1
|
Zhang WJ, Shi QM, Li TZ, Huang YW. G protein coupled P2Y2 receptor as a regulatory molecule in cancer progression. Arch Biochem Biophys 2024; 762:110194. [PMID: 39486566 DOI: 10.1016/j.abb.2024.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/01/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The occurrence and development of cancer involves the participation of many factors, its pathological mechanism is far more complicated than other diseases, and the treatment is also extremely difficult. Although the treatment of cancer adopts diversified methods to improve the survival rate and quality of life of patients, but the drug resistance, metastasis and recurrence of cancer cause most patients to fail in treatment. Therefore, exploring new molecular targets in cancer pathology is of great value for improving and preventing the treatment of cancer. Fortunately, the P2Y2 purinergic receptor (P2Y2 receptor) in the G protein-coupled receptor family has been recognized for regulating cancer progression. Agonist activated P2Y2 receptor has a certain contribution to the growth and metastasis of tumor cells. P2Y2 receptor activation participates in cancer progression by regulating calcium ion channels and classical signaling pathways (such as PLC-PKC and PI3K/AKT). It has the effect of anti-tumor therapy by inhibiting the activation of P2Y2 receptor (the use of antagonist) and reducing its expression. Therefore, in this article, we focus on the expression patterns of P2Y2 receptor in cancer and potential pharmacological targets as anti-cancer treatments.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Rehabilitation Medicine Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi province, China
| | - Qing-Ming Shi
- Orthopedic Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi province, China
| | - Teng-Zheng Li
- Orthopedic Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi province, China
| | - Ya-Wei Huang
- Urology Department, The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang City, China.
| |
Collapse
|
2
|
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
|
3
|
Shatat AAS. P2Y 11 receptor is a critical regulator of extracellular ATP-mediated premature senescence in lung fibroblasts: Implications of ER-Ca +2 release/mitochondrial ROS production signaling pathway. Purinergic Signal 2024:10.1007/s11302-024-10036-9. [PMID: 38977636 DOI: 10.1007/s11302-024-10036-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Affiliation(s)
- Abdel-Aziz S Shatat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt.
| |
Collapse
|
4
|
Mull ML, Pratt SJP, Thompson KN, Annis DA, Gad AA, Lee RM, Chang KT, Stemberger MB, Ju JA, Gilchrist DE, Boyman L, Vitolo MI, Lederer WJ, Martin SS. Disruption of P2Y2 signaling promotes breast tumor cell dissemination by reducing ATP-dependent calcium elevation and actin localization to cell junctions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.31.533191. [PMID: 37034765 PMCID: PMC10081304 DOI: 10.1101/2023.03.31.533191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The tumor microenvironment and wound healing after injury both contain extremely high concentrations of the extracellular signaling molecule, adenosine triphosphate (ATP) compared to normal tissue. P2Y2 receptor, an ATP-activated purinergic receptor, is typically associated with pulmonary, endothelial, and neurological cell signaling. Here we report its role and importance in breast epithelial cell signaling and how it is altered in metastatic breast cancer. In response to ATP activation, P2Y2 receptor signaling causes an increase of intracellular Ca 2+ in non-tumorigenic breast epithelial cells, while their tumorigenic and metastatic counterparts have significantly reduced Ca 2+ responses. The non-tumorigenic cells respond to increased Ca 2+ with actin polymerization and localization to cell edges, while the metastatic cells remained unaffected. The increase in intracellular Ca 2+ after ATP stimulation was blunted using a P2Y2 antagonist, which also prevented actin mobilization and caused cell dissemination from spheroids in non-tumorigenic breast epithelial cells. Furthermore, the lack of Ca 2+ concentration changes and actin mobilization in the metastatic breast cancer cells could be due to reduced P2Y2 expression, which correlates with poorer overall survival in breast cancer patients. This study elucidates rapid changes that occur after elevated intracellular Ca 2+ in breast epithelial cells and how metastatic cancer cells have adapted to evade this cellular response.
Collapse
|
5
|
Bischof H, Maier S, Koprowski P, Kulawiak B, Burgstaller S, Jasińska J, Serafimov K, Zochowska M, Gross D, Schroth W, Matt L, Juarez Lopez DA, Zhang Y, Bonzheim I, Büttner FA, Fend F, Schwab M, Birkenfeld AL, Malli R, Lämmerhofer M, Bednarczyk P, Szewczyk A, Lukowski R. mitoBK Ca is functionally expressed in murine and human breast cancer cells and potentially contributes to metabolic reprogramming. eLife 2024; 12:RP92511. [PMID: 38808578 PMCID: PMC11136494 DOI: 10.7554/elife.92511] [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: 05/30/2024] Open
Abstract
Alterations in the function of K+ channels such as the voltage- and Ca2+-activated K+ channel of large conductance (BKCa) reportedly promote breast cancer (BC) development and progression. Underlying molecular mechanisms remain, however, elusive. Here, we provide electrophysiological evidence for a BKCa splice variant localized to the inner mitochondrial membrane of murine and human BC cells (mitoBKCa). Through a combination of genetic knockdown and knockout along with a cell permeable BKCa channel blocker, we show that mitoBKCa modulates overall cellular and mitochondrial energy production, and mediates the metabolic rewiring referred to as the 'Warburg effect', thereby promoting BC cell proliferation in the presence and absence of oxygen. Additionally, we detect mitoBKCa and BKCa transcripts in low or high abundance, respectively, in clinical BC specimens. Together, our results emphasize, that targeting mitoBKCa could represent a treatment strategy for selected BC patients in future.
Collapse
Affiliation(s)
- Helmut Bischof
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
| | - Selina Maier
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
- Dr Margarete Fischer-Bosch Institute of Clinical PharmacologyStuttgartGermany
| | - Piotr Koprowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Sandra Burgstaller
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
- NMI Natural and Medical Sciences Institute at the University of TübingenReutlingenGermany
- Center for Medical Research, CF Bioimaging, Medical University of GrazGrazAustria
| | - Joanna Jasińska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Kristian Serafimov
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, University of TübingenTübingenGermany
| | - Monika Zochowska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Dominic Gross
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
| | - Werner Schroth
- Dr Margarete Fischer-Bosch Institute of Clinical PharmacologyStuttgartGermany
- University of TübingenTübingenGermany
| | - Lucas Matt
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
| | | | - Ying Zhang
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, University Hospital TübingenTübingenGermany
| | - Florian A Büttner
- Dr Margarete Fischer-Bosch Institute of Clinical PharmacologyStuttgartGermany
- University of TübingenTübingenGermany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital TübingenTübingenGermany
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical PharmacologyStuttgartGermany
- iFIT Cluster of Excellence (EXC 2180) “Image-guided and Functionally Instructed Tumor Therapies”, University of TübingenTübingenGermany
- Department of Clinical Pharmacology, Universityhostpital of TübingenTübingenGermany
- Department of Biochemistry and Pharmacy, University of TübingenTübingenGermany
- German Cancer Consortium (DKTK), German Cancer Research Center, Partner Site TübingenTübingenGermany
| | - Andreas L Birkenfeld
- Medical Clinic IV, University Hospital TübingenTübingenGermany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, University of TübingenTübingenGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Roland Malli
- Center for Medical Research, CF Bioimaging, Medical University of GrazGrazAustria
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of GrazGrazAustria
- BioTechMed GrazGrazAustria
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, University of TübingenTübingenGermany
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW)WarsawPoland
| | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of SciencesWarsawPoland
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of TübingenTübingenGermany
| |
Collapse
|
6
|
Volonte D, Benson CJ, Daugherty SL, Beckel JM, Trebak M, Galbiati F. Purinergic signaling promotes premature senescence. J Biol Chem 2024; 300:107145. [PMID: 38460941 PMCID: PMC11002311 DOI: 10.1016/j.jbc.2024.107145] [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: 01/24/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
Extracellular ATP activates P2 purinergic receptors. Whether purinergic signaling is functionally coupled to cellular senescence is largely unknown. We find that oxidative stress induced release of ATP and caused senescence in human lung fibroblasts. Inhibition of P2 receptors limited oxidative stress-induced senescence, while stimulation with exogenous ATP promoted premature senescence. Pharmacological inhibition of P2Y11 receptor (P2Y11R) inhibited premature senescence induced by either oxidative stress or ATP, while stimulation with a P2Y11R agonist was sufficient to induce cellular senescence. Our data show that both extracellular ATP and a P2Y11R agonist induced calcium (Ca++) release from the endoplasmic reticulum (ER) and that either inhibition of phospholipase C or intracellular Ca++ chelation impaired ATP-induced senescence. We also find that Ca++ that was released from the ER, following ATP-mediated activation of phospholipase C, entered mitochondria in a manner dependent on P2Y11R activation. Once in mitochondria, excessive Ca++ promoted the production of reactive oxygen species in a P2Y11R-dependent fashion, which drove development of premature senescence of lung fibroblasts. Finally, we show that conditioned medium derived from senescent lung fibroblasts, which were induced to senesce through the activation of ATP/P2Y11R-mediated signaling, promoted the proliferation of triple-negative breast cancer cells and their tumorigenic potential by secreting amphiregulin. Our study identifies the existence of a novel purinergic signaling pathway that links extracellular ATP to the development of a protumorigenic premature senescent phenotype in lung fibroblasts that is dependent on P2Y11R activation and ER-to-mitochondria calcium signaling.
