in influencing potential of glioma cells, as well as the possibility of using these isoforms as therapeutic targets.
Collapse
Affiliation(s)
- Bartosz Szymczak
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Toruń, Poland
| | - Anna Pegoraro
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Elena De Marchi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Marianna Grignolo
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Bartosz Maciejewski
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Toruń, Poland
| | - Joanna Czarnecka
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Toruń, Poland
| | - Elena Adinolfi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Toruń, Poland.
| |
Collapse
|
7
|
McGown A, Renault N, Barczyk A, Nafie J, Barluzzi L, Guest D, Tizzard GJ, Coles SJ, Leach D, von Emloh D, Sutton L, Bailey K, Edmunds L, Greenland BW, Millet R, Spencer J, Dezitter X. Characterization of the Active Enantiomer and Mapping of the Stereospecific Intermolecular Pattern of a Reference P2X7 Allosteric Antagonist. ACS Pharmacol Transl Sci 2025; 8:446-459. [PMID: 39974629 PMCID: PMC11833722 DOI: 10.1021/acsptsci.4c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 02/21/2025]
Abstract
The P2X purinergic receptor 7 (P2X7) has an essential role in inflammation, innate immunity, tumor progression, neurodegenerative diseases, and several other diseases, leading subsequently to the development of P2X7 modulators. AZ11645373 is a frequently studied P2X7 antagonist tool compound but always used as a racemic mixture. Racemic AZ11645373 can be separated into its respective enantiomers by chiral chromatography, albeit in small batches, and these were stereochemically intact over two years later, by chiral high-performance liquid chromatography (HPLC) analysis. On a higher scale, significant decomposition is observed during purification. One of the enantiomers was crystallized as a palladium complex, and its (R)-configuration was determined by single-crystal X-ray diffraction, further confirmed, in solution, by vibrational circular dichroism. Biological studies demonstrated that both (S)- and (R)-forms were able to fully inhibit human P2X7, but (R)-AZ11645373 was more potent, with an IC50 of 32.9 nM. Contrary to its effect on human P2X7, (S)-AZ11645373 was ineffective on mouse P2X7, while the (R)-AZ11645373 enantiomer was a full antagonist. These results demonstrated that the antagonistic effects of racemic AZ11645373 are mainly due to its (R)-enantiomer. Site-directed mutagenesis and molecular dynamics simulations indicated that the (R)-enantiomer may form specific interactions with Phe95 and the antagonists bound to other P2X7 monomers. Phe95 is situated in the allosteric binding site at the edge of the upper vestibule and appears to be the pivotal molecular gateway between AZ11645373 allosteric binding and locking of the closed state of the P2X7 channel. All together, these structure-function relationships should be helpful for drug design of P2X7 modulators.
Collapse
Affiliation(s)
- Andrew McGown
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
- Sussex
Drug Discovery Centre, Department of Chemistry, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Nicolas Renault
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| | - Amélie Barczyk
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| | - Jordan Nafie
- Biotools,
Inc., 17546 Beeline Highway, Jupiter, Florida 33458, United States
| | - Luciano Barluzzi
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Daniel Guest
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Graham J. Tizzard
- National
Crystallography Service, School of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Simon J. Coles
- National
Crystallography Service, School of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - David Leach
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Daniel von Emloh
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Léa Sutton
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Kiera Bailey
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Lewis Edmunds
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Barnaby W. Greenland
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Régis Millet
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| | - John Spencer
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
- Sussex
Drug Discovery Centre, Department of Chemistry, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Xavier Dezitter
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| |
Collapse
|
8
|
Loaiza-Moss J, Braun U, Leitges M. Transcriptome Analysis Suggests PKD3 Regulates Proliferative Glucose Metabolism, Calcium Homeostasis and Microtubule Dynamics After MEF Spontaneous Immortalization. Int J Mol Sci 2025; 26:596. [PMID: 39859313 PMCID: PMC11765705 DOI: 10.3390/ijms26020596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Cell immortalization corresponds to a biologically relevant clinical feature that allows cells to acquire a high proliferative potential during carcinogenesis. In multiple cancer types, Protein Kinase D3 (PKD3) has often been reported as a dysregulated oncogenic kinase that promotes cell proliferation. Using mouse embryonic fibroblasts (MEFs), in a spontaneous immortalization model, PKD3 has been demonstrated as a critical regulator of cell proliferation after immortalization. However, the mechanisms by which PKD3 regulates proliferation in immortalized MEFs require further elucidation. Using a previously validated Prkd3-deficient MEF model, we performed a poly-A transcriptomic analysis to identify putative Prkd3-regulated biological processes and downstream targets in MEFs after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were identified and further analyzed by gene ontology (GO) enrichment and protein-protein interaction (PPI) network analyses to identify potential hub genes. Our results suggest that Prkd3 modulates proliferation through the regulation of gene expression associated with glucose metabolism (Tnf, Ucp2, Pgam2, Angptl4), calcium homeostasis and transport (Calcr and P2rx7) and microtubule dynamics (Stmn2 and Map10). These candidate processes and associated genes represent potential mechanisms involved in Prkd3-induced proliferation in spontaneously immortalized cells as well as clinical targets in several cancer types.
Collapse
Affiliation(s)
| | | | - Michael Leitges
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. Johns, NL A1B 3V6, Canada; (J.L.-M.); (U.B.)
| |
Collapse
|
9
|
Si Q, Yang L, Liu J, Liu H, Bu R, Cui N. Nucleotide receptor P2X7/STAT6 pathway regulates macrophage M2 polarization and its application in CAR-T immunotherapy. Immunobiology 2025; 230:152863. [PMID: 39693800 DOI: 10.1016/j.imbio.2024.152863] [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: 09/17/2024] [Revised: 11/07/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND A key factor underlying the failure of Chimeric Antigen Receptor-T Cell (CAR-T) therapy in ovarian cancer (OC) is the presence of an immunosuppressive tumor microenvironment, which is intricately linked to M2 polarization among tumor-infiltrating macrophages. P2X7 receptor has been previously documented as expressed within these macrophages and its correlation with M2 polarization is evident. This investigation scrutinizes whether silencing of P2X7 receptor within macrophages could lead to augmented anti-tumor potency of CAR-T. METHOD Human peripheral blood mononuclear cells were artificially differentiated into macrophages or M2 macrophage in vitro. After silencing P2X7 receptor and/or overexpressing STAT6 within macrophages, the M1 and M2 markers were evaluated via flow cytometry, ELISA, and qRT-PCR. Additionally, the phosphorylation level of STAT6 was monitored by western blot. We engineered CAR-T cells targeting the non-functional P2X7 (nfP2X7) receptor, and co-cultured them with macrophages silencing P2X7 receptor along with OC cells. The anti-tumor effect of these CAR-T cells was assessed through evaluating OC cell viability, lactate dehydrogenase release, and IFN-γ levels. RESULT P2X7 receptor silencing promotes M1 macrophage marker expression (CD86, TNF-α, IL-6, IL-1β), diminishes M2 macrophage marker expression (CD206 and IL-10) and suppresses STAT6 phosphorylation, whereas STAT6 overexpression reverses these phenomena. Furthermore, M2 macrophage suppresses the toxic effect of CAR-T cells on OC cells, while silencing the P2X7 receptor nullifies the immunosuppressive effect of M2 macrophages on CAR-T cells. CONCLUSION Silencing P2X7 receptor can reverse M2 macrophage polarization by suppressing STAT6 activation, thereby enhancing the anti-tumor efficacy of CAR-T cells targeting nfP2X7 receptor in OC cell lines.
Collapse
Affiliation(s)
- Qin Si
- the Inner Mongolia Key Laboratory for Molecular Regulation of the Cell and the State Key Laboratory of Reproductive Regulation, School of Life Sciences, Inner Mongolia University, Hohhot 010010, Inner Mongolia Autonomous Region, P.R. China
| | - Lu Yang
- Department of Gyneacological Oncology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010010, Inner Mongolia Autonomous Region, P.R. China
| | - Jie Liu
- Department of Ultrasound, International Mongolian Hospital of Inner Mongolia, Hohhot 010020, Inner Mongolia Autonomous Region, P.R. China
| | - Hui Liu
- Department of Gyneacological Oncology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010010, Inner Mongolia Autonomous Region, P.R. China
| | - Ruifang Bu
- Department of Gyneacological Oncology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010010, Inner Mongolia Autonomous Region, P.R. China
| | - Na Cui
- Research Service Office, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical, Hohhot 010010, Inner Mongolia Autonomous Region, P.R. China.
| |
Collapse
|
10
|
Najdaghi S, Davani DN, Fouladseresht H, Ebrahimi N, Sullman MJM, Moradi M, Eskandari N. The Role of Extracellular Vesicles and Microparticles in Central Nervous System Disorders: Mechanisms, Biomarkers, and Therapeutic Potential. Cell Mol Neurobiol 2024; 44:82. [PMID: 39625540 PMCID: PMC11614997 DOI: 10.1007/s10571-024-01518-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
Microscopic, membranous vesicles known as extracellular vesicles (EVs) have been proposed to play a role in the mechanisms underlying central nervous system (CNS) diseases. EVs are secreted by a variety of cells, including myeloid, endothelial, microglial, oligodendroglial, and mesenchymal stem cells (MSCs). Body fluids such as plasma, urine, and cerebrospinal fluid (CSF) contain microparticles (MPs). The detection of MPs in CSF may indicate genetic or environmental susceptibility to conditions such as schizophrenia, schizoaffective disorder, and bipolar disorder. MPs of different origins can exhibit changes in specific biomarkers at various stages of the disease, aiding in the diagnosis and monitoring of neurological conditions. However, understanding the role and clinical applications of MPs is complicated by challenges such as their isolation and dual roles within the CNS. In this review, we discuss the history, characteristics, and roles of MPs in CNS diseases. We also provide practical insights for future research and highlight the challenges that obscure the therapeutic potential of MPs.
Collapse
Affiliation(s)
- Soroush Najdaghi
- Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hamed Fouladseresht
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Narges Ebrahimi
- Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mark J M Sullman
- Department of Social Sciences, School of Humanities and Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, School of Humanities and Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Marjan Moradi
- Departement of Genetics, School of Science, Shahrekord University, Shahrakord, Iran
| | - Nahid Eskandari
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran.
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
11
|
de Carvalho Braga G, Coiado JV, de Melo VC, Loureiro BB, Bagatini MD. Cutaneous melanoma and purinergic modulation by phenolic compounds. Purinergic Signal 2024; 20:581-593. [PMID: 38498100 PMCID: PMC11555167 DOI: 10.1007/s11302-024-10002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Cutaneous melanoma is a complex pathology that still has only treatments that lack efficiency and offer many adverse effects. Due to this scenario emerges the need to analyze other possible treatments against this disease, such as the effect of phenolic compounds. These substances have proven antitumor effects, but still have not been fully explored as a form of therapy to combat melanoma. Also, the purinergic receptors, along with its system molecules, take part in the formation of tumors from many pathways, such as the actions of ectoenzymes and receptors activity, especially P2Rs family, and are formed by structures that can be modulated by the phenolic compounds. Therefore, more studies have to be made with the aim of explaining the purinergic system activity in carcinogenesis of cutaneous melanoma and the effects of its modulation by phenolic compound, in order to enable the development of new therapies to combat this aggressive and feared cancer.