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Cory J Benson
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stephanie L Daugherty
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jonathan M Beckel
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ferruccio Galbiati
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
7
|
Vázquez-Cuevas FG, Reyna-Jeldes M, Velázquez-Miranda E, Coddou C. Transactivation of receptor tyrosine kinases by purinergic P2Y and adenosine receptors. Purinergic Signal 2023; 19:613-621. [PMID: 36529846 PMCID: PMC10754767 DOI: 10.1007/s11302-022-09913-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Transactivation of receptor tyrosine kinases (RTK) is a crosstalk mechanism exhibited by G-protein-coupled receptors (GPCR) to activate signaling pathways classically associated with growth factors. The discovery of RTK transactivation was a breakthrough in signal transduction that contributed to developing current concepts in intracellular signaling. RTK transactivation links GPCR signaling to important cellular processes, such as cell proliferation and differentiation, and explains the functional diversity of these receptors. Purinergic (P2Y and adenosine) receptors belong to class A of GPCR; in the present work, we systematically review the experimental evidence showing that purinergic receptors have the ability to transactivate RTK in multiple tissues and physiopathological conditions resulting in the modulation of cellular physiology. Of particular relevance, the crosstalk between purinergic receptors and epidermal growth factor receptor is a redundant pathway that participates in multiple pathophysiological processes. Specific and detailed knowledge of purinergic receptor-regulated pathways advances our understanding of the complexity of GPCR signal transduction and opens the way for pharmacologic intervention in the pathological context.
Collapse
Affiliation(s)
- F G Vázquez-Cuevas
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla # 3001, Juriquilla, Querétaro, 76230, México.
| | - M Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo, 1781421, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, 8331150, Chile
- Núcleo Para El Estudio del Cáncer a Nivel Básico, Aplicado Y Clínico, Universidad Católica del Norte, Larrondo 1281, Coquimbo , 1781421, Chile
| | - E Velázquez-Miranda
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla # 3001, Juriquilla, Querétaro, 76230, México
| | - C Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo, 1781421, Chile.
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, 8331150, Chile.
- Núcleo Para El Estudio del Cáncer a Nivel Básico, Aplicado Y Clínico, Universidad Católica del Norte, Larrondo 1281, Coquimbo , 1781421, Chile.
| |
Collapse
|
8
|
Nuñez-Rios JD, Ulrich H, Díaz-Muñoz M, Lameu C, Vázquez-Cuevas FG. Purinergic system in cancer stem cells. Purinergic Signal 2023:10.1007/s11302-023-09976-5. [PMID: 37966629 DOI: 10.1007/s11302-023-09976-5] [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: 06/10/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Accumulating evidence supports the idea that cancer stem cells (CSCs) are those with the capacity to initiate tumors, generate phenotypical diversity, sustain growth, confer drug resistance, and orchestrate the spread of tumor cells. It is still controversial whether CSCs originate from normal stem cells residing in the tissue or cancer cells from the tumor bulk that have dedifferentiated to acquire stem-like characteristics. Although CSCs have been pointed out as key drivers in cancer, knowledge regarding their physiology is still blurry; thus, research focusing on CSCs is essential to designing novel and more effective therapeutics. The purinergic system has emerged as an important autocrine-paracrine messenger system with a prominent role at multiple levels of the tumor microenvironment, where it regulates cellular aspects of the tumors themselves and the stromal and immune systems. Recent findings have shown that purinergic signaling also participates in regulating the CSC phenotype. Here, we discuss updated information regarding CSCs in the purinergic system and present evidence supporting the idea that elements of the purinergic system expressed by this subpopulation of the tumor represent attractive pharmacological targets for proposing innovative anti-cancer therapies.
Collapse
Affiliation(s)
- J D Nuñez-Rios
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México
| | - H Ulrich
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - M Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México
| | - C Lameu
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México.
| |
Collapse
|
9
|
Bustamante Eduardo M, Keller I, Schuster N, Aebi S, Jaggi R. Molecular characterization of breast cancer cell pools with normal or reduced ability to respond to progesterone: a study based on RNA-seq. J Genet Eng Biotechnol 2023; 21:81. [PMID: 37550554 PMCID: PMC10406740 DOI: 10.1186/s43141-023-00541-6] [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: 05/25/2022] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND About one-third of patients with estrogen receptor alpha (ERα)-positive breast cancer have tumors which are progesterone receptor (PR) negative. PR is an important prognostic factor in breast cancer. Patients with ERα-positive/PR-negative tumors have shorter disease-free and overall survival than patients with ERα-positive/PR-positive tumors. New evidence has shown that progesterone (P4) has an anti-proliferative effect in ERα-positive breast cancer cells. However, the role of PR in breast cancer is only poorly understood. METHODS We disrupted the PR gene (PGR) in ERα-positive/PR-positive T-47D cells using the CRISPR/Cas9 system. This resulted in cell pools we termed PR-low as P4 mediated effects were inhibited or blocked compared to control T-47D cells. We analyzed the gene expression profiles of PR-low and control T-47D cells in the absence of hormone and upon treatment with P4 alone or P4 together with estradiol (E2). Differentially expressed (DE) genes between experimental groups were characterized based on RNA-seq and Gene Ontology (GO) enrichment analyses. RESULTS The overall gene expression pattern was very similar between untreated PR-low and untreated control T-47D cells. More than 6000 genes were DE in control T-47D cells upon stimulation with P4 or P4 plus E2. When PR-low pools were subjected to the same hormonal treatment, up- or downregulation was either blocked/absent or consistently lower. We identified more than 3000 genes that were DE between hormone-treated PR-low and control T-47D cells. GO analysis revealed seven significantly enriched biological processes affected by PR and associated with G protein-coupled receptor (GPCR) pathways which have been described to support growth, invasiveness, and metastasis in breast cancer cells. CONCLUSIONS The present study provides new insights into the complex role of PR in ERα-positive/PR-positive breast cancer cells. Many of the genes affected by PR are part of central biological processes of tumorigenesis.
Collapse
Affiliation(s)
- Mariana Bustamante Eduardo
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, USA.
| | - Irene Keller
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nathalie Schuster
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stefan Aebi
- Department of Medical Oncology, Cantonal Hospital, Lucerne, Switzerland
| | - Rolf Jaggi
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
10
|
Cui Y, Li C, Zeng X, Wei X, Li P, Cheng J, Xu Q, Yang Y. ATP purinergic receptor signalling promotes Sca-1 + cell proliferation and migration for vascular remodelling. Cell Commun Signal 2023; 21:173. [PMID: 37430253 PMCID: PMC10332060 DOI: 10.1186/s12964-023-01185-2] [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: 02/16/2023] [Accepted: 06/06/2023] [Indexed: 07/12/2023] Open
Abstract
AIMS Vascular resident stem cells expressing stem cell antigen-1 (Sca-1+ cells) promote vascular regeneration and remodelling following injury through migration, proliferation and differentiation. The aim of this study was to examine the contributions of ATP signalling through purinergic receptor type 2 (P2R) isoforms in promoting Sca-1+ cell migration and proliferation after vascular injury and to elucidate the main downstream signalling pathways. METHODS AND RESULTS ATP-evoked changes in isolated Sca-1+ cell migration were examined by transwell assays, proliferation by viable cell counting assays and intracellular Ca2+ signalling by fluorometry, while receptor subtype contributions and downstream signals were examined by pharmacological or genetic inhibition, immunofluorescence, Western blotting and quantitative RT-PCR. These mechanisms were further examined in mice harbouring TdTomato-labelled Sca-1+ cells with and without Sca-1+-targeted P2R knockout following femoral artery guidewire injury. Stimulation with ATP promoted cultured Sca-1+ cell migration, induced intracellular free calcium elevations primarily via P2Y2R stimulation and accelerated proliferation mainly via P2Y6R stimulation. Enhanced migration was inhibited by the ERK blocker PD98059 or P2Y2R-shRNA, while enhanced proliferation was inhibited by the P38 inhibitor SB203580. Femoral artery guidewire injury of the neointima increased the number of TdTomato-labelled Sca-1+ cells, neointimal area and the ratio of neointimal area to media area at 3 weeks post-injury, and all of these responses were reduced by P2Y2R knockdown. CONCLUSIONS ATP induces Sca-1+ cell migration through the P2Y2R-Ca2+-ERK signalling pathway, and enhances proliferation through the P2Y6R-P38-MAPK signalling pathway. Both pathways are essential for vascular remodelling following injury. Video Abstract.
Collapse
Affiliation(s)
- Yiqin Cui
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Chunshu Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Xinyi Zeng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Xiaoyu Wei
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Jun Cheng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Qingbo Xu
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China.