Collapse
Affiliation(s)
| | - João Victor Coiado
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | | | | | | |
Collapse
|
12
|
Tantai X, Yang X, Liu X, Yang X. Antagonism of the ATP-gated P2X7 receptor inhibits the proliferation of hepatocellular carcinoma cells. Purinergic Signal 2024:10.1007/s11302-024-10064-5. [PMID: 39549156 DOI: 10.1007/s11302-024-10064-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
The P2X7 receptor, an ATP-gated ion channel which belongs to the P2X receptor family, plays critical roles in recognizing extracellular adenosine 5'-triphosphate (ATP) and is widely expressed in most tumor cells as well as inflammatory cells. Previously, the P2X7 receptor has been demonstrated to modulate the progression of various malignancies, including glioblastoma, pancreatic cancer, lung cancer, leukemia, and lymphoma. However, the biological function and prognostic values of P2X7 receptor in hepatocellular carcinoma remain to be determined. Here, we investigated the expression level of P2X7 receptor in patients with hepatocellular carcinoma. Then MTS and EdU assays were carried out to study the role of P2X7 receptor blockade in the proliferation of hepatocellular carcinoma cells. In addition, the underlying mechanism was further elucidated by bulk RNAseq. Compared to the control group, the P2X7 receptor was significantly up-regulated in the hepatocellular carcinoma group. Interestingly, A740003 and A438079, two selective antagonists at P2X7 receptor, significantly blocked Ca2+ influx and decreased the proliferative rate of hepatocellular carcinoma cells. Furthermore, the expression level of chondroitin sulfate synthase 1 (CHSY1), an enzyme that mediates the polymerization step of chondroitin sulfate, was reduced by both A740003 and A438079. In conclusion, inhibition of the P2X7 receptor attenuated the proliferation of hepatocellular carcinoma cells, and this process was largely modulated by CHSY1. Thus, our findings reveal a previously unknown role for P2X7 receptor in the proliferation of hepatocellular carcinoma cells and imply that the P2X7 receptor may represent a new target for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xinxing Tantai
- Department of Gastroenterology and Scientific Research Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Xin Yang
- Department of Rheumatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Xinyuan Liu
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511462, Guangdong, China.
| | - Xiao Yang
- Department of Gastroenterology and Scientific Research Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
13
|
Ashmore AA, Balasubramanian B, Phillips A, Asher V, Bali A, Ordóñez-Morán P, Khan R. Bioinformatic and experimental data pertaining to the role of the NLRP3 inflammasome in ovarian cancer. J Cancer Res Clin Oncol 2024; 150:488. [PMID: 39516433 PMCID: PMC11549120 DOI: 10.1007/s00432-024-05988-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
The Nod-Like Receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome plays a role in regulating inflammatory signaling and is a well-established contributor to pyroptotic cell death. It has been investigated extensively in cancer but there remains limited evidence of its role within ovarian cancer (OC). Bioinformatic investigation of gene expression data has highlighted that higher expression of NLRP3 and genes associated with the NLRP3 complex appear to be positively correlated with OC and may also have prognostic significance. However, heterogeneity exists within the results and experimental data is limited and contradictory. If the NLRP3 inflammasome is to be exploited as a therapeutic target, further laboratory-based investigation is required to determine its role in cancer. Furthermore, its relationship with clinically important characteristics such as histopathological subtype may be of key significance in developing targeted therapies towards specific cohorts of patients.
Collapse
Affiliation(s)
- Ayisha A Ashmore
- Derby Gynaecological Cancer Centre, Royal Derby Hospital, University Hospitals of Derby and Burton, Derby, UK.
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK.
| | - Brinda Balasubramanian
- Translational Medical Sciences Unit, Biodiscovery Institute, Centre for Cancer Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Andrew Phillips
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Viren Asher
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Anish Bali
- Derby Gynaecological Cancer Centre, Royal Derby Hospital, University Hospitals of Derby and Burton, Derby, UK
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Paloma Ordóñez-Morán
- Translational Medical Sciences Unit, Biodiscovery Institute, Centre for Cancer Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Raheela Khan
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
14
|
Oken AC, Ditter IA, Lisi NE, Krishnamurthy I, Godsey MH, Mansoor SE. P2X 7 receptors exhibit at least three modes of allosteric antagonism. SCIENCE ADVANCES 2024; 10:eado5084. [PMID: 39365862 PMCID: PMC11451537 DOI: 10.1126/sciadv.ado5084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/28/2024] [Indexed: 10/06/2024]
Abstract
P2X receptors are trimeric ion channels activated by adenosine triphosphate (ATP) that contribute to pathophysiological processes ranging from asthma to neuropathic pain and neurodegeneration. A number of small-molecule antagonists have been identified for these important pharmaceutical targets. However, the molecular pharmacology of P2X receptors is poorly understood because of the chemically disparate nature of antagonists and their differential actions on the seven constituent subtypes. Here, we report high-resolution cryo-electron microscopy structures of the homomeric rat P2X7 receptor bound to five previously known small-molecule allosteric antagonists and a sixth antagonist that we identify. Our structural, biophysical, and electrophysiological data define the molecular determinants of allosteric antagonism in this pharmacologically relevant receptor, revealing three distinct classes of antagonists that we call shallow, deep, and starfish. Starfish binders, exemplified by the previously unidentified antagonist methyl blue, represent a unique class of inhibitors with distinct functional properties that could be exploited to develop potent P2X7 ligands with substantial clinical impact.
Collapse
Affiliation(s)
- Adam C. Oken
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ismayn A. Ditter
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Nicolas E. Lisi
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ipsita Krishnamurthy
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Michael H. Godsey
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Steven E. Mansoor
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
- Division of Cardiovascular Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
15
|
Giorgioni L, Ambrosone A, Cometa MF, Salvati AL, Nisticò R, Magrelli A. Revolutionizing CAR T-Cell Therapies: Innovations in Genetic Engineering and Manufacturing to Enhance Efficacy and Accessibility. Int J Mol Sci 2024; 25:10365. [PMID: 39408696 PMCID: PMC11476879 DOI: 10.3390/ijms251910365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved notable success in treating hematological cancers but faces significant challenges in solid-tumor treatment and overall efficacy. Key limitations include T-cell exhaustion, tumor relapse, immunosuppressive tumor microenvironments (TME), immunogenicity, and antigen heterogeneity. To address these issues, various genetic engineering strategies have been proposed. Approaches such as overexpression of transcription factors or metabolic armoring and dynamic CAR regulation are being explored to improve CAR T-cell function and safety. Other efforts to improve CAR T-cell efficacy in solid tumors include targeting novel antigens or developing alternative strategies to address antigen diversity. Despite the promising preclinical results of these solutions, challenges remain in translating CAR T-cell therapies to the clinic to enable economically viable access to these transformative medicines. The efficiency and scalability of autologous CAR T-cell therapy production are hindered by traditional, manual processes which are costly, time-consuming, and prone to variability and contamination. These high-cost, time-intensive processes have complex quality-control requirements. Recent advancements suggest that smaller, decentralized solutions such as microbioreactors and automated point-of-care systems could improve production efficiency, reduce costs, and shorten manufacturing timelines, especially when coupled with innovative manufacturing methods such as transposons and lipid nanoparticles. Future advancements may include harmonized consumables and AI-enabled technologies, which promise to streamline manufacturing, reduce costs, and enhance production quality.
Collapse
Affiliation(s)
- Lorenzo Giorgioni
- Faculty of Physiology and Pharmacology “V. Erspamer”, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Alessandra Ambrosone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| | - Maria Francesca Cometa
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| | - Anna Laura Salvati
- Faculty of Pharmacy, Tor Vergata University of Rome, 00133 Rome, Italy (R.N.)
| | - Robert Nisticò
- Faculty of Pharmacy, Tor Vergata University of Rome, 00133 Rome, Italy (R.N.)
- Agenzia Italiana del Farmaco, Via del Tritone 181, 00187 Rome, Italy
| | - Armando Magrelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| |
Collapse
|
16
|
Pei S, Piao HL. Exploring Protein S-Palmitoylation: Mechanisms, Detection, and Strategies for Inhibitor Discovery. ACS Chem Biol 2024; 19:1868-1882. [PMID: 39160165 DOI: 10.1021/acschembio.4c00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
S-palmitoylation is a reversible and dynamic process that involves the addition of long-chain fatty acids to proteins. This protein modification regulates various aspects of protein function, including subcellular localization, stability, conformation, and biomolecular interactions. The zinc finger DHHC (ZDHHC) domain-containing protein family is the main group of enzymes responsible for catalyzing protein S-palmitoylation, and 23 members have been identified in mammalian cells. Many proteins that undergo S-palmitoylation have been linked to disease pathogenesis and progression, suggesting that the development of effective inhibitors is a promising therapeutic strategy. Reducing the protein S-palmitoylation level can target either the PATs directly or their substrates. However, there are rare clinically effective S-palmitoylation inhibitors. This review aims to provide an overview of the S-palmitoylation field, including the catalytic mechanism of ZDHHC, S-palmitoylation detection methods, and the functional impact of protein S-palmitoylation. Additionally, this review focuses on current strategies for expanding the chemical toolbox to develop novel and effective inhibitors that can reduce the level of S-palmitoylation of the target protein.
Collapse
Affiliation(s)
- Shaojun Pei
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, 110122 Shenyang, China
| |
Collapse
|
17
|
Logan IE, Nguyen KT, Chatterjee T, Manivannan B, Paul NP, Kim SR, Sixta EM, Bastian LP, Marean-Reardon C, Karajannis MA, Fernández-Valle C, Estevez AG, Franco MC. Selective nitration of Hsp90 acts as a metabolic switch promoting tumor cell proliferation. Redox Biol 2024; 75:103249. [PMID: 38945076 PMCID: PMC11261529 DOI: 10.1016/j.redox.2024.103249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024] Open
Abstract
Tumors develop in an oxidative environment characterized by peroxynitrite production and downstream protein tyrosine (Y) nitration. We showed that tyrosine nitration supports schwannoma cell proliferation and regulates cell metabolism in the inheritable tumor disorder NF2-related Schwannomatosis (NF2-SWN). Here, we identified the chaperone Heat shock protein 90 (Hsp90) as the first nitrated protein that acts as a metabolic switch to promote schwannoma cell proliferation. Doubling the endogenous levels of nitrated Hsp90 in schwannoma cells or supplementing nitrated Hsp90 into normal Schwann cells increased their proliferation. Metabolically, nitration on either Y33 or Y56 conferred Hsp90 distinct functions; nitration at Y33 (Hsp90NY33) down-regulated mitochondrial oxidative phosphorylation, while nitration at Y56 (Hsp90NY56) increased glycolysis by activating the purinergic receptor P2X7 in both schwannoma and normal Schwann cells. Hsp90NY33 and Hsp90NY56 showed differential subcellular and spatial distribution corresponding with their metabolic and proliferative functions in schwannoma three-dimensional cell culture models. Collectively, these results underscore the role of tyrosine nitration as a post-translational modification regulating critical cellular processes. Nitrated proteins, particularly nitrated Hsp90, emerge as a novel category of tumor-directed therapeutic targets.