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China.
| |
Collapse
|
11
|
Mata-Martínez E, Gonzalez-Gallardo A, Díaz-Muñoz M, Vázquez-Cuevas FG. Purinergic Activation of Store-Operated Calcium Entry (SOCE) Regulates Cell Migration in Metastatic Ovarian Cancer Cells. Pharmaceuticals (Basel) 2023; 16:944. [PMID: 37513856 PMCID: PMC10384695 DOI: 10.3390/ph16070944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Store-operated calcium entry (SOCE) is an important process in calcium signaling. Its role in physiological and pathological events is well recognized. However, in cancerous systems, the importance of SOCE in relation to the degree of cancer aggressiveness, as well as its regulation by ligands such as purinergic molecules, are not well documented. This study aimed to characterize a differential effect of the P2Y2 receptor (promoted by UTP of 10 µM and inhibited by ARC118925XX of 1 µM) on intracellular calcium response between metastatic (SKOV-3) and non-metastatic (CAOV-3) ovarian cell lines in conditions of normal (1.5 mM) and zero extracellular calcium concentration. The sustained calcium influx observed exclusively in SKOV-3 cells was associated with the presence of SOCE (promoted by thapsigargin (74.81 ± 0.94 ΔF) and sensitive to 2-APB (20.60 ± 0.85 ΔF)), whereas its absence in CAOV-3 cells (26.2 ± 6.1 ΔF) was correlated with a low expression of ORAI1. The relevance of SOCE in metastatic SKOV-3 cells was further corroborated when 2-APB significantly inhibited (40.4 ± 2.8% of covered area) UTP-induced cell migration (54.6 ± 3.7% of covered area). In conclusion, our data suggest that SOCE activation elicited by the P2Y2 receptor is involved in the aggressiveness of ovarian cancer cells.
Collapse
Affiliation(s)
- Esperanza Mata-Martínez
- Departamento de Neurobiología Celular y Molecular, Universidad Nacional Autónoma de México, Boulevard Juriquilla#3001, Juriquilla 76230, Querétaro, Mexico
| | - Adriana Gonzalez-Gallardo
- Unidad de Proteogenómica, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla#3001, Juriquilla 76230, Querétaro, Mexico
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Universidad Nacional Autónoma de México, Boulevard Juriquilla#3001, Juriquilla 76230, Querétaro, Mexico
| | - Francisco G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Universidad Nacional Autónoma de México, Boulevard Juriquilla#3001, Juriquilla 76230, Querétaro, Mexico
| |
Collapse
|
12
|
Kaur J, Dora S. Purinergic signaling: Diverse effects and therapeutic potential in cancer. Front Oncol 2023; 13:1058371. [PMID: 36741002 PMCID: PMC9889871 DOI: 10.3389/fonc.2023.1058371] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Regardless of improved biological insights and therapeutic advances, cancer is consuming multiple lives worldwide. Cancer is a complex disease with diverse cellular, metabolic, and physiological parameters as its hallmarks. This instigates a need to uncover the latest therapeutic targets to advance the treatment of cancer patients. Purines are building blocks of nucleic acids but also function as metabolic intermediates and messengers, as part of a signaling pathway known as purinergic signaling. Purinergic signaling comprises primarily adenosine triphosphate (ATP) and adenosine (ADO), their analogous membrane receptors, and a set of ectonucleotidases, and has both short- and long-term (trophic) effects. Cells release ATP and ADO to modulate cellular function in an autocrine or paracrine manner by activating membrane-localized purinergic receptors (purinoceptors, P1 and P2). P1 receptors are selective for ADO and have four recognized subtypes-A1, A2A, A2B, and A3. Purines and pyrimidines activate P2 receptors, and the P2X subtype is ligand-gated ion channel receptors. P2X has seven subtypes (P2X1-7) and forms homo- and heterotrimers. The P2Y subtype is a G protein-coupled receptor with eight subtypes (P2Y1/2/4/6/11/12/13/14). ATP, its derivatives, and purinoceptors are widely distributed in all cell types for cellular communication, and any imbalance compromises the homeostasis of the cell. Neurotransmission, neuromodulation, and secretion employ fast purinergic signaling, while trophic purinergic signaling regulates cell metabolism, proliferation, differentiation, survival, migration, invasion, and immune response during tumor progression. Thus, purinergic signaling is a prospective therapeutic target in cancer and therapy resistance.
Collapse
Affiliation(s)
- Jasmeet Kaur
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sanchit Dora
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
13
|
Henningsen MB, McWhan K, Dam VS, Mele M, Hauerslev KR, Voss NCS, Dabir PD, Balling E, Pedersen HL, Vahl P, Johansen T, Tramm T, Christiansen PM, Boedtkjer E. Amplified Ca 2+ dynamics and accelerated cell proliferation in breast cancer tissue during purinergic stimulation. Int J Cancer 2022; 151:1150-1165. [PMID: 35657342 PMCID: PMC9544627 DOI: 10.1002/ijc.34147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022]
Abstract
Intracellular Ca2+ dynamics shape malignant behaviors of cancer cells. Whereas previous studies focused on cultured cancer cells, we here used breast organoids and colonic crypts freshly isolated from human and murine surgical biopsies. We performed fluorescence microscopy to evaluate intracellular Ca2+ concentrations in breast and colon cancer tissue with preferential focus on intracellular Ca2+ release in response to purinergic and cholinergic stimuli. Inhibition of the sarco‐/endoplasmic reticulum Ca2+ ATPase with cyclopiazonic acid elicited larger Ca2+ responses in breast cancer tissue, but not in colon cancer tissue, relative to respective normal tissue. The resting intracellular Ca2+ concentration was elevated, and ATP, UTP and acetylcholine induced strongly augmented intracellular Ca2+ responses in breast cancer tissue compared with normal breast tissue. In contrast, resting intracellular Ca2+ levels and acetylcholine‐induced increases in intracellular Ca2+ concentrations were unaffected and ATP‐ and UTP‐induced Ca2+ responses were smaller in colon cancer tissue compared with normal colon tissue. In accordance with the amplified Ca2+ responses, ATP and UTP substantially increased proliferative activity—evaluated by bromodeoxyuridine incorporation—in breast cancer tissue, whereas the effect was minimal in normal breast tissue. ATP caused cell death—identified with ethidium homodimer‐1 staining—in breast cancer tissue only at concentrations above the expected pathophysiological range. We conclude that intracellular Ca2+ responses are amplified in breast cancer tissue, but not in colon cancer tissue, and that nucleotide signaling stimulates breast cancer cell proliferation within the extracellular concentration range typical for solid cancer tissue.
Collapse
Affiliation(s)
| | - Kezia McWhan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Vibeke S Dam
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marco Mele
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Katrine R Hauerslev
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Ninna C S Voss
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Parag D Dabir
- Department on Pathology, Randers Regional Hospital, Randers, Denmark
| | - Eva Balling
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Helene L Pedersen
- Department on Pathology, Randers Regional Hospital, Randers, Denmark
| | - Pernille Vahl
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Tonje Johansen
- Department on Pathology, Randers Regional Hospital, Randers, Denmark
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Peer M Christiansen
- Department of Surgery, Randers Regional Hospital, Randers, Denmark.,Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Lau CC, Aminuddin A, Chan KM, Paterson IC, Law LM, Ng PY. Extracellular ATP Induced S-Phase Cell Cycle Arrest via P2Y Receptor-Activated ERK Signaling in Poorly Differentiated Oral Squamous Cell Carcinoma SAS Cells. Life (Basel) 2021; 11:1170. [PMID: 34833046 PMCID: PMC8624497 DOI: 10.3390/life11111170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/23/2021] [Accepted: 10/29/2021] [Indexed: 12/19/2022] Open
Abstract
Extracellular ATP in the tumor microenvironment exhibits either pro- or antitumor effect via interaction with P2Y receptors, but the intracellular signaling and functional roles of P2Y receptors in oral squamous cell carcinoma (OSCC) are unclear. We aimed to study the effect of ATP on OSCC cell lines and the potential mechanisms involved. Through GEPIA dataset analysis, high expression levels of mRNA encoding P2Y receptors, the ATP-induced G protein-coupled receptors, were associated with better overall patient survival in head and neck squamous cell carcinoma. qPCR analysis showed that the poorly differentiated OSCC SAS cell line, had higher P2RY1 expression level compared to the well-differentiated H103 and H376 cell lines. Western blotting and flow cytometry analyses revealed that ATP phosphorylated ERK and elevated intracellular calcium signaling in all tested cell lines. A significant S-phase cell cycle arrest was observed in SAS, and preincubation with the MEK inhibitor PD0325901 reversed the ATP-induced S-phase arrest. We further demonstrated that ATP induced a slight reduction in cell count and colony formation yet significant apoptosis in SAS. Overall, we postulate that the ATP-induced S-phase arrest effect in SAS cells may be regulated through P2Y receptor-mediated ERK signaling, thus suggesting a potential antitumor effect of ATP via interaction with its distinct profile of P2Y receptors.