Collapse
Affiliation(s)
- Isabelle E Logan
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA; Center for Translational Science, Florida International University, Florida, 34987, USA
| | - Kyle T Nguyen
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA
| | - Tilottama Chatterjee
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA
| | | | - Ngozi P Paul
- Center for Translational Science, Florida International University, Florida, 34987, USA
| | - Sharon R Kim
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA
| | - Evelyn M Sixta
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA
| | - Lydia P Bastian
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA
| | - Carrie Marean-Reardon
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA
| | - Matthias A Karajannis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Cristina Fernández-Valle
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Alvaro G Estevez
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Florida, 33199, USA
| | - Maria Clara Franco
- Department of Biochemistry and Biophysics, College of Science, Oregon State University, Corvallis, OR, 97331, USA; Center for Translational Science, Florida International University, Florida, 34987, USA; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Florida, 33199, USA.
| |
Collapse
|
18
|
Oken AC, Lisi NE, Krishnamurthy I, McCarthy AE, Godsey MH, Glasfeld A, Mansoor SE. High-affinity agonism at the P2X 7 receptor is mediated by three residues outside the orthosteric pocket. Nat Commun 2024; 15:6662. [PMID: 39107314 PMCID: PMC11303814 DOI: 10.1038/s41467-024-50771-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
P2X receptors are trimeric ATP-gated ion channels that activate diverse signaling cascades. Due to its role in apoptotic pathways, selective activation of P2X7 is a potential experimental tool and therapeutic approach in cancer biology. However, mechanisms of high-affinity P2X7 activation have not been defined. We report high-resolution cryo-EM structures of wild-type rat P2X7 bound to the high-affinity agonist BzATP as well as significantly improved apo receptor structures in the presence and absence of sodium. Apo structures define molecular details of pore architecture and reveal how a partially hydrated Na+ ion interacts with the conductance pathway in the closed state. Structural, electrophysiological, and direct binding data of BzATP reveal that three residues just outside the orthosteric ATP-binding site are responsible for its high-affinity agonism. This work provides insights into high-affinity agonism for any P2X receptor and lays the groundwork for development of subtype-specific agonists applicable to cancer therapeutics.
Collapse
Affiliation(s)
- Adam C Oken
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Nicolas E Lisi
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Ipsita Krishnamurthy
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Alanna E McCarthy
- Division of Cardiovascular Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Michael H Godsey
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Arthur Glasfeld
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Steven E Mansoor
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA.
- Division of Cardiovascular Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
19
|
Liu X, Li Y, Huang L, Kuang Y, Wu X, Ma X, Zhao B, Lan J. Unlocking the therapeutic potential of P2X7 receptor: a comprehensive review of its role in neurodegenerative disorders. Front Pharmacol 2024; 15:1450704. [PMID: 39139642 PMCID: PMC11319138 DOI: 10.3389/fphar.2024.1450704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The P2X7 receptor (P2X7R), an ATP-gated ion channel, has emerged as a crucial player in neuroinflammation and a promising therapeutic target for neurodegenerative disorders. This review explores the current understanding of P2X7R's structure, activation, and physiological roles, focusing on its expression and function in microglial cells. The article examines the receptor's involvement in calcium signaling, microglial activation, and polarization, as well as its role in the pathogenesis of Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis. The review highlights the complex nature of P2X7R signaling, discussing its potential neuroprotective and neurotoxic effects depending on the disease stage and context. It also addresses the development of P2X7R antagonists and their progress in clinical trials, identifying key research gaps and future perspectives for P2X7R-targeted therapy development. By providing a comprehensive overview of the current state of knowledge and future directions, this review serves as a valuable resource for researchers and clinicians interested in exploring the therapeutic potential of targeting P2X7R for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Xiaoming Liu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yiwen Li
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Liting Huang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yingyan Kuang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiaoxiong Wu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiangqiong Ma
- Henan Hospital of Integrated Chinese and Western Medicine, Zhengzhou, China
| | - Beibei Zhao
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jiao Lan
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
20
|
Du Y, Cao Y, Song W, Wang X, Yu Q, Peng X, Zhao R. Role of the P2X7 receptor in breast cancer progression. Purinergic Signal 2024:10.1007/s11302-024-10039-6. [PMID: 39039304 DOI: 10.1007/s11302-024-10039-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
Breast cancer is a common malignant tumor, whose incidence is increasing year by year, and it has become the malignant tumor with the highest incidence rate in women. Purine ligand-gated ion channel 7 receptor (P2X7R) is a cation channel receptor with Adenosine triphosphate ( ATP) as a ligand, which is widely distributed in cells and tissues, and is closely related to tumorigenesis and progression. P2X7R plays an important role in cancer by interacting with ATP. Studies have shown that P2X7R is up-regulated in breast cancer and can promote tumor invasion and metastasis by activating the protein kinase B (AKT) signaling pathway, promoting epithelial-mesenchymal transition (EMT), controlling the generation of extracellular vesicle (EV), and regulating the expression of the inflammatory protein cyclooxygenase 2 (COX-2). Furthermore, P2X7R was proven to play an essential role in the proliferation and apoptosis of breast cancer cells. Recently, inhibitors targeting P2X7R have been found to inhibit the progression of breast cancer. Natural P2X7R antagonists, such as rhodopsin, and the isoquinoline alkaloid berberine, have also been shown to be effective in inhibiting breast cancer progression. In this article, we review the research progress of P2X7R and breast cancer intending to provide new targets and directions for breast cancer treatment.
Collapse
Affiliation(s)
- Yanan Du
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Yahui Cao
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Wei Song
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Xin Wang
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Qingqing Yu
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Xiaoxiang Peng
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China.
| | - Ronglan Zhao
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China.
| |
Collapse
|
21
|
Stauffer PE, Brinkley J, Jacobson DA, Quaranta V, Tyson DR. Purinergic Ca 2+ Signaling as a Novel Mechanism of Drug Tolerance in BRAF-Mutant Melanoma. Cancers (Basel) 2024; 16:2426. [PMID: 39001489 PMCID: PMC11240618 DOI: 10.3390/cancers16132426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Drug tolerance is a major cause of relapse after cancer treatment. Despite intensive efforts, its molecular basis remains poorly understood, hampering actionable intervention. We report a previously unrecognized signaling mechanism supporting drug tolerance in BRAF-mutant melanoma treated with BRAF inhibitors that could be of general relevance to other cancers. Its key features are cell-intrinsic intracellular Ca2+ signaling initiated by P2X7 receptors (purinergic ligand-gated cation channels) and an enhanced ability for these Ca2+ signals to reactivate ERK1/2 in the drug-tolerant state. Extracellular ATP, virtually ubiquitous in living systems, is the ligand that can initiate Ca2+ spikes via P2X7 channels. ATP is abundant in the tumor microenvironment and is released by dying cells, ironically implicating treatment-initiated cancer cell death as a source of trophic stimuli that leads to ERK reactivation and drug tolerance. Such a mechanism immediately offers an explanation of the inevitable relapse after BRAFi treatment in BRAF-mutant melanoma and points to actionable strategies to overcome it.
Collapse
Affiliation(s)
- Philip E. Stauffer
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jordon Brinkley
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Vito Quaranta
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Darren R. Tyson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
22
|
Pegoraro A, Grignolo M, Ruo L, Ricci L, Adinolfi E. P2X7 Variants in Pathophysiology. Int J Mol Sci 2024; 25:6673. [PMID: 38928378 PMCID: PMC11204217 DOI: 10.3390/ijms25126673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
P2X7 receptor activation by extracellular adenosine triphosphate (eATP) modulates different intracellular pathways, including pro-inflammatory and tumor-promoting cascades. ATP is released by cells and necrotic tissues during stressful conditions and accumulates mainly in the inflammatory and tumoral microenvironments. As a consequence, both the P2X7 blockade and agonism have been proposed as therapeutic strategies in phlogosis and cancer. Nevertheless, most studies have been carried out on the WT fully functional receptor variant. In recent years, the discovery of P2X7 variants derived by alternative splicing mechanisms or single-nucleotide substitutions gave rise to the investigation of these new P2X7 variants' roles in different processes and diseases. Here, we provide an overview of the literature covering the function of human P2X7 splice variants and polymorphisms in diverse pathophysiological contexts, paying particular attention to their role in oncological and neuroinflammatory conditions.
Collapse
Affiliation(s)
- Anna Pegoraro
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.G.); (L.R.); (L.R.)
| | | | | | | | - Elena Adinolfi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.G.); (L.R.); (L.R.)
| |
Collapse
|
23
|
Lindo J, Nogueira C, Soares R, Cunha N, Almeida MR, Rodrigues L, Coelho P, Rodrigues F, Cunha RA, Gonçalves T. Genetic Polymorphisms of P2RX7 but Not of ADORA2A Are Associated with the Severity of SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:6135. [PMID: 38892324 PMCID: PMC11173306 DOI: 10.3390/ijms25116135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
SARS-CoV-2 infection ranges from mild to severe presentations, according to the intensity of the aberrant inflammatory response. Purinergic receptors dually control the inflammatory response: while adenosine A2A receptors (A2ARs) are anti-inflammatory, ATP P2X7 receptors (P2X7Rs) exert pro-inflammatory effects. The aim of this study was to assess if there were differences in allelic and genotypic frequencies of a loss-of-function SNP of ADORA2A (rs2298383) and a gain-of-function single nucleotide polymorphism (SNP) of P2RX7 (rs208294) in the severity of SARS-CoV-2-associated infection. Fifty-five individuals were enrolled and categorized according to the severity of the infection. Endpoint genotyping was performed in blood cells to screen for both SNPs. The TT genotype (vs. CT + CC) and the T allele (vs. C allele) of P2RX7 SNP were found to be associated with more severe forms of COVID-19, whereas the association between ADORA2A SNP and the severity of infection was not significantly different. The T allele of P2RX7 SNP was more frequent in people with more than one comorbidity and with cardiovascular conditions and was associated with colorectal cancer. Our findings suggest a more prominent role of P2X7R rather than of A2AR polymorphisms in SARS-CoV-2 infection, although larger population-based studies should be performed to validate our conclusions.
Collapse
Affiliation(s)
- Jorge Lindo
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Célia Nogueira
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Rui Soares
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
- Clinical Pathology Service, Portuguese Oncology Institute of Coimbra, 3004-011 Coimbra, Portugal;
| | - Nuno Cunha
- Clinical Pathology Service, Portuguese Oncology Institute of Coimbra, 3004-011 Coimbra, Portugal;
| | - Maria Rosário Almeida
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Lisa Rodrigues
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Patrícia Coelho
- IPCB/ESALD—Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, SPRINT-IPCB—Sport Physical Activity and Health Research & Innovation Center, 6000-767 Castelo Branco, Portugal; (P.C.); (F.R.)
| | - Francisco Rodrigues
- IPCB/ESALD—Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, SPRINT-IPCB—Sport Physical Activity and Health Research & Innovation Center, 6000-767 Castelo Branco, Portugal; (P.C.); (F.R.)
| | - Rodrigo A. Cunha
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Gonçalves
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
24
|
Bandara V, Niktaras VM, Willett VJ, Chapman H, Lokman NA, Macpherson AM, Napoli S, Gundsambuu B, Foeng J, Sadlon TJ, Coombs J, McColl SR, Barry SC, Oehler MK, Ricciardelli C. Engineered CAR-T cells targeting the non-functional P2X purinoceptor 7 (P2X7) receptor as a novel treatment for ovarian cancer. Clin Transl Immunology 2024; 13:e1512. [PMID: 38800555 PMCID: PMC11116765 DOI: 10.1002/cti2.1512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/01/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives Recent studies have identified expression of the non-functional P2X7 (nfP2X7) receptor on various malignant cells including ovarian cancer, but not on normal cells, which makes it a promising tumour-associated antigen candidate for chimeric antigen receptor (CAR)-T-cell immunotherapies. In this study, we assessed the cytotoxic effects of nfP2X7-CAR-T cells on ovarian cancer using in vitro and in vivo models. Methods We evaluated the effects of nfP2X7-CAR-T cells on ovarian cancer cell lines (SKOV-3, OVCAR3, OVCAR5), normal peritoneal cells (LP-9) and primary serous ovarian cancer cells derived from patient ascites in vitro using monolayer and 3D spheroid assays. We also evaluated the effects of nfP2X7-CAR-T cells on patient-derived tissue explants, which recapitulate an intact tumour microenvironment. In addition, we investigated the effect of nfP2X7-CAR-T cells in vivo using the OVCAR-3 xenograft model in NOD-scid IL2Rγnull (NSG) mice. Results Our study found that nfP2X7-CAR-T cells were cytotoxic and significantly inhibited survival of OVCAR3, OVCAR5 and primary serous ovarian cancer cells compared with un-transduced CD3+ T cells in vitro. However, no significant effects of nfP2X7-CAR-T cells were observed for SKOV3 or normal peritoneal cells (LP-9) cells with low P2X7 receptor expression. Treatment with nfP2X7-CAR-T cells increased apoptosis compared with un-transduced T cells in patient-derived explants and correlated with CD3 positivity. Treatment with nfP2X7-CAR-T cells significantly reduced OVCAR3 tumour burden in mice compared with un-transduced CD3 cells for 7-8 weeks. Conclusion This study demonstrates that nfP2X7-CAR-T cells have great potential to be developed as a novel immunotherapy for ovarian cancer.
Collapse
Affiliation(s)
- Veronika Bandara
- Molecular Immunology, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Victoria M Niktaras
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Vasiliki J Willett
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Hayley Chapman
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Noor A Lokman
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Anne M Macpherson
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Silvana Napoli
- Molecular Immunology, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Batjargal Gundsambuu
- Molecular Immunology, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Jade Foeng
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological SciencesThe University of AdelaideAdelaideSAAustralia
| | - Timothy J Sadlon
- Molecular Immunology, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Justin Coombs
- Carina Biotech, Level 2 Innovation & Collaboration CentreAdelaideSAAustralia
| | - Shaun R McColl
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological SciencesThe University of AdelaideAdelaideSAAustralia
- Carina Biotech, Level 2 Innovation & Collaboration CentreAdelaideSAAustralia
| | - Simon C Barry
- Molecular Immunology, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| | - Martin K Oehler
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
- Department of Gynaecological OncologyRoyal Adelaide HospitalAdelaideSAAustralia
| | - Carmela Ricciardelli
- Reproductive Cancer Research Group, Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research InstituteUniversity of AdelaideAdelaideSAAustralia
| |
Collapse
|
25
|
Ferreira NCDS, Viviani LG, Lima LM, do Amaral AT, Romano JVP, Fortunato AL, Soares RF, Alberto AVP, Coelho Neto JA, Alves LA. A Hybrid Approach Combining Shape-Based and Docking Methods to Identify Novel Potential P2X7 Antagonists from Natural Product Databases. Pharmaceuticals (Basel) 2024; 17:592. [PMID: 38794162 PMCID: PMC11123696 DOI: 10.3390/ph17050592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 05/26/2024] Open
Abstract
P2X7 is an ATP-activated purinergic receptor implicated in pro-inflammatory responses. It is associated with the development of several diseases, including inflammatory and neurodegenerative conditions. Although several P2X7 receptor antagonists have recently been reported in the literature, none of them is approved for clinical use. However, the structure of the known antagonists can serve as a scaffold for discovering effective compounds in clinical therapy. This study aimed to propose an improved virtual screening methodology for the identification of novel potential P2X7 receptor antagonists from natural products through the combination of shape-based and docking approaches. First, a shape-based screening was performed based on the structure of JNJ-47965567, a P2X7 antagonist, using two natural product compound databases, MEGx (~5.8 × 103 compounds) and NATx (~32 × 103 compounds). Then, the compounds selected by the proposed shape-based model, with Shape-Tanimoto score values ranging between 0.624 and 0.799, were filtered for drug-like properties. Finally, the compounds that met the drug-like filter criteria were docked into the P2X7 allosteric binding site, using the docking programs GOLD and DockThor. The docking poses with the best score values were submitted to careful visual inspection of the P2X7 allosteric binding site. Based on our established visual inspection criteria, four compounds from the MEGx database and four from the NATx database were finally selected as potential P2X7 receptor antagonists. The selected compounds are structurally different from known P2X7 antagonists, have drug-like properties, and are predicted to interact with key P2X7 allosteric binding pocket residues, including F88, F92, F95, F103, M105, F108, Y295, Y298, and I310. Therefore, the combination of shape-based screening and docking approaches proposed in our study has proven useful in selecting potential novel P2X7 antagonist candidates from natural-product-derived compounds databases. This approach could also be useful for selecting potential inhibitors/antagonists of other receptors and/or biological targets.
Collapse
Affiliation(s)
- Natiele Carla da Silva Ferreira
- Laboratory of Cellular Communication, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; (N.C.d.S.F.); (L.M.L.); (J.V.P.R.); (A.L.F.); (A.V.P.A.)
| | - Lucas Gasparello Viviani
- Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil; (L.G.V.); (A.T.d.A.)
| | - Lauro Miranda Lima
- Laboratory of Cellular Communication, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; (N.C.d.S.F.); (L.M.L.); (J.V.P.R.); (A.L.F.); (A.V.P.A.)
| | | | - João Victor Paiva Romano
- Laboratory of Cellular Communication, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; (N.C.d.S.F.); (L.M.L.); (J.V.P.R.); (A.L.F.); (A.V.P.A.)
- Laboratory of Immunobiotechnology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Anderson Lage Fortunato
- Laboratory of Cellular Communication, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; (N.C.d.S.F.); (L.M.L.); (J.V.P.R.); (A.L.F.); (A.V.P.A.)
| | - Rafael Ferreira Soares
- Laboratory of Applied Genomics and Bioinnovations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil;
| | - Anael Viana Pinto Alberto
- Laboratory of Cellular Communication, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; (N.C.d.S.F.); (L.M.L.); (J.V.P.R.); (A.L.F.); (A.V.P.A.)
| | - Jose Aguiar Coelho Neto
- National Institute of Industrial Property, Rio de Janeiro 20090-910, Brazil;
- Tijuca Campus, Veiga de Almeida University, Rio de Janeiro 20271-020, Brazil
| | - Luiz Anastacio Alves
- Laboratory of Cellular Communication, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; (N.C.d.S.F.); (L.M.L.); (J.V.P.R.); (A.L.F.); (A.V.P.A.)
| |
Collapse
|
26
|
Notarangelo MP, Penolazzi L, Lambertini E, Falzoni S, De Bonis P, Capanni C, Di Virgilio F, Piva R. The NFATc1/P2X7 receptor relationship in human intervertebral disc cells. Front Cell Dev Biol 2024; 12:1368318. [PMID: 38638530 PMCID: PMC11024252 DOI: 10.3389/fcell.2024.1368318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 04/20/2024] Open
Abstract
A comprehensive understanding of the molecules that play key roles in the physiological and pathological homeostasis of the human intervertebral disc (IVD) remains challenging, as does the development of new therapeutic treatments. We recently found a positive correlation between IVD degeneration (IDD) and P2X7 receptor (P2X7R) expression increases both in the cytoplasm and in the nucleus. Using immunocytochemistry, reverse transcription PCR (RT-PCR), overexpression, and chromatin immunoprecipitation, we found that NFATc1 and hypoxia-inducible factor-1α (HIF-1α) are critical regulators of P2X7R. Both transcription factors are recruited at the promoter of the P2RX7 gene and involved in its positive and negative regulation, respectively. Furthermore, using the proximity ligation assay, we revealed that P2X7R and NFATc1 form a molecular complex and that P2X7R is closely associated with lamin A/C, a major component of the nuclear lamina. Collectively, our study identifies, for the first time, P2X7R and NFATc1 as markers of IVD degeneration and demonstrates that both NFATc1 and lamin A/C are interaction partners of P2X7R.
Collapse
Affiliation(s)
| | - Letizia Penolazzi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Elisabetta Lambertini
- Department of Chemical, Pharmaceutical and Agricultural Sciences of the University of Ferrara, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Pasquale De Bonis
- Neurosurgery Department, Sant’Anna University Hospital, Ferrara, Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | | | - Roberta Piva
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| |
Collapse
|
27
|
Stauffer PE, Brinkley J, Jacobson D, Quaranta V, Tyson DR. Purinergic Ca 2+ signaling as a novel mechanism of drug tolerance in BRAF mutant melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565532. [PMID: 37961267 PMCID: PMC10635130 DOI: 10.1101/2023.11.03.565532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Drug tolerance is a major cause of relapse after cancer treatment. In spite of intensive efforts1-9, its molecular basis remains poorly understood, hampering actionable intervention. We report a previously unrecognized signaling mechanism supporting drug tolerance in BRAF-mutant melanoma treated with BRAF inhibitors that could be of general relevance to other cancers. Its key features are cell-intrinsic intracellular Ca2+ signaling initiated by P2X7 receptors (purinergic ligand-gated cation channels), and an enhanced ability for these Ca2+ signals to reactivate ERK1/2 in the drug-tolerant state. Extracellular ATP, virtually ubiquitous in living systems, is the ligand that can initiate Ca2+ spikes via P2X7 channels. ATP is abundant in the tumor microenvironment and is released by dying cells, ironically implicating treatment-initiated cancer cell death as a source of trophic stimuli that leads to ERK reactivation and drug tolerance. Such a mechanism immediately offers an explanation of the inevitable relapse after BRAFi treatment in BRAF-mutant melanoma, and points to actionable strategies to overcome it.