Collapse
Affiliation(s)
- Chia Chih Lau
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (C.C.L.); (A.A.); (L.M.L.)
| | - Amnani Aminuddin
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (C.C.L.); (A.A.); (L.M.L.)
| | - Kok Meng Chan
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Ian C. Paterson
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Lok Mun Law
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (C.C.L.); (A.A.); (L.M.L.)
| | - Pei Yuen Ng
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (C.C.L.); (A.A.); (L.M.L.)
| |
Collapse
|
15
|
Alvarez CL, Troncoso MF, Espelt MV. Extracellular ATP and adenosine in tumor microenvironment: Roles in epithelial-mesenchymal transition, cell migration, and invasion. J Cell Physiol 2021; 237:389-400. [PMID: 34514618 DOI: 10.1002/jcp.30580] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022]
Abstract
Under nonpathological conditions, the extracellular nucleotide concentration remains constant and low (nM range) because of a close balance between ATP release and ATP consumption. This balance is completely altered in cancer disease. Adenine and uridine nucleotides are found in the extracellular space of tumors in high millimolar (mM) concentrations acting as extracellular signaling molecules. In general, although uridine nucleotides may be involved in different tumor cell responses, purinergic signaling in cancer is preferentially focused on adenine nucleotides and nucleosides. Extracellular ATP can bind to specific receptors (P receptors) triggering different responses, or it can be hydrolyzed by ectoenzymes bound to cell membranes to render the final product adenosine. The latter pathway plays an important role in the increase of adenosine in tumor microenvironment. In this study, we will focus on extracellular ATP and adenosine, their effects acting as ligands of specific receptors, activating ectoenzymes, and promoting epithelial-mesenchymal transition, migration, and invasion in cancer cells. Finding the roles that these nucleotides play in tumor microenvironment may be important to design new intervention strategies in cancer therapies.
Collapse
Affiliation(s)
- Cora L Alvarez
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini", Buenos Aires, Argentina
| | - María F Troncoso
- CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini", Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María V Espelt
- CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini", Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
16
|
Reyna-Jeldes M, Díaz-Muñoz M, Madariaga JA, Coddou C, Vázquez-Cuevas FG. Autocrine and paracrine purinergic signaling in the most lethal types of cancer. Purinergic Signal 2021; 17:345-370. [PMID: 33982134 PMCID: PMC8410929 DOI: 10.1007/s11302-021-09785-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer comprises a collection of diseases that occur in almost any tissue and it is characterized by an abnormal and uncontrolled cell growth that results in tumor formation and propagation to other tissues, causing tissue and organ malfunction and death. Despite the undeniable improvement in cancer diagnostics and therapy, there is an urgent need for new therapeutic and preventive strategies with improved efficacy and fewer side effects. In this context, purinergic signaling emerges as an interesting candidate as a cancer biomarker or therapeutic target. There is abundant evidence that tumor cells have significant changes in the expression of purinergic receptors, which comprise the G-protein coupled P2Y and AdoR families of receptors and the ligand-gated ion channel P2X receptors. Tumor cells also exhibit changes in the expression of nucleotidases and other enzymes involved in nucleotide metabolism, and the concentrations of extracellular nucleotides are significantly higher than those observed in normal cells. In this review, we will focus on the potential role of purinergic signaling in the ten most lethal cancers (lung, breast, colorectal, liver, stomach, prostate, cervical, esophagus, pancreas, and ovary), which together are responsible for more than 5 million annual deaths.
Collapse
Affiliation(s)
- M Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- Núcleo para el Estudio del Cáncer a nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta, Chile
| | - M Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, México
| | - J A Madariaga
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Núcleo para el Estudio del Cáncer a nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta, Chile
| | - C Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile.
- Núcleo para el Estudio del Cáncer a nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta, Chile.
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, México.
| |
Collapse
|
17
|
El-Ashmawy NE, El-Zamarany EA, Khedr NF, Selim HM, Khedr EG. Inhibition of PKC/MEK pathway suppresses β1-integrin and mitigates breast cancer cells proliferation. Toxicol Rep 2021; 8:1530-1537. [PMID: 34408972 PMCID: PMC8361284 DOI: 10.1016/j.toxrep.2021.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
PGE2 enhanced β1- integrin expression via EP1 receptor, PKC, MEK and NfҡB. FOXC2, E2F1 and survivin play a role in PGE2 mediated effect in MCF7 cells. PGE2 enhances breast cancer cell cycle through E2F1, FOXC2, survivin and β integrin. Biochemical mediators of PKC/MEK pathway could be considered as targets for breast cancer treatment.
Prostaglandin E2 (PGE2) and β1-integrin have been correlated with breast cancer, where both could enhance progression and metastasis. Protein kinase C (PKC) and MEK have played a vital role in breast cancer development. Our study was conducted to elucidate the effect of inhibition of E-prostanoid receptor 1 (EP1)/ PKC/ MEK/ β1-integrin pathway in mitigating breast cancer progression and to evaluate the role of the intermediate signals FOXC2, E2F1, NF-ҡB and survivin. MCF7 cells were treated with 17 -PT-PGE2, an EP1 agonist, for 24 h, and β1-integrin was measured. To MCF7 cells treated with 17-PT-PGE2, inhibitors of either EP1, MEK, PKC or NF-ҡB were added followed by measurement of β1-integrin gene expression and cell proliferation in each case. Addition of 17- PT-PGE2 to MCF7 cells showed enhancement of both cell proliferation, and cell cycle transition from G1 to S phase. In addition, activation of EP1 receptor increased β1-integrin expression. On the contrary, inhibition of EP1 receptor showed a decrease in the cell proliferation, β1-integrin expression and cells transition to S phase, but increased cell count in apoptotic phase. Selective inhibition of each of MEK, PKC, and NF-ҡB suppressed 17 -PT-PGE2-mediated β1-integrin expression as well as cell proliferation. Furthermore, FOXC2, phosphorylated NF-ҡB, E2F1, and survivin levels were upregulated with 17- PT-PGE2 and suppressed by MEK, PKC and NF-ҡB inhibitors. Targeting the biochemical mediators of PKC/MEK pathway may be of value in developing new chemical entities for cancer treatment.
Collapse
Affiliation(s)
| | - Enas A El-Zamarany
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Egypt
| | - Naglaa F Khedr
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Egypt
| | - Hend M Selim
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Egypt
| | - Eman G Khedr
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Egypt
| |
Collapse
|
18
|
Zhao Y, Chen X, Lyu S, Ding Z, Wu Y, Gao Y, Du J. Identification of novel P2X7R antagonists by using structure-based virtual screening and cell-based assays. Chem Biol Drug Des 2021; 98:192-205. [PMID: 33993620 DOI: 10.1111/cbdd.13867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
In the tumor microenvironment, inflammation and necrosis cause the accumulations of ATP extracellularly, and high concentrations of ATP can activate P2X7 receptors (P2X7R), which leads to the influx of Na+ , K+ , or Ca2+ into cells and trigger the downstream signaling pathways. P2X7R is a relatively unique ligand-gated ion channel, which is over-expressed in most tumor cells. The activated P2X7R facilitates the tumor growth, invasion, and metastasis. Inhibition of the P2X7R activation can be applied as a potential anti-tumor therapy strategy. There are currently no anti-tumor agents against P2X7R, though several P2X7R antagonists for indications such as anti-inflammatory and anti-depression were reported. In this study, we combined homology modeling (HM), virtual screening, and EB intake assay to characterize the structural features of P2X7R and identify several novel antagonists, which were chemically different from any other known P2X7R antagonists. The identified antagonists could effectively prevent the pore opening of P2X7R with IC50 values ranging from 29.14 to 35.34 μM. HM model showed the area between ATP-binding pocket, and allosteric sides were hydrophobic and suitable for small molecule interaction. Molecular docking indicated a universal binding mode, of which residues R294 and K311 were used as hydrogen bond donors to participate in antagonist interactions. The binding mode can potentially be utilized for inhibitor optimization for increased affinity, and the identified antagonists can be further tested for anti-cancer activity or may serve as chemical agents to study P2X7R related functions.
Collapse
Affiliation(s)
- Yunshuo Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaotong Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Sifan Lyu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhe Ding
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Kwon K, Hwang Y, Jung J, Tae G. Enhanced Transport and Permeation of a Polymeric Nanocarrier across the Retina by Mixing with ATP upon Intravitreal Injection. Pharmaceutics 2021; 13:463. [PMID: 33805533 PMCID: PMC8065980 DOI: 10.3390/pharmaceutics13040463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/18/2022] Open
Abstract
The outer part of the retina pigment epithelium (RPE) in the retina is the main site of neovascularization associated with retinal diseases. However, various obstacles interrupt the delivery of medicines across the RPE, mainly due to the well-developed tight junctions in the RPE. Currently, there is no practical formulation to overcome this issue. In this study, we demonstrated that simple mixing with adenosine tetraphosphate (ATP) has the potential to greatly enhance the transport and permeation of a polymeric nanocarrier across the retina via intravitreal administration. Chitosan-functionalized, pluronic-based nanocarrier (NC), which can deliver various biomolecules efficiently, was used as a polymeric nanocarrier. Mixing with ATP facilitated the diffusion of the nanocarrier in the vitreous humor by reducing the electrostatic interaction between NC and negatively charged glycosaminoglycans (GAGs) in the vitreous humor. Mixing with ATP also allowed the penetration of NC across the whole retina, and it resulted in a great increase (approximately nine times) in the transport of NC across the retina, as well as spreading it throughout the whole retina upon intravitreal administration in a mouse model. This enhanced permeation across the retina was specific to ATP but not to GTP, suggesting the possibility of P2Y receptor-mediated tight junction disruption by ATP.