Collapse
Affiliation(s)
- Philip E Stauffer
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Jordon Brinkley
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - David Jacobson
- Departments of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Vito Quaranta
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Darren R Tyson
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| |
Collapse
|
28
|
Charras A, Hofmann SR, Cox A, Schulze F, Russ S, Northey S, Liu X, Fang Y, Haldenby S, Hartmann H, Bassuk AG, Carvalho A, Sposito F, Grinstein L, Rösen-Wolff A, Meyer-Bahlburg A, Beresford MW, Lainka E, Foell D, Wittkowski H, Girschick HJ, Morbach H, Uebe S, Hüffmeier U, Ferguson PJ, Hedrich CM. P2RX7 gene variants associate with altered inflammasome assembly and reduced pyroptosis in chronic nonbacterial osteomyelitis (CNO). J Autoimmun 2024; 144:103183. [PMID: 38401466 DOI: 10.1016/j.jaut.2024.103183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024]
Abstract
Chronic nonbacterial osteomyelitis (CNO), an autoinflammatory bone disease primarily affecting children, can cause pain, hyperostosis and fractures, affecting quality-of-life and psychomotor development. This study investigated CNO-associated variants in P2RX7, encoding for the ATP-dependent trans-membrane K+ channel P2X7, and their effects on NLRP3 inflammasome assembly. Whole exome sequencing in two related transgenerational CNO patients, and target sequencing of P2RX7 in a large CNO cohort (N = 190) were conducted. Results were compared with publicly available datasets and regional controls (N = 1873). Findings were integrated with demographic and clinical data. Patient-derived monocytes and genetically modified THP-1 cells were used to investigate potassium flux, inflammasome assembly, pyroptosis, and cytokine release. Rare presumably damaging P2RX7 variants were identified in two related CNO patients. Targeted P2RX7 sequencing identified 62 CNO patients with rare variants (32.4%), 11 of which (5.8%) carried presumably damaging variants (MAF <1%, SIFT "deleterious", Polyphen "probably damaging", CADD >20). This compared to 83 of 1873 controls (4.4%), 36 with rare and presumably damaging variants (1.9%). Across the CNO cohort, rare variants unique to one (Median: 42 versus 3.7) or more (≤11 patients) participants were over-represented when compared to 190 randomly selected controls. Patients with rare damaging variants more frequently experienced gastrointestinal symptoms and lymphadenopathy while having less spinal, joint and skin involvement (psoriasis). Monocyte-derived macrophages from patients, and genetically modified THP-1-derived macrophages reconstituted with CNO-associated P2RX7 variants exhibited altered potassium flux, inflammasome assembly, IL-1β and IL-18 release, and pyroptosis. Damaging P2RX7 variants occur in a small subset of CNO patients, and rare P2RX7 variants may represent a CNO risk factor. Observations argue for inflammasome inhibition and/or cytokine blockade and may allow future patient stratification and individualized care.
Collapse
Affiliation(s)
- Amandine Charras
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Sigrun R Hofmann
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Allison Cox
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, USA
| | - Felix Schulze
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Susanne Russ
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Sarah Northey
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Xuan Liu
- Centre of Genome Research, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, UK
| | - Yongxiang Fang
- Centre of Genome Research, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, UK
| | - Sam Haldenby
- Centre of Genome Research, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, UK
| | - Hella Hartmann
- Light Microscopy Facility, Centre for Regenerative Therapies, Technische Universität Dresden, Germany
| | - Alexander G Bassuk
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, USA
| | - Ana Carvalho
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Francesca Sposito
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Lev Grinstein
- Department of Pediatrics, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Angela Rösen-Wolff
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology and Immunology, Department of Pediatrics, University Medicine Greifswald, Greifswald, Germany
| | - Michael W Beresford
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, UK; Department of Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Elke Lainka
- Department of Pediatrics II, University Hospital Essen, University of Duisburg-Essen, Essen, Germany on behalf of the German Autoinflammatory Disease Network (AID Net), Germany
| | - Dirk Foell
- Department for Pediatric Rheumatology & Immunology, University Hospital Münster, Germany on behalf of the German Autoinflammatory Disease Network (AID Net), Germany
| | - Helmut Wittkowski
- Department for Pediatric Rheumatology & Immunology, University Hospital Münster, Germany on behalf of the German Autoinflammatory Disease Network (AID Net), Germany
| | | | - Henner Morbach
- Department of Pediatrics, University Hospital Würzburg, Germany
| | - Steffen Uebe
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrike Hüffmeier
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Polly J Ferguson
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, USA
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, UK; Department of Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
29
|
Wang H, Wei Y, Wang N. Purinergic pathways and their clinical use in the treatment of acute myeloid leukemia. Purinergic Signal 2024:10.1007/s11302-024-09997-8. [PMID: 38446337 DOI: 10.1007/s11302-024-09997-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Despite the use of various therapies such as hematopoietic stem cell transplantation and chimeric antigen receptor T cell therapy (CAR-T), the prognosis of patients with acute myeloid leukemia (AML) is still generally poor. However, immunotherapy is currently a hot topic in the treatment of hematological tumors. Extracellular adenosine triphosphate (ATP) can be converted to adenosine diphosphate (ADP) via CD39, and ADP can be converted to adenosine via CD73, which can bind to P1 and P2 receptors to exert immunomodulatory effects. Research on the mechanism of the purinergic signaling pathway can provide a new direction for the treatment of AML, and inhibitors of this signaling pathway have been discovered by several researchers and gradually applied in the clinic. In this paper, the mechanism of the purinergic signaling pathway and its clinical application are described, revealing a new target for the treatment of AML and subsequent improvement in patient prognosis.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yujie Wei
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Na Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
30
|
Acuña-Castillo C, Escobar A, García-Gómez M, Bachelet VC, Huidobro-Toro JP, Sauma D, Barrera-Avalos C. P2X7 Receptor in Dendritic Cells and Macrophages: Implications in Antigen Presentation and T Lymphocyte Activation. Int J Mol Sci 2024; 25:2495. [PMID: 38473744 DOI: 10.3390/ijms25052495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
The P2X7 receptor, a member of the P2X purinergic receptor family, is a non-selective ion channel. Over the years, it has been associated with various biological functions, from modulating to regulating inflammation. However, its emerging role in antigen presentation has captured the scientific community's attention. This function is essential for the immune system to identify and respond to external threats, such as pathogens and tumor cells, through T lymphocytes. New studies show that the P2X7 receptor is crucial for controlling how antigens are presented and how T cells are activated. These studies focus on antigen-presenting cells, like dendritic cells and macrophages. This review examines how the P2X7 receptor interferes with effective antigen presentation and activates T cells and discusses the fundamental mechanisms that can affect the immune response. Understanding these P2X7-mediated processes in great detail opens up exciting opportunities to create new immunological therapies.
Collapse
Affiliation(s)
- Claudio Acuña-Castillo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Alejandro Escobar
- Laboratorio Biología Celular y Molecular, Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380000, Chile
| | - Moira García-Gómez
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| | - Vivienne C Bachelet
- Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Juan Pablo Huidobro-Toro
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
- Centro Ciencia & Vida, Av. Del Valle Norte 725, Huechuraba 8580000, Chile
| | - Carlos Barrera-Avalos
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| |
Collapse
|
31
|
Zheng H, Liu Q, Zhou S, Luo H, Zhang W. Role and therapeutic targets of P2X7 receptors in neurodegenerative diseases. Front Immunol 2024; 15:1345625. [PMID: 38370420 PMCID: PMC10869479 DOI: 10.3389/fimmu.2024.1345625] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
The P2X7 receptor (P2X7R), a non-selective cation channel modulated by adenosine triphosphate (ATP), localizes to microglia, astrocytes, oligodendrocytes, and neurons in the central nervous system, with the most incredible abundance in microglia. P2X7R partake in various signaling pathways, engaging in the immune response, the release of neurotransmitters, oxidative stress, cell division, and programmed cell death. When neurodegenerative diseases result in neuronal apoptosis and necrosis, ATP activates the P2X7R. This activation induces the release of biologically active molecules such as pro-inflammatory cytokines, chemokines, proteases, reactive oxygen species, and excitotoxic glutamate/ATP. Subsequently, this leads to neuroinflammation, which exacerbates neuronal involvement. The P2X7R is essential in the development of neurodegenerative diseases. This implies that it has potential as a drug target and could be treated using P2X7R antagonists that are able to cross the blood-brain barrier. This review will comprehensively and objectively discuss recent research breakthroughs on P2X7R genes, their structural features, functional properties, signaling pathways, and their roles in neurodegenerative diseases and possible therapies.
Collapse
Affiliation(s)
- Huiyong Zheng
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiang Liu
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Siwei Zhou
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wenjun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
32
|
Zhang GP, Liao JX, Liu YY, Zhu FQ, Huang HJ, Zhang WJ. Ion channel P2X7 receptor in the progression of cancer. Front Oncol 2024; 13:1297775. [PMID: 38273855 PMCID: PMC10808724 DOI: 10.3389/fonc.2023.1297775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
P2X7 receptor (P2X7) is a non-selective and ATP-sensitive ligand-gated cation channel. Studies have confirmed that it is expressed in a variety of cells and correlates with their function, frequently in immune cells and tumor cells. We found increased expression of this receptor in many tumor cells, and it has a role in tumor survival and progression. In immune cells, upregulation of the receptor has a double effect on tumor suppression as well as tumor promotion. This review describes the structure of P2X7 and its role in the tumor microenvironment and presents possible mechanisms of P2X7 in tumor invasion and metastasis. Understanding the potential of P2X7 for tumor treatment, we also present several therapeutic agents targeting P2X7 and their mechanisms of action. In conclusion, the study of P2X7 is an important guideline for the use of clinical tumor therapy and may be able to provide a new idea for tumor treatment, but considering the complexity of the biological effects of P2X7, the drugs should be used with caution in clinical practice.
Collapse
Affiliation(s)
- Guang-ping Zhang
- The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Department of Critical Medicine, Ganzhou people’s Hospital, Ganzhou, Jiangxi, China
| | - Jun-xiang Liao
- The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Yi-yi Liu
- The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Fu-qi Zhu
- The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Hui-jin Huang
- The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Wen-jun Zhang
- Department of Rehabilitation Medicine, the Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
33
|
Zhang J, Gao L, Zhang Y, Wang H, Sun S, Wu L. Involvement of microglial P2X7 receptor in pain modulation. CNS Neurosci Ther 2024; 30:e14496. [PMID: 37950524 PMCID: PMC10805404 DOI: 10.1111/cns.14496] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/23/2023] [Accepted: 10/02/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Pain is a rapid response mechanism that compels organisms to retreat from the harmful stimuli and triggers a repair response. Nonetheless, when pain persists for extended periods, it can lead to adverse changes into in the individual's brain, negatively impacting their emotional state and overall quality of life. Microglia, the resident immune cells in the central nervous system (CNS), play a pivotal role in regulating a variety of pain-related disorders. Specifically, recent studies have shed light on the central role that microglial purinergic ligand-gated ion channel 7 receptor (P2X7R) plays in regulating pain. In this respect, the P2X7R on microglial membranes represents a potential therapeutic target. AIMS To expound on the intricate link between microglial P2X7R and pain, offering insights into potential avenues for future research. METHODS We reviewed 140 literature and summarized the important role of microglial P2X7R in regulating pain, including the structure and function of P2X7R, the relationship between P2X7R and microglial polarization, P2X7R-related signaling pathways, and the effects of P2X7R antagonists on pain regulation. RESULTS P2X7R activation is related to M1 polarization of microglia, while suppressing P2X7R can transfer microglia from M1 into M2 phenotype. And targeting the P2X7R-mediated signaling pathways helps to explore new therapy for pain alleviation. P2X7R antagonists also hold potential for translational and clinical applications in pain management. CONCLUSIONS Microglial P2X7R holds promise as a potential novel pharmacological target for clinical treatments due to its distinctive structure, function, and the development of antagonists.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Lei Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Yaoyuan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Haozhen Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Shukai Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Li‐an Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
34
|
Mao X, Shen J. Potential roles of enteric glial cells in Crohn's disease: A critical review. Cell Prolif 2024; 57:e13536. [PMID: 37551711 PMCID: PMC10771111 DOI: 10.1111/cpr.13536] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/09/2023] Open
Abstract
Enteric glial cells in the enteric nervous system are critical for the regulation of gastrointestinal homeostasis. Increasing evidence suggests two-way communication between enteric glial cells and both enteric neurons and immune cells. These interactions may be important in the pathogenesis of Crohn's disease (CD), a chronic relapsing disease characterized by a dysregulated immune response. Structural abnormalities in glial cells have been identified in CD. Furthermore, classical inflammatory pathways associated with CD (e.g., the nuclear factor kappa-B pathway) function in enteric glial cells. However, the specific mechanisms by which enteric glial cells contribute to CD have not been summarized in detail. In this review, we describe the possible roles of enteric glial cells in the pathogenesis of CD, including the roles of glia-immune interactions, neuronal modulation, neural plasticity, and barrier integrity. Additionally, the implications for the development of therapeutic strategies for CD based on enteric glial cell-mediated pathogenic processes are discussed.
Collapse
Affiliation(s)
- Xinyi Mao
- Division of Gastroenterology and HepatologyBaoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and HepatologyMinistry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive DiseaseShanghaiChina
| | - Jun Shen
- Division of Gastroenterology and HepatologyBaoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and HepatologyMinistry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive DiseaseShanghaiChina
| |
Collapse
|
35
|
Olivier E, Rat P. Role of Oxysterols in Ocular Degeneration Mechanisms and Involvement of P2X7 Receptor. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:277-292. [PMID: 38036885 DOI: 10.1007/978-3-031-43883-7_14] [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: 12/02/2023]
Abstract
Ocular degeneration, including cataracts, glaucoma, macular degeneration, and diabetic retinopathy, is a major public health challenge, as it affects the quality of life of millions of people worldwide and, in its advanced stages, leads to blindness. Ocular degeneration, although it can affect different parts of the eye, shares common characteristics such as oxysterols and the P2X7 receptor. Indeed, oxysterols, which are cholesterol derivatives, are associated with ocular degeneration pathogenesis and trigger inflammation and cell death pathways. Activation of the P2X7 receptor is also linked to ocular degeneration and triggers the same pathways. In age-related macular degeneration, these two key players have been associated, but further studies are needed to extrapolate this interrelationship to other ocular degenerations.
Collapse
Affiliation(s)
| | - Patrice Rat
- Université Paris Cité, CNRS, CiTCoM, Paris, France
| |
Collapse
|
36
|
Calderón-García AÁ, Valencia-Nieto L, Valencia-Sandonis C, López-de la Rosa A, Blanco-Vazquez M, Fernández I, García-Vázquez C, Arroyo-Del Arroyo C, González-García MJ, Enríquez-de-Salamanca A. Gene expression changes in conjunctival cells associated with contact lens wear and discomfort. Ocul Surf 2024; 31:31-42. [PMID: 38128761 DOI: 10.1016/j.jtos.2023.12.004] [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: 09/01/2023] [Revised: 10/27/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE This study aimed to analyze the differences in the expression of pain-related genes in conjunctival epithelial cells among symptomatic contact lens (CL) wearers (SCLWs), asymptomatic CL wearers (ACLWs), and non-CL wearers (non-CLWs). METHODS For this study, 60 participants (20 non-CLWs, 40 CLWs) were enrolled. The CLW group comprised 20 ACLWs and 20 SCLWs according to the Contact Lens Dry Eye Questionnaire short form©. Conjunctival cells were collected using impression cytology, and RNA was isolated and used to determine the expression levels of 85 human genes involved in neuropathic and inflammatory pain. The effects of CL wear and discomfort were evaluated using mixed-effects ANOVA with partially nested fixed-effects model. Gene set enrichment analysis was performed to assign biological meaning to sets of differentially expressed genes. RESULTS Six genes (CD200, EDN1, GRIN1, PTGS1, P2RX7, and TNF) were significantly upregulated in CLWs compared to non-CLWs. Eleven genes (ADORA1, BDKRB1, CACNA1B, DBH, GRIN1, GRM1, HTR1A, PDYN, PTGS1, P2RX3, and TNF) were downregulated in SCLWs compared to ACLWs. These genes were mainly related to pain, synaptic transmission and signaling, ion transport, calcium transport and concentration, and cell-cell signaling. CONCLUSIONS CL wear modified the expression of pain- and inflammation-related genes in conjunctival epithelial cells. These changes may be in part, along with other mechanisms, responsible for CL discomfort in SCLWs.
Collapse
Affiliation(s)
- Andrés Ángel Calderón-García
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Laura Valencia-Nieto
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Cristina Valencia-Sandonis
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Alberto López-de la Rosa
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - Marta Blanco-Vazquez
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain
| | - Itziar Fernández
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain; Department of Statistics and Operations Research, Universidad de Valladolid, Valladolid, Spain
| | - Carmen García-Vázquez
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain
| | - Cristina Arroyo-Del Arroyo
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain
| | - María J González-García
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Department of Theoretical Physics, Atomic and Optics, Universidad de Valladolid, Valladolid, Spain; Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain.
| | - Amalia Enríquez-de-Salamanca
- Institute of Applied Ophthalmobiology (IOBA), Universidad de Valladolid, Valladolid, Spain; Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Valladolid, Spain
| |
Collapse
|
37
|
Okon E, Gaweł-Bęben K, Jarzab A, Koch W, Kukula-Koch W, Wawruszak A. Therapeutic Potential of 1,8-Dihydroanthraquinone Derivatives for Breast Cancer. Int J Mol Sci 2023; 24:15789. [PMID: 37958772 PMCID: PMC10648492 DOI: 10.3390/ijms242115789] [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: 10/09/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy among women worldwide. In recent years, significant progress has been made in BC therapy. However, serious side effects resulting from the use of standard chemotherapeutic drugs, as well as the phenomenon of multidrug resistance (MDR), limit the effectiveness of approved therapies. Advanced research in the BC area is necessary to create more effective and safer forms of therapy to improve the outlook for individuals diagnosed with this aggressive neoplasm. For decades, plants and natural products with anticancer properties have been successfully utilized in treating various medical conditions. Anthraquinone derivatives are tricyclic secondary metabolites of natural origin that have been identified in plants, lichens, and fungi. They represent a few botanical families, e.g., Rhamnaceae, Rubiaceae, Fabaceae, Polygonaceae, and others. The review comprehensively covers and analyzes the most recent advances in the anticancer activity of 1,8-dihydroanthraquinone derivatives (emodin, aloe-emodin, hypericin, chrysophanol, rhein, and physcion) applied both individually, or in combination with other chemotherapeutic agents, in in vitro and in vivo BC models. The application of nanoparticles for in vitro and in vivo evidence in the context of 1,8-dihydroanthraquinone derivatives was also described.
Collapse
Affiliation(s)
- Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (E.O.); (A.J.)
| | - Katarzyna Gaweł-Bęben
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, 2 Sucharskiego, 35-225 Rzeszów, Poland;
| | - Agata Jarzab
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (E.O.); (A.J.)
| | - Wojciech Koch
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland;
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medical Plants Garden, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (E.O.); (A.J.)
| |
Collapse
|
38
|
Sainz RM, Rodriguez-Quintero JH, Maldifassi MC, Stiles BM, Wennerberg E. Tumour immune escape via P2X7 receptor signalling. Front Immunol 2023; 14:1287310. [PMID: 38022596 PMCID: PMC10643160 DOI: 10.3389/fimmu.2023.1287310] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
While P2X7 receptor expression on tumour cells has been characterized as a promotor of cancer growth and metastasis, its expression by the host immune system is central for orchestration of both innate and adaptive immune responses against cancer. The role of P2X7R in anti-tumour immunity is complex and preclinical studies have described opposing roles of the P2X7R in regulating immune responses against tumours. Therefore, few P2X7R modulators have reached clinical testing in cancer patients. Here, we review the prognostic value of P2X7R in cancer, how P2X7R have been targeted to date in tumour models, and we discuss four aspects of how tumours skew immune responses to promote immune escape via the P2X7R; non-pore functional P2X7Rs, mono-ADP-ribosyltransferases, ectonucleotidases, and immunoregulatory cells. Lastly, we discuss alternative approaches to offset tumour immune escape via P2X7R to enhance immunotherapeutic strategies in cancer patients.
Collapse
Affiliation(s)
- Ricardo M. Sainz
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jorge Humberto Rodriguez-Quintero
- Department of Cardiovascular and Thoracic Surgery, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY, United States
| | - Maria Constanza Maldifassi
- Department of Cardiovascular and Thoracic Surgery, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY, United States
| | - Brendon M. Stiles
- Department of Cardiovascular and Thoracic Surgery, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY, United States
| | - Erik Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
39
|
Mafra JCM, Boechat N, Teixeira GP, Faria RX. Synthetic molecules as P2X7 receptor antagonists: A medicinal chemistry update focusing the therapy of inflammatory diseases. Eur J Pharmacol 2023; 957:175999. [PMID: 37619787 DOI: 10.1016/j.ejphar.2023.175999] [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: 04/04/2023] [Revised: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Stimulation of the P2X7 receptor by extracellular adenosine 5'-triphosphate induces a series of responses in the organism, exceptionally protein cascades related to the proinflammatory process. This has made P2X7 a target for research on inflammatory diseases such as rheumatoid arthritis. Thus, the incessant search for new prototypes that aim to antagonize the action of P2X7 has been remarkable in recent decades, a factor that has already led to numerous clinical studies in humans. In this review, we present the key molecules developed over the years with potential inhibition of P2X7 and inflammation. In addition, an update with newly developed chemical classes with promising activity and results in clinical studies for human pathologies focusing on P2X7 inhibition.