Collapse
Affiliation(s)
| | | | | | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; (K.K.); (Y.H.); (J.J.)
| |
Collapse
|
20
|
Zefferino R, Piccoli C, Di Gioia S, Capitanio N, Conese M. How Cells Communicate with Each Other in the Tumor Microenvironment: Suggestions to Design Novel Therapeutic Strategies in Cancer Disease. Int J Mol Sci 2021; 22:ijms22052550. [PMID: 33806300 PMCID: PMC7961918 DOI: 10.3390/ijms22052550] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
Connexin- and pannexin (Panx)-formed hemichannels (HCs) and gap junctions (GJs) operate an interaction with the extracellular matrix and GJ intercellular communication (GJIC), and on account of this they are involved in cancer onset and progression towards invasiveness and metastatization. When we deal with cancer, it is not correct to omit the immune system, as well as neglecting its role in resisting or succumbing to formation and progression of incipient neoplasia until the formation of micrometastasis, nevertheless what really occurs in the tumor microenvironment (TME), which are the main players and which are the tumor or body allies, is still unclear. The goal of this article is to discuss how the pivotal players act, which can enhance or contrast cancer progression during two important process: "Activating Invasion and Metastasis" and the "Avoiding Immune Destruction", with a particular emphasis on the interplay among GJIC, Panx-HCs, and the purinergic system in the TME without disregarding the inflammasome and cytokines thereof derived. In particular, the complex and contrasting roles of Panx1/P2X7R signalosome in tumor facilitation and/or inhibition is discussed in regard to the early/late phases of the carcinogenesis. Finally, considering this complex interplay in the TME between cancer cells, stromal cells, immune cells, and focusing on their means of communication, we should be capable of revealing harmful messages that help the cancer growth and transform them in body allies, thus designing novel therapeutic strategies to fight cancer in a personalized manner.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
- Correspondence: ; Tel.: +39-0881-884673
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (N.C.)
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (N.C.)
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| |
Collapse
|
21
|
Targeting the purinergic pathway in breast cancer and its therapeutic applications. Purinergic Signal 2021; 17:179-200. [PMID: 33576905 PMCID: PMC7879595 DOI: 10.1007/s11302-020-09760-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/06/2020] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is the most frequent cause of death among women, representing a global public health problem. Here, we aimed to discuss the correlation between the purinergic system and BC, recognizing therapeutic targets. For this, we analyzed the interaction of extracellular nucleotides and nucleosides with the purinergic receptors P1 and P2, as well as the influence of ectonucleotidase enzymes (CD39 and CD73) on tumor progression. A comprehensive bibliographic search was carried out. The relevant articles for this review were found in the PubMed, Scielo, Lilacs, and ScienceDirect databases. It was observed that among the P1 receptors, the A1, A2A, and A2B receptors are involved in the proliferation and invasion of BC, while the A3 receptor is related to the inhibition of tumor growth. Among the P2 receptors, the P2X7 has a dual function. When activated for a short time, it promotes metastasis, but when activated for long periods, it is related to BC cell death. P2Y2 and P2Y6 receptors are related to BC proliferation and invasiveness. Also, the high expression of CD39 and CD73 in BC is strongly related to a worse prognosis. The receptors and ectonucleotidases involved with BC become possible therapeutic targets. Several purinergic pathways have been found to be involved in BC cell survival and progression. In this review, in addition to analyzing the pathways involved, we reviewed the therapeutic interventions already studied for BC related to the purinergic system, as well as to other possible therapeutic targets.
Collapse
|
22
|
Sharma S, Kalra H, Akundi RS. Extracellular ATP Mediates Cancer Cell Migration and Invasion Through Increased Expression of Cyclooxygenase 2. Front Pharmacol 2021; 11:617211. [PMID: 33584298 PMCID: PMC7873692 DOI: 10.3389/fphar.2020.617211] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment plays a major role in the ability of the tumor cells to undergo metastasis. A major player of tumors gaining metastatic property is the inflammatory protein, cyclooxygenase 2 (COX-2). Several tumors show upregulation of this protein, which has been implicated in mediating metastasis in various cancer types such as of colon, breast and lung. In this report, we show that the concentration of extracellular ATP (eATP) is increased in response to cell death mediated by chemotherapeutic agents such as doxorubicin. By using three different cell-lines-HeLa (cervical), IMR-32 (neuronal) and MCF-7 (breast)-we show that this eATP goes on to act on purinergic (P2) receptors. Among the various P2 receptors expressed in these cells we identified P2X7, in IMR-32 and MCF-7 cells, and P2Y12, in HeLa cells, as important in modulating cell migration and invasion. Downstream of the P2 receptor activation, both p42/44 mitogen-activated protein kinase (MAPK) and the p38 MAPK are activated in these cells. These result in an increase in the expression of COX-2 mRNA and protein. We also observe an increase in the activity of matrix metalloproteinase 2 (MMP-2) enzyme in these cells. Blocking the P2 receptors not only blocks migration and invasion, but also COX-2 synthesis and MMP-2 activity. Our results show the link between purinergic receptors and COX-2 expression. Increased levels of ATP in the tumor microenvironment, therefore, leads to increased COX-2 expression, which, in turn, affords migratory and invasive properties to the tumor. This provides P2 receptor-based anti-inflammatory drugs (PBAIDs) a potential opportunity to be explored as cancer therapeutics.
Collapse
Affiliation(s)
- Shilpa Sharma
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Harshit Kalra
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Ravi Shankar Akundi
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| |
Collapse
|
23
|
Shishido K, Kuroishi T, Sugawara S. P2 purinergic receptor signaling and interleukin-1 synergistically induce interleukin-6 production in a human oral squamous carcinoma cell line. J Oral Biosci 2021; 63:80-90. [PMID: 33497843 DOI: 10.1016/j.job.2021.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the inflammatory roles of P2 purinergic receptor (P2R) signaling in oral squamous cell carcinoma (OSCC). METHODS Human OSCC cell lines HSC-2, Ca9-22, and HO-1-u-1 were stimulated with P2R agonists. The concentration of interleukin (IL)-6 in culture supernatants was measured using an enzyme-linked immune sorbent assay. Expression levels of messenger RNAs (mRNAs) were analyzed using reverse transcription polymerase chain reaction. Phosphorylation of intracellular signaling molecules was analyzed using western blotting. RESULTS HSC-2 cells expressed the mRNAs for P2X4-6 and all P2YRs. ATP or ADP induced significantly greater production of IL-6 by HSC-2 cells. Ca9-22 cells expressed mRNAs for P2X4-6 and all P2YRs except P2Y4. ATP or ADP induced the production of IL-6 by Ca9-22 cells, but the IL-6 concentration was much lower than that in HSC-2 cells. Although HO-1-u-1 cells expressed the mRNAs for P2X4-6 and all P2YRs, ATP or ADP did not induce IL-6 production. The production of IL-6 by HSC-2 cells stimulated with adenine nucleotides was significantly inhibited by P2R antagonists and a p38 mitogen-activated protein kinase inhibitor, but not by extracellular signal-related kinase or c-Jun N-terminal kinase inhibitors. The proinflammatory cytokine IL-1 significantly augmented P2R-induced IL-6 production by HSC-2 cells via the nuclear factor-κB signaling pathway. CONCLUSIONS The present study suggests that P2Rs signaling and IL-1 synergistically induce chronic inflammation in OSCC. Because chronic inflammation is a well-known driving force of tumor progression, these results support therapeutic strategies that target P2Rs signaling in OSCC.
Collapse
Affiliation(s)
- Kaori Shishido
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan; Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Toshinobu Kuroishi
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan.
| | - Shunji Sugawara
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
24
|
Purinergic Signaling Within the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1270:73-87. [PMID: 33123994 DOI: 10.1007/978-3-030-47189-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Accumulating studies have clearly demonstrated high concentrations of extracellular ATP (eATP) within the tumor microenvironment (TME). Implications of these findings are multifold as ATP-mediated purinergic signaling has been shown to mediate a variety of cancer-related processes, including cell migration, resistance to cytotoxic therapy, and immune regulation. Broad roles of ATP within the tumor microenvironment are linked to the abundance of ATP-regulated purinergic receptors on cancer and stromal and various immune cell types, as well as on the importance of ATP release and signaling in the regulation of multiple cellular processes. ATP release and downstream purinergic signaling are emerging as a central regulator of tumor growth and an important target for therapeutic intervention. In this chapter, we summarize the major roles of purinergic signaling in the tumor microenvironment with a specific focus on its critical roles in the induction of immunogenic cancer cell death and immune modulation.
Collapse
|
25
|
Woods LT, Forti KM, Shanbhag VC, Camden JM, Weisman GA. P2Y receptors for extracellular nucleotides: Contributions to cancer progression and therapeutic implications. Biochem Pharmacol 2021; 187:114406. [PMID: 33412103 DOI: 10.1016/j.bcp.2021.114406] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022]
Abstract
Purinergic receptors for extracellular nucleotides and nucleosides contribute to a vast array of cellular and tissue functions, including cell proliferation, intracellular and transmembrane ion flux, immunomodulation and thrombosis. In mammals, the purinergic receptor system is composed of G protein-coupled P1 receptors A1, A2A, A2B and A3 for extracellular adenosine, P2X1-7 receptors that are ATP-gated ion channels and G protein-coupled P2Y1,2,4,6,11,12,13 and 14 receptors for extracellular ATP, ADP, UTP, UDP and/or UDP-glucose. Recent studies have implicated specific P2Y receptor subtypes in numerous oncogenic processes, including cancer tumorigenesis, metastasis and chemotherapeutic drug resistance, where G protein-mediated signaling cascades modulate intracellular ion concentrations and activate downstream protein kinases, Src family kinases as well as numerous mitogen-activated protein kinases. We are honored to contribute to this special issue dedicated to the founder of the field of purinergic signaling, Dr. Geoffrey Burnstock, by reviewing the diverse roles of P2Y receptors in the initiation, progression and metastasis of specific cancers with an emphasis on pharmacological and genetic strategies employed to delineate cell-specific and P2Y receptor subtype-specific responses that have been investigated using in vitro and in vivo cancer models. We further highlight bioinformatic and empirical evidence on P2Y receptor expression in human clinical specimens and cover clinical perspectives where P2Y receptor-targeting interventions may have therapeutic relevance to cancer treatment.
Collapse
Affiliation(s)
- Lucas T Woods
- Department of Biochemistry, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Kevin Muñoz Forti
- Department of Biochemistry, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Vinit C Shanbhag
- Department of Biochemistry, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Jean M Camden
- Department of Biochemistry, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Gary A Weisman
- Department of Biochemistry, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
26
|
Ding Z, Xiang X, Li J, Wu S. Long-term 1800MHz electromagnetic radiation did not induce Balb/c-3T3 cells malignant transformation. Electromagn Biol Med 2021; 40:169-178. [PMID: 33211539 DOI: 10.1080/15368378.2020.1846194] [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: 07/28/2020] [Accepted: 11/01/2020] [Indexed: 10/23/2022]
Abstract
There is an increased public concern about potential health hazards of exposure to electromagnetic radiation (EMR). To declare the carcinogenic effects of 1800 MHz EMR. In this study, Balb/c-3T3 cells were exposed to 1800 MHz EMR for 80 days. The cells were harvested for cell proliferation detection, cell cycle assay, plate clone, and soft agar formation assay, transwell assay, and mRNA microarray detection. 1800 MHz EMR promoted Balb/c-3T3 proliferation. No clones were observed in both plate clone and soft agar clone formation assay. The percentage of cells in S phase in Balb/c-3T3 cells of 80d Expo was obviously higher than the percetage in 80d Sham cells. 80d Expo Balb/c-3T3 cells had stronger migration ability than Sham cells. The mRNA microarray results indicated that cell cycle, cell division, and DNA replication were the main biological processes the significant genes enriched, with higher expression of RPs and Mcms. 1800 MHz EMR promoted Balb/c-3T3 cells proliferation and migration. The mRNA microarray results indicated that cell cycle, cell division, and DNA replication were the main biological processes the significant genes enriched.
Collapse
Affiliation(s)
- Zhen Ding
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Shenzhen, China
| | - Xiaoyong Xiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Shenzhen, China
| | - Jintao Li
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Life Science and Bio-Engineering, Beijing University of Technology , Beijing, China
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing, China
| | - Shuicai Wu
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Life Science and Bio-Engineering, Beijing University of Technology , Beijing, China
| |
Collapse
|
27
|
Di Virgilio F, Vultaggio-Poma V, Sarti AC. P2X receptors in cancer growth and progression. Biochem Pharmacol 2020; 187:114350. [PMID: 33253643 DOI: 10.1016/j.bcp.2020.114350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
It is increasingly appreciated that ion channels have a crucial role in tumors, either as promoters of cancer cell growth, or modulators of immune cell functions, or both. Among ion channels, P2X receptors have a special status because they are gated by ATP, a common and abundant component of the tumor microenvironment. Furthermore, one P2X receptor, i.e. P2X7, may also function as a conduit for ATP release, thus fuelling the increased extracellular ATP level in the tumor interstitium. These findings show that P2X receptors and extracellular ATP are indissoluble partners and key regulators of tumor growth, and suggest the exploitation of the extracellular ATP-P2X partnership to develop innovative therapeutic approaches to cancer.
Collapse
|
28
|
Sankhe R, Pai SRK, Kishore A. Tumour suppression through modulation of neprilysin signaling: A comprehensive review. Eur J Pharmacol 2020; 891:173727. [PMID: 33160935 DOI: 10.1016/j.ejphar.2020.173727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/31/2020] [Accepted: 11/01/2020] [Indexed: 02/09/2023]
Abstract
Peptidases are emerging as promising drug targets in tumour suppression. Neprilysin, also known as neutral endopeptidase, is a cell surface peptidase that degrades various peptides such as angiotensin II, endothelin I, Substance P, etc., and reduces their local concentration. Neprilysin is expressed in various tissues such as kidney, prostate, lung, breast, brain, intestine, adrenal gland, etc. The tumour-suppressor mechanisms of neprilysin include its peptidase activity that degrades mitogenic growth factors such as fibroblast growth factor-2 and insulin-like growth factors, and the protein-protein interaction of neprilysin with phosphatase and tensin homolog, focal adhesion kinase, ezrin/radixin/moesin, and phosphoinositide 3-kinase. Studies have shown that the levels of neprilysin play an important role in malignancies. NEP is downregulated in prostate, renal, lung, breast, urothelial, cervical, hepatic cancers, etc. Histone deacetylation and hypermethylation of the neprilysin promoter region are the common mechanisms involved in the downregulation of neprilysin. Downregulation of the peptidase promotes angiogenesis, cell survival and cell migration. This review presents an overview of the role of neprilysin in malignancy, the tumour suppression mechanisms of neprilysin, the epigenetic mechanisms responsible for downregulation of neprilysin, and the potential pharmacological approaches to upregulate neprilysin levels and its activity.
Collapse
Affiliation(s)
- Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
29
|
Zhang WJ, Luo C, Pu FQ, Zhu JF, Zhu Z. The role and pharmacological characteristics of ATP-gated ionotropic receptor P2X in cancer pain. Pharmacol Res 2020; 161:105106. [DOI: 10.1016/j.phrs.2020.105106] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
|
30
|
Zaparte A, Cappellari AR, Brandão CA, de Souza JB, Borges TJ, Kist LW, Bogo MR, Zerbini LF, Ribeiro Pinto LF, Glaser T, Gonçalves MCB, Naaldijk Y, Ulrich H, Morrone FB. P2Y 2 receptor activation promotes esophageal cancer cells proliferation via ERK1/2 pathway. Eur J Pharmacol 2020; 891:173687. [PMID: 33130276 DOI: 10.1016/j.ejphar.2020.173687] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 02/08/2023]
Abstract
Esophageal cancer is a prominent worldwide illness that is divided into two main subtypes: esophageal squamous cell carcinoma and esophageal adenocarcinoma. Mortality rates are alarming, and the understanding of the mechanisms involved in esophageal cancer development, becomes essential. Purinergic signaling is related to many diseases and among these various types of tumors. Here we studied the effects of the P2Y2 receptor activation in different types of esophageal cancer. Esophageal tissue samples of healthy controls were used for P2Y2R expression quantification. Two human esophageal cancer cell lines Kyse-450 (squamous cell carcinoma) and OE-33 (adenocarcinoma) were used to perform in vitro analysis of cell proliferation, migration, adhesion, and the signaling pathways involved in P2Y2R activation. Data showed that P2Y2R was expressed in biopsies of patients with ESCC and adenocarcinoma, as well as in the two human esophageal cancer cell lines studied. The RT-qPCR analysis demonstrated that OE-33 cells have higher P2RY2 expression than Kyse-450 squamous cell line. Results showed that P2Y2R activation, induced by ATP or UTP, promoted esophageal cancer cells proliferation and colony formation. P2Y2R blockage with the selective antagonist, AR-C 118925XX, led to decreased proliferation, colony formation and adhesion. Treatments with ATP or UTP activated ERK 1/2 pathway in ESCC and ECA cells. The P2Y2R antagonism did not alter the migration of esophageal cancer cells. Interestingly, the esophageal cancer cell lines presented a distinct profile of nucleotide hydrolysis activity. The modulation of P2Y2 receptors may be a promising target for esophageal cancer treatment.
Collapse
Affiliation(s)
- Aline Zaparte
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Avenida Ipiranga, 6690. Partenon, 90619-900, Porto Alegre, RS, Brazil; Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil
| | - Angélica R Cappellari
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil
| | - Caroline A Brandão
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil
| | - Júlia B de Souza
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil
| | - Thiago J Borges
- Transplant Research Center, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Luíza W Kist
- Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil
| | - Maurício R Bogo
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Avenida Ipiranga, 6690. Partenon, 90619-900, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil
| | - Luiz F Zerbini
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cancer Genomics Group, Cape Town, South Africa
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Coordenação de Pesquisa, Instituto Nacional de Cancer, Rua Andre Cavalcante, 37, Centro, Rio de Janeiro, RJ, Brazil
| | - Talita Glaser
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Prof. Lineu Prestes, 748. Butantã, 05508-000, São Paulo, SP, Brazil
| | - Maria Carolina B Gonçalves
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Prof. Lineu Prestes, 748. Butantã, 05508-000, São Paulo, SP, Brazil
| | - Yahaira Naaldijk
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Prof. Lineu Prestes, 748. Butantã, 05508-000, São Paulo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Prof. Lineu Prestes, 748. Butantã, 05508-000, São Paulo, SP, Brazil
| | - Fernanda B Morrone
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, PUCRS, Avenida Ipiranga, 6690. Partenon, 90619-900, Porto Alegre, RS, Brazil; Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, PUCRS, Avenida Ipiranga, 6681, Partenon, 90619-900, Porto Alegre, RS, Brazil.