Collapse
Affiliation(s)
- João Carlos Martins Mafra
- Laboratório de Síntese de Fármacos (LASFAR) - Farmanguinhos - Fiocruz Brazil; Instituto Federal do Rio de Janeiro - IFRJ, Rio de Janeiro, Brazil.
| | - Nubia Boechat
- Laboratório de Síntese de Fármacos (LASFAR) - Farmanguinhos - Fiocruz Brazil.
| | - Guilherme Pegas Teixeira
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz (IOC), Rio de Janeiro Fiocruz Brazil.
| | - Robson Xavier Faria
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz (IOC), Rio de Janeiro Fiocruz Brazil.
| |
Collapse
|
40
|
Huo A, Xiong X. PAICS as a potential target for cancer therapy linking purine biosynthesis to cancer progression. Life Sci 2023; 331:122070. [PMID: 37673296 DOI: 10.1016/j.lfs.2023.122070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/02/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
Tumor cells are required to undergo metabolic reprogramming for rapid development and progression, and one of the metabolic characteristics of cancer cells is the excessive synthesis and utilization of nucleotides. Abnormally increased nucleotides and their metabolites not only directly accelerate tumor cell progression but also indirectly act on stromal cells in the tumor microenvironment (TME) via a paracrine manner to regulate tumor progression. Purine nucleotides are mainly produced via de novo nucleotide synthesis in tumor cells; therefore, intervening in their synthesis has emerged as a promising strategy in anti-tumor therapy. De novo purine synthesis is a 10-step reaction catalyzed by six enzymes to synthesize inosine 5-monophosphate (IMP) and subsequently synthesize AMP and GMP. Phosphoribosylaminoimidazole carboxylase/phosphori-bosylaminoimidazole succinocarboxamide synthetase (PAICS) is a bifunctional enzyme that catalyzes de novo purine synthesis. Aberrantly elevated PAICS expression in various tumors is associated with poor prognosis. Evidence suggests that PAICS and its catalytic product, N-succinylcarboxamide-5-aminoimidazole ribonucleotide (SAICAR), could inhibit tumor cell apoptosis and promote the growth, epithelial-mesenchymal transition (EMT), invasion, and metastasis by regulating signaling pathways such as pyruvate kinase M2 (PKM2), extracellular signal-related kinases 1 and 2 (ERK1/2), focal adhesion kinase (FAK) and so on. This review summarizes the structure, biological functions and the molecular mechanisms of PAICS in cancer development and discusses its potential to be a target for tumor therapy.
Collapse
Affiliation(s)
- Anqi Huo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, China; The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, China; Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
41
|
Ai Y, Wang H, Liu L, Qi Y, Tang S, Tang J, Chen N. Purine and purinergic receptors in health and disease. MedComm (Beijing) 2023; 4:e359. [PMID: 37692109 PMCID: PMC10484181 DOI: 10.1002/mco2.359] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Purines and purinergic receptors are widely distributed throughout the human body. Purine molecules within cells play crucial roles in regulating energy metabolism and other cellular processes, while extracellular purines transmit signals through specific purinergic receptors. The ubiquitous purinergic signaling maintains normal neural excitability, digestion and absorption, respiratory movement, and other complex physiological activities, and participates in cell proliferation, differentiation, migration, and death. Pathological dysregulation of purinergic signaling can result in the development of various diseases, including neurodegeneration, inflammatory reactions, and malignant tumors. The dysregulation or dysfunction of purines and purinergic receptors has been demonstrated to be closely associated with tumor progression. Compared with other subtypes of purinergic receptors, the P2X7 receptor (P2X7R) exhibits distinct characteristics (i.e., a low affinity for ATP, dual functionality upon activation, the mediation of ion channels, and nonselective pores formation) and is considered a promising target for antitumor therapy, particularly in patients with poor response to immunotherapy This review summarizes the physiological and pathological significance of purinergic signaling and purinergic receptors, analyzes their complex relationship with tumors, and proposes potential antitumor immunotherapy strategies from tumor P2X7R inhibition, tumor P2X7R overactivation, and host P2X7R activation. This review provides a reference for clinical immunotherapy and mechanism investigation.
Collapse
Affiliation(s)
- Yanling Ai
- Department of OncologyHospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Hengyi Wang
- Department of Infectious DiseasesHospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Lu Liu
- School of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Yulin Qi
- Department of OphthalmologyThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
- Postdoctoral Research Station of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan ProvinceHospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Nianzhi Chen
- State Key Laboratory of Ultrasound in Medicine and EngineeringCollege of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| |
Collapse
|
42
|
Hetmann A, Szymczak B, Czarnecka J, Rusak T, Wiśniewski M, Wujak M, Roszek K. Adenylate kinase immobilized on graphene oxide impairs progression of human lung carcinoma epithelial cells through adenosinergic pathway. J Biomed Mater Res A 2023; 111:1565-1576. [PMID: 37162101 DOI: 10.1002/jbm.a.37555] [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: 01/04/2023] [Revised: 04/04/2023] [Accepted: 04/26/2023] [Indexed: 05/11/2023]
Abstract
Purinergic signaling, the oldest evolutionary transmitter system, has been increasingly studied as a pivotal target for novel anti-cancer therapies. In the present work, the developed nanobiocatalytic system consisting of adenylate kinase immobilized on graphene oxide (AK-GO) was characterized in terms of its physicochemical and biochemical properties. We put special emphasis on the AK-GO influence on purinergic signaling components, that is, ecto-nucleotides concentration and ecto-enzymes expression and activity in human lung carcinoma epithelial (A549) cells. The immobilization-dependent modification of AK kinetic parameters allowed for the removal of ATP excess while maintaining low ATP concentrations, efficient decrease in adenosine concentration, and control of the nucleotide balance in carcinoma cells. The cyto- and hemocompatibility of developed AK-GO nanobiocatalytic system indicates that it can be successfully harnessed for biomedical applications. In A549 cells treated with AK-GO nanobiocatalytic system, the significantly decreased adenosinergic signaling results in reduction of the proliferation and migration capability of cancer cells. This finding is particularly relevant in regard to AK-GO prospective anti-cancer applications.
Collapse
Affiliation(s)
- Anna Hetmann
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Bartosz Szymczak
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Joanna Czarnecka
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Tomasz Rusak
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Bialystok, Bialystok, Poland
| | - Marek Wiśniewski
- Department of Materials Chemistry, Adsorption and Catalysis, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Magdalena Wujak
- Department of Medicinal Chemistry, Collegium Medicum in Bydgoszcz, Faculty of Pharmacy, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Torun, Poland
| |
Collapse
|
43
|
Adinolfi E, De Marchi E, Grignolo M, Szymczak B, Pegoraro A. The P2X7 Receptor in Oncogenesis and Metastatic Dissemination: New Insights on Vesicular Release and Adenosinergic Crosstalk. Int J Mol Sci 2023; 24:13906. [PMID: 37762206 PMCID: PMC10531279 DOI: 10.3390/ijms241813906] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The tumor niche is an environment rich in extracellular ATP (eATP) where purinergic receptors have essential roles in different cell subtypes, including cancer, immune, and stromal cells. Here, we give an overview of recent discoveries regarding the role of probably the best-characterized purinergic receptor in the tumor microenvironment: P2X7. We cover the activities of the P2X7 receptor and its human splice variants in solid and liquid cancer proliferation, dissemination, and crosstalk with immune and endothelial cells. Particular attention is paid to the P2X7-dependent release of microvesicles and exosomes, their content, including ATP and miRNAs, and, in general, P2X7-activated mechanisms favoring metastatic spread and niche conditioning. Moreover, the emerging role of P2X7 in influencing the adenosinergic axis, formed by the ectonucleotidases CD39 and CD73 and the adenosine receptor A2A in cancer, is analyzed. Finally, we cover how antitumor therapy responses can be influenced by or can change P2X7 expression and function. This converging evidence suggests that P2X7 is an attractive therapeutic target for oncological conditions.
Collapse
Affiliation(s)
- Elena Adinolfi
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.D.M.); (M.G.); (A.P.)
| | - Elena De Marchi
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.D.M.); (M.G.); (A.P.)
| | - Marianna Grignolo
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.D.M.); (M.G.); (A.P.)
| | - Bartosz Szymczak
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Anna Pegoraro
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.D.M.); (M.G.); (A.P.)
| |
Collapse
|
44
|
Bandara V, Foeng J, Gundsambuu B, Norton TS, Napoli S, McPeake DJ, Tyllis TS, Rohani-Rad E, Abbott C, Mills SJ, Tan LY, Thompson EJ, Willet VM, Nikitaras VJ, Zheng J, Comerford I, Johnson A, Coombs J, Oehler MK, Ricciardelli C, Cowin AJ, Bonder CS, Jensen M, Sadlon TJ, McColl SR, Barry SC. Pre-clinical validation of a pan-cancer CAR-T cell immunotherapy targeting nfP2X7. Nat Commun 2023; 14:5546. [PMID: 37684239 PMCID: PMC10491676 DOI: 10.1038/s41467-023-41338-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell immunotherapy is a novel treatment that genetically modifies the patients' own T cells to target and kill malignant cells. However, identification of tumour-specific antigens expressed on multiple solid cancer types, remains a major challenge. P2X purinoceptor 7 (P2X7) is a cell surface expressed ATP gated cation channel, and a dysfunctional version of P2X7, named nfP2X7, has been identified on cancer cells from multiple tissues, while being undetectable on healthy cells. We present a prototype -human CAR-T construct targeting nfP2X7 showing potential antigen-specific cytotoxicity against twelve solid cancer types (breast, prostate, lung, colorectal, brain and skin). In xenograft mouse models of breast and prostate cancer, CAR-T cells targeting nfP2X7 exhibit robust anti-tumour efficacy. These data indicate that nfP2X7 is a suitable immunotherapy target because of its broad expression on human tumours. CAR-T cells targeting nfP2X7 have potential as a wide-spectrum cancer immunotherapy for solid tumours in humans.