| |
Collapse
|
31
|
da Silva Ferreira NC, Alves LA, Soares-Bezerra RJ. Potential Therapeutic Applications of P2 Receptor Antagonists: From Bench to Clinical Trials. Curr Drug Targets 2020; 20:919-937. [PMID: 30760187 DOI: 10.2174/1389450120666190213095923] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Extracellular purines and pyrimidines have important physiological functions in mammals. Purines and pyrimidines act on P1 and P2 purinergic receptors, which are widely expressed in the plasma membrane in various cell types. P2 receptors act as important therapeutic targets and are associated with several disorders, such as pain, neurodegeneration, cancer, inflammation, and thrombosis. However, the use of antagonists for P2 receptors in clinical therapy, with the exception of P2Y12, is a great challenge. Currently, many research groups and pharmaceutical companies are working on the development of specific antagonist molecules for each receptor subtype that could be used as new medicines to treat their respective disorders. OBJECTIVE The present review compiles some interesting findings on the application of P2 receptor antagonists in different in vitro and in vivo experimental models as well as the progress of advanced clinical trials with these compounds. CONCLUSION Despite all of the exciting results obtained on the bench, few antagonists of P2 receptors advanced to the clinical trials, and once they reach this stage, the effectiveness of the therapy is not guaranteed, as in the example of P2X7 antagonists. Despite this, P2Y12 receptor antagonists have a history of success and have been used in therapy for at least two decades to prevent thrombosis in patients at risk for myocardial infarctions. This breakthrough is the motivation for scientists to develop new drugs with antagonistic activity for the other P2 receptors; thus, in a matter of years, we will have an evolution in the field of purinergic therapy.
Collapse
Affiliation(s)
- Natiele C da Silva Ferreira
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040- 360, Brazil
| | - Luiz A Alves
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040- 360, Brazil
| | - Rômulo J Soares-Bezerra
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-360, Brazil
| |
Collapse
|
32
|
Tao Z, Zhao Y, Chen X. Role of methyltransferase-like enzyme 3 and methyltransferase-like enzyme 14 in urological cancers. PeerJ 2020; 8:e9589. [PMID: 32765970 PMCID: PMC7382367 DOI: 10.7717/peerj.9589] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
N6-methyladenosine (m6A) modifications can be found in eukaryotic messenger RNA (mRNA), long non-coding RNA (lncRNA), and microRNA (miRNA). Several studies have demonstrated a close relationship between m6A modifications and cancer cells. Methyltransferase-like enzyme 3 (METTL3) and methyltransferase-like enzyme 14 (METTL14) are two major enzymes involved in m6A modifications that play vital roles in various cancers. However, the roles and regulatory mechanisms of METTL3 and METTL14 in urological cancers are largely unknown. In this review, we summarize the current research results for METTL3 and METTL14 and identify potential pathways involving these enzymes in kidney, bladder, prostate, and testicular cancer. We found that METTL3 and METTL14 have different expression patterns in four types of urological cancers. METTL3 is highly expressed in bladder and prostate cancer and plays an oncogenic role on cancer cells; however, its expression and role are opposite in kidney cancer. METTL14 is expressed at low levels in kidney and bladder cancer, where it has a tumor suppressive role. Low METTL3 or METTL14 expression in cancer cells negatively regulates cell growth-related pathways (e.g., mTOR, EMT, and P2XR6) but positively regulates cell death-related pathways (e.g., P53, PTEN, and Notch1). When METTL3 is highly expressed, it positively regulates the NF-kB and SHH-GL1pathways but negatively regulates PTEN. These results suggest that although METTL3 and METTL14 have different expression levels and regulatory mechanisms in urological cancers, they control cancer cell fate via cell growth- and cell death-related pathways. These findings suggest that m6A modification may be a potential new therapeutic target in urological cancer.
Collapse
Affiliation(s)
- Zijia Tao
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiqiao Zhao
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaonan Chen
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
33
|
Purinergic Signaling in the Hallmarks of Cancer. Cells 2020; 9:cells9071612. [PMID: 32635260 PMCID: PMC7407645 DOI: 10.3390/cells9071612] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/29/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is a complex expression of an altered state of cellular differentiation associated with severe clinical repercussions. The effort to characterize this pathological entity to understand its underlying mechanisms and visualize potential therapeutic strategies has been constant. In this context, some cellular (enhanced duplication, immunological evasion), metabolic (aerobic glycolysis, failure in DNA repair mechanisms) and physiological (circadian disruption) parameters have been considered as cancer hallmarks. The list of these hallmarks has been growing in recent years, since it has been demonstrated that various physiological systems misfunction in well-characterized ways upon the onset and establishment of the carcinogenic process. This is the case with the purinergic system, a signaling pathway formed by nucleotides/nucleosides (mainly adenosine triphosphate (ATP), adenosine (ADO) and uridine triphosphate (UTP)) with their corresponding membrane receptors and defined transduction mechanisms. The dynamic equilibrium between ATP and ADO, which is accomplished by the presence and regulation of a set of ectonucleotidases, defines the pro-carcinogenic or anti-cancerous final outline in tumors and cancer cell lines. So far, the purinergic system has been recognized as a potential therapeutic target in cancerous and tumoral ailments.
Collapse
|
34
|
Woods LT, Jasmer KJ, Muñoz Forti K, Shanbhag VC, Camden JM, Erb L, Petris MJ, Weisman GA. P2Y 2 receptors mediate nucleotide-induced EGFR phosphorylation and stimulate proliferation and tumorigenesis of head and neck squamous cell carcinoma cell lines. Oral Oncol 2020; 109:104808. [PMID: 32540611 PMCID: PMC7736485 DOI: 10.1016/j.oraloncology.2020.104808] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/16/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To assess functional expression of the P2Y2 nucleotide receptor (P2Y2R) in head and neck squamous cell carcinoma (HNSCC) cell lines and define its role in nucleotide-induced epidermal growth factor receptor (EGFR) transactivation. The use of anti-EGFR therapeutics to treat HNSCC is hindered by intrinsic and acquired drug resistance. Defining novel pathways that modulate EGFR signaling could identify additional targets to treat HNSCC. MATERIALS AND METHODS In human HNSCC cell lines CAL27 and FaDu and the mouse oral cancer cell line MOC2, P2Y2R contributions to extracellular nucleotide-induced changes in intracellular free Ca2+ concentration and EGFR and extracellular signal-regulated kinase (ERK1/2) phosphorylation were determined using the ratiometric Ca2+ indicator fura-2 and immunoblot analysis, respectively. Genetic knockout of P2Y2Rs using CRISPR technology or pharmacological inhibition with P2Y2R-selective antagonist AR-C118925 defined P2Y2R contributions to in vivo tumor growth. RESULTS P2Y2R agonists UTP and ATP increased intracellular Ca2+ levels and ERK1/2 and EGFR phosphorylation in CAL27 and FaDu cells, responses that were inhibited by AR-C118925 or P2Y2R knockout. P2Y2R-mediated EGFR phosphorylation was also attenuated by inhibition of the adamalysin family of metalloproteases or Src family kinases. P2Y2R knockout reduced UTP-induced CAL27 cell proliferation in vitro and significantly reduced CAL27 and FaDu tumor xenograft volume in vivo. In a syngeneic mouse model of oral cancer, AR-C118925 administration reduced MOC2 tumor volume. CONCLUSION P2Y2Rs mediate HNSCC cell responses to extracellular nucleotides and genetic or pharmacological blockade of P2Y2R signaling attenuates tumor cell proliferation and tumorigenesis, suggesting that the P2Y2R represents a novel therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Lucas T Woods
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA
| | - Kimberly J Jasmer
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA
| | - Kevin Muñoz Forti
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA
| | - Vinit C Shanbhag
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA
| | - Jean M Camden
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA
| | - Laurie Erb
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA
| | - Michael J Petris
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA; Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65211-7310 USA
| | - Gary A Weisman
- Division of Biochemistry, University of Missouri, Columbia, MO 65211-7310 USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310 USA.
| |
Collapse
|
35
|
Lara R, Adinolfi E, Harwood CA, Philpott M, Barden JA, Di Virgilio F, McNulty S. P2X7 in Cancer: From Molecular Mechanisms to Therapeutics. Front Pharmacol 2020; 11:793. [PMID: 32581786 PMCID: PMC7287489 DOI: 10.3389/fphar.2020.00793] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022] Open
Abstract
P2X7 is a transmembrane receptor expressed in multiple cell types including neurons, dendritic cells, macrophages, monocytes, B and T cells where it can drive a wide range of physiological responses from pain transduction to immune response. Upon activation by its main ligand, extracellular ATP, P2X7 can form a nonselective channel for cations to enter the cell. Prolonged activation of P2X7, via high levels of extracellular ATP over an extended time period can lead to the formation of a macropore, leading to depolarization of the plasma membrane and ultimately to cell death. Thus, dependent on its activation state, P2X7 can either drive cell survival and proliferation, or induce cell death. In cancer, P2X7 has been shown to have a broad range of functions, including playing key roles in the development and spread of tumor cells. It is therefore unsurprising that P2X7 has been reported to be upregulated in several malignancies. Critically, ATP is present at high extracellular concentrations in the tumor microenvironment (TME) compared to levels observed in normal tissues. These high levels of ATP should present a survival challenge for cancer cells, potentially leading to constitutive receptor activation, prolonged macropore formation and ultimately to cell death. Therefore, to deliver the proven advantages for P2X7 in driving tumor survival and metastatic potential, the P2X7 macropore must be tightly controlled while retaining other functions. Studies have shown that commonly expressed P2X7 splice variants, distinct SNPs and post-translational receptor modifications can impair the capacity of P2X7 to open the macropore. These receptor modifications and potentially others may ultimately protect cancer cells from the negative consequences associated with constitutive activation of P2X7. Significantly, the effects of both P2X7 agonists and antagonists in preclinical tumor models of cancer demonstrate the potential for agents modifying P2X7 function, to provide innovative cancer therapies. This review summarizes recent advances in understanding of the structure and functions of P2X7 and how these impact P2X7 roles in cancer progression. We also review potential therapeutic approaches directed against P2X7.