Collapse
Affiliation(s)
- Veronika Bandara
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Jade Foeng
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Batjargal Gundsambuu
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Todd S Norton
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Silvana Napoli
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Dylan J McPeake
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Timona S Tyllis
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Elaheh Rohani-Rad
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Caitlin Abbott
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Stuart J Mills
- University of South Australia, STEM (Future Industries Institute) SA, Adelaide, 5095, Australia
| | - Lih Y Tan
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
| | - Emma J Thompson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
| | - Vasiliki M Willet
- Reproductive Cancer Research Group, Discipline Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Victoria J Nikitaras
- Reproductive Cancer Research Group, Discipline Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jieren Zheng
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Iain Comerford
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Adam Johnson
- Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Justin Coombs
- Carina Biotech, Level 2 Innovation & Collaboration Centre, UniSA Bradley Building, Adelaide, SA, 5001, Australia
| | - Martin K Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA, 5005, Australia
| | - Carmela Ricciardelli
- Reproductive Cancer Research Group, Discipline Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Allison J Cowin
- University of South Australia, STEM (Future Industries Institute) SA, Adelaide, 5095, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Michael Jensen
- Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Timothy J Sadlon
- Department of Gastroenterology, Women's and Children's Health Network, North Adelaide, SA, 5006, Australia
| | - Shaun R McColl
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
- Carina Biotech, Level 2 Innovation & Collaboration Centre, UniSA Bradley Building, Adelaide, SA, 5001, Australia
| | - Simon C Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia.
- Carina Biotech, Level 2 Innovation & Collaboration Centre, UniSA Bradley Building, Adelaide, SA, 5001, Australia.
- Department of Gastroenterology, Women's and Children's Health Network, North Adelaide, SA, 5006, Australia.
| |
Collapse
|
45
|
Jia W, Huang Z, Zhou L, Liou YC, Di Virgilio F, Ulrich H, Illes P, Zhang W, Huang C, Tang Y. Purinergic signalling in cancer therapeutic resistance: From mechanisms to targeting strategies. Drug Resist Updat 2023; 70:100988. [PMID: 37413937 DOI: 10.1016/j.drup.2023.100988] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023]
Abstract
Purinergic signalling, consisting of extracellular purines and purinergic receptors, modulates cell proliferation, invasion and immunological reaction during cancer progression. Here, we focus on current evidence that suggests the crucial role of purinergic signalling in mediating cancer therapeutic resistance, the major obstacle in cancer treatment. Mechanistically, purinergic signalling can modulate the tumor microenvironment (TME), epithelial-mesenchymal transition (EMT) and anti-tumor immunity, thus affecting drug sensitivity of tumor cells. Currently, some agents attempting to target purinergic signalling either in tumor cells or in tumor-associated immune cells are under preclinical or clinical investigation. Moreover, nano-based delivery technologies significantly improve the efficacy of agents targeting purinergic signalling. In this review article, we summarize the mechanisms of purinergic signalling in promoting cancer therapeutic resistance and discuss the potentials and challenges of targeting purinergic signalling in future cancer treatment.
Collapse
Affiliation(s)
- Wenhui Jia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhao Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yih-Cherng Liou
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117573, Singapore; Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117573, Singapore
| | | | - Henning Ulrich
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, Leipzig, Germany
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; Institute of TCM-Based Stress Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
46
|
Cevoli F, Arnould B, Peralta FA, Grutter T. Untangling Macropore Formation and Current Facilitation in P2X7. Int J Mol Sci 2023; 24:10896. [PMID: 37446075 DOI: 10.3390/ijms241310896] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Macropore formation and current facilitation are intriguing phenomena associated with ATP-gated P2X7 receptors (P2X7). Macropores are large pores formed in the cell membrane that allow the passage of large molecules. The precise mechanisms underlying macropore formation remain poorly understood, but recent evidence suggests two alternative pathways: a direct entry through the P2X7 pore itself, and an indirect pathway triggered by P2X7 activation involving additional proteins, such as TMEM16F channel/scramblase. On the other hand, current facilitation refers to the progressive increase in current amplitude and activation kinetics observed with prolonged or repetitive exposure to ATP. Various mechanisms, including the activation of chloride channels and intrinsic properties of P2X7, have been proposed to explain this phenomenon. In this comprehensive review, we present an in-depth overview of P2X7 current facilitation and macropore formation, highlighting new findings and proposing mechanistic models that may offer fresh insights into these untangled processes.
Collapse
Affiliation(s)
- Federico Cevoli
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Benoit Arnould
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Francisco Andrés Peralta
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
- Instituto de Neurociencias, CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - Thomas Grutter
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
- University of Strasbourg Institute for Advanced Studies (USIAS), 67000 Strasbourg, France
| |
Collapse
|
47
|
Norouzi A, Liaghat M, Bakhtiyari M, Noorbakhsh Varnosfaderani SM, Zalpoor H, Nabi-Afjadi M, Molania T. The potential role of COVID-19 in progression, chemo-resistance, and tumor recurrence of oral squamous cell carcinoma (OSCC). Oral Oncol 2023; 144:106483. [PMID: 37421672 DOI: 10.1016/j.oraloncology.2023.106483] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023]
Abstract
Numerous studies have revealed that cancer patients are more likely to develop severe Coronavirus disease-2019 (COVID-19), which can cause mortality, as well as cancer progression and treatment failure. Among these patients who may be particularly vulnerable to severe COVID-19 and COVID-19-associated cancer progression are those with oral squamous cell carcinoma (OSCC). In this regard, therapeutic approaches must be developed to lower the risk of cancer development, chemo-resistance, tumor recurrence, and death in OSCC patients with COVID-19. It may be helpful to comprehend the cellular and molecular mechanisms by which the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) contributes to these problems. In this line, in this review, we described the potential cellular and molecular mechanisms that SARS-CoV-2 can exert its role and based on them pharmacological targeted therapies were suggested. However, in this study, we encourage more investigations in the future to uncover other cellular and molecular mechanisms of action of SARS-CoV-2 to develop beneficial therapeutic strategies for such patients.
Collapse
Affiliation(s)
- Ali Norouzi
- Department of Oral Medicine, Dental Research Center, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahsa Liaghat
- Department of Medical Laboratory sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran; Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran; Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran; Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of biological sciences, Tarbiat Modares University, Tehran, Iran.
| | - Tahereh Molania
- Department of Oral Medicine, Dental Research Center, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
48
|
Kao YC, Chang YW, Lai CP, Chang NW, Huang CH, Chen CS, Huang HC, Juan HF. Ectopic ATP synthase stimulates the secretion of extracellular vesicles in cancer cells. Commun Biol 2023; 6:642. [PMID: 37322056 PMCID: PMC10272197 DOI: 10.1038/s42003-023-05008-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
ABSTARCT Ectopic ATP synthase on the plasma membrane (eATP synthase) has been found in various cancer types and is a potential target for cancer therapy. However, whether it provides a functional role in tumor progression remains unclear. Here, quantitative proteomics reveals that cancer cells under starvation stress express higher eATP synthase and enhance the production of extracellular vesicles (EVs), which are vital regulators within the tumor microenvironment. Further results show that eATP synthase generates extracellular ATP to stimulate EV secretion by enhancing P2X7 receptor-triggered Ca2+ influx. Surprisingly, eATP synthase is also located on the surface of tumor-secreted EVs. The EVs-surface eATP synthase increases the uptake of tumor-secreted EVs in Jurkat T-cells via association with Fyn, a plasma membrane protein found in immune cells. The eATP synthase-coated EVs uptake subsequently represses the proliferation and cytokine secretion of Jurkat T-cells. This study clarifies the role of eATP synthase on EV secretion and its influence on immune cells.
Collapse
Affiliation(s)
- Yi-Chun Kao
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Yi-Wen Chang
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Charles P Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
| | - Nai-Wen Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 106, Taiwan
| | - Chen-Hao Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 106, Taiwan
| | - Chien-Sheng Chen
- Department of Food Safety / Hygiene and Risk Management, National Cheng Kung University, Tainan, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Hsueh-Fen Juan
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan.
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 106, Taiwan.
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 106, Taiwan.
- Center for Computational and Systems Biology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
49
|
Rupert M, Bhattacharya A, Sivcev S, Knezu M, Cimicka J, Zemkova H. Identification of residues in the first transmembrane domain of the P2X7 that regulates receptor trafficking, sensitization, and dye uptake function. J Neurochem 2023; 165:874-891. [PMID: 36945903 DOI: 10.1111/jnc.15813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
P2X receptors (P2X1-7) are trimeric ion channels activated by extracellular ATP. Each P2X subunit contains two transmembrane helices (TM1 and TM2). We substituted all residues in TM1 of rat P2X7 with alanine or leucine one by one, expressed mutants in HEK293T cells, and examined the pore permeability by recording both membrane currents and fluorescent dye uptake in response to agonist application. Alanine substitution of G27, K30, H34, Y40, F43, L45, M46, and D48 inhibited agonist-stimulated membrane current and dye uptake, and all but one substitution, D48A, prevented surface expression. Mutation V41A partially reduced both membrane current and dye uptake, while W31A and A44L showed reduced dye uptake not accompanied by reduced membrane current. Mutations T28A, I29A, and L33A showed small changes in agonist sensitivity, but they had no or small impact on dye uptake function. Replacing charged residues with residues of the same charge (K30R, H34K, and D48E) rescued receptor function, while replacement with residues of opposite charge inhibited (K30E and H34E) or potentiated (D48K) receptor function. Prolonged stimulation with agonist-induced current facilitation and a leftward shift in the dose-response curve in the P2X7 wild-type and most functional mutants, but sensitization was absent in the W31A, L33A, and A44L. Detailed analysis of the decay of responses revealed two kinetically distinct mechanisms of P2X7 deactivation: fast represents agonist unbinding, and slow might represent resetting of the receptor to the resting closed state. These results indicate that conserved and receptor-specific TM1 residues control surface expression of the P2X7 protein, non-polar residues control receptor sensitization, and D48 regulates intrinsic channel properties.
Collapse
Affiliation(s)
- Marian Rupert
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Anirban Bhattacharya
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Sonja Sivcev
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Faculty of Sciences, Charles University in Prague, Prague, Czech Republic
| | - Michal Knezu
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Faculty of Sciences, Charles University in Prague, Prague, Czech Republic
| | - Jana Cimicka
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Hana Zemkova
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
50
|
Pacheco PAF, Faria JV, Silva AC, von Ranke NL, Silva RC, Rodrigues CR, da Rocha DR, Faria RX. In silico and pharmacological study of N,S-acetal juglone derivatives as inhibitors of the P2X7 receptor-promoted in vitro and in vivo inflammatory response. Biomed Pharmacother 2023; 162:114608. [PMID: 37003033 DOI: 10.1016/j.biopha.2023.114608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/01/2023] Open
Abstract
Purinergic receptors are transmembrane proteins responsive to extracellular nucleotides and are expressed by several cell types throughout the human body. Among all identified subtypes, the P2×7 receptor has emerged as a relevant target for the treatment of inflammatory disease. Several clinical trials have been conducted to evaluate the effectiveness of P2×7R antagonists. However, to date, no selective antagonist has reached clinical use. In this work, we report the pharmacological evaluation of eleven N, S-acetal juglone derivatives as P2×7R inhibitors. Using in vitro assays and in vivo experimental models, we identified one derivative with promising inhibitory activity and low toxicity. Our in silico studies indicate that the 1,4-naphthoquinone moiety might be a valuable molecular scaffold for the development of novel P2×7R antagonists, as suggested by our previous studies.
Collapse
|