Collapse
Affiliation(s)
- Romain Lara
- Biosceptre (UK) Limited, Cambridge, United Kingdom
| | - Elena Adinolfi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mike Philpott
- Centre for Cutaneous Research, Blizard Institute, Bart's & The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
36
|
Zhang WJ, Hu CG, Zhu ZM, Luo HL. Effect of P2X7 receptor on tumorigenesis and its pharmacological properties. Biomed Pharmacother 2020; 125:109844. [PMID: 32004973 DOI: 10.1016/j.biopha.2020.109844] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/27/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
The occurrence and development of tumors is a multi-factor, multi-step, multi-gene pathological process, and its treatment has been the most difficult problem in the field of medicine today. Therefore, exploring the relevant factors involved in the pathogenesis of tumors, improving the diagnostic rate, treatment rate, and prognosis survival rate of tumors have become an urgent problem to be solved. A large number of studies have shown that the P2X7 receptor (P2X7R) and the tumor microenvironment play an important role in regulating the growth, apoptosis, migration and invasion of tumor cells. P2X7R is an ATP ligand-gated cationic channel receptor, which exists in most tissues of the human body. The main function of P2X7R is to regulate the relevant cells (such as macrophages, lymphocytes, and glial cells) to release damaging factors and induce apoptosis and cell death. In recent years, with continuous research and exploration of P2X7R, it has been found that P2X7R exists on the surface of most tumor cells and plays an important role in tumor pathogenesis. The activation of the P2X7R can open the ion channels on the tumor cell membrane (sodium ion, calcium ion influx and potassium ion outflow), trigger rearrangement of the cytoskeleton and changes in membrane fluidity, allow small molecule substances to enter the cell, activate enzymes and kinases in related signaling pathways in cells (such as PKA, PKC, ERK1/2, AKT, and JNK), thereby affecting the development of tumor cells, and can also indirectly affect the growth, apoptosis and migration of tumor cells through tumor microenvironment. At present, P2X7R has been widely recognized for its important role in tumorigenesis and development. In this paper, we give a comprehensive description of the structure and function of the P2X7R gene. We also clarified the concept of tumor microenvironment and its effect on tumors, discussed the relevant pathological mechanisms in the development of tumors, and revealed the intrinsic relationship between P2X7R and tumors. We explored the pharmacological properties of P2X7R antagonists or inhibitors in reducing its expression as targeted therapy for tumors.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, 343000, China
| | - Ce-Gui Hu
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, 343000, China
| | - Zheng-Ming Zhu
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, 343000, China
| | - Hong-Liang Luo
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, 343000, China.
| |
Collapse
|
37
|
Han S, Wei R, Zhang X, Jiang N, Fan M, Huang JH, Xie B, Zhang L, Miao W, Butler ACP, Coleman MA, Vaughan AT, Wang Y, Chen HW, Liu J, Li JJ. CPT1A/2-Mediated FAO Enhancement-A Metabolic Target in Radioresistant Breast Cancer. Front Oncol 2019; 9:1201. [PMID: 31803610 PMCID: PMC6873486 DOI: 10.3389/fonc.2019.01201] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
Tumor cells, including cancer stem cells (CSCs) resistant to radio- and chemotherapy, must enhance metabolism to meet the extra energy demands to repair and survive such genotoxic conditions. However, such stress-induced adaptive metabolic alterations, especially in cancer cells that survive radiotherapy, remain unresolved. In this study, we found that CPT1 (Carnitine palmitoyl transferase I) and CPT2 (Carnitine palmitoyl transferase II), a pair of rate-limiting enzymes for mitochondrial fatty acid transportation, play a critical role in increasing fatty acid oxidation (FAO) required for the cellular fuel demands in radioresistant breast cancer cells (RBCs) and radiation-derived breast cancer stem cells (RD-BCSCs). Enhanced CPT1A/CPT2 expression was detected in the recurrent human breast cancers and associated with a worse prognosis in breast cancer patients. Blocking FAO via a FAO inhibitor or by CRISPR-mediated CPT1A/CPT2 gene deficiency inhibited radiation-induced ERK activation and aggressive growth and radioresistance of RBCs and RD-BCSCs. These results revealed that switching to FAO contributes to radiation-induced mitochondrial energy metabolism, and CPT1A/CPT2 is a potential metabolic target in cancer radiotherapy.
Collapse
Affiliation(s)
- Shujun Han
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Center for Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ryan Wei
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Lewis Katz School of Medicine/St. Luke's University Regional Campus, Temple University, Philadelphia, PA, United States
| | - Xiaodi Zhang
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Nian Jiang
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Ming Fan
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Jie Hunter Huang
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Bowen Xie
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Lu Zhang
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Weili Miao
- Department of Chemistry, University of California, Riverside, Riverside, CA, United States
| | - Ashley Chen-Ping Butler
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Matthew A. Coleman
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
- NCI-Designated Compressive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Andrew T. Vaughan
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
- NCI-Designated Compressive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, Riverside, CA, United States
| | - Hong-Wu Chen
- NCI-Designated Compressive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, United States
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jian Jian Li
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
- NCI-Designated Compressive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
38
|
Wei L, Mousawi F, Li D, Roger S, Li J, Yang X, Jiang LH. Adenosine Triphosphate Release and P2 Receptor Signaling in Piezo1 Channel-Dependent Mechanoregulation. Front Pharmacol 2019; 10:1304. [PMID: 31780935 PMCID: PMC6853025 DOI: 10.3389/fphar.2019.01304] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Organs and tissues and their constituent cells are physiologically submitted to diverse types of mechanical forces or stress, one common sequence of which is release of intracellular ATP into extracellular space. Extracellular ATP is a well-established autocrine or paracrine signaling molecule that regulates multiple cell functions and mediates cell-to-cell communications via activating the purinergic P2 receptors, more specifically, ligand-gated ion channel P2X receptors and some of the G-protein-coupled P2Y receptors. The molecular mechanisms that sense mechanical and transduce forces to trigger ATP release are poorly understood. The Piezo1, a newly identified mechanosensing ion channel, shows widespread expression and confers mechanosensitivity in many different types of cells. In this mini-review, we briefly introduce the Piezo1 channel and discuss the evidence that supports its important role in the mechanoregulation of diverse cell functions and, more specifically, critical engagement of ATP release and subsequent P2 receptor activation in Piezo1 channel-dependent mechanoregulation. Such ATP release-mediated coupling of the Piezo1 channel and P2 receptors may serve a signaling mechanism that is more common than we currently understand in transducing mechanical information to regulation of the attendant cell functions in various organs and tissues.
Collapse
Affiliation(s)
- Linyu Wei
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Dongliang Li
- Department of Physiology, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Sébastien Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| | - Jing Li
- Lingnan Medical Research Centre, School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuebin Yang
- Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| |
Collapse
|
39
|
Chen S, Shenk T, Nogalski MT. P2Y2 purinergic receptor modulates virus yield, calcium homeostasis, and cell motility in human cytomegalovirus-infected cells. Proc Natl Acad Sci U S A 2019; 116:18971-18982. [PMID: 31481624 PMCID: PMC6754545 DOI: 10.1073/pnas.1907562116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (HCMV) manipulates many aspects of host cell biology to create an intracellular milieu optimally supportive of its replication and spread. Our study reveals that levels of several components of the purinergic signaling system, including the P2Y2 and P2X5 receptors, are elevated in HCMV-infected fibroblasts. Knockdown and drug treatment experiments demonstrated that P2Y2 enhances the yield of virus, whereas P2X5 reduces HCMV production. The HCMV IE1 protein induces P2Y2 expression; and P2Y2-mediated signaling is important for efficient HCMV gene expression, DNA synthesis, and the production of infectious HCMV progeny. P2Y2 cooperates with the viral UL37x1 protein to regulate cystolic Ca2+ levels. P2Y2 also regulates PI3K/Akt signaling and infected cell motility. Thus, P2Y2 functions at multiple points within the viral replication cycle to support the efficient production of HCMV progeny, and it may facilitate in vivo viral spread through its role in cell migration.
Collapse
Affiliation(s)
- Saisai Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014
| | - Thomas Shenk
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014
| | - Maciej T Nogalski
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014
| |
Collapse
|
40
|
Signaling pathways involved in adaptive responses to cell membrane disruption. CURRENT TOPICS IN MEMBRANES 2019; 84:99-127. [PMID: 31610867 DOI: 10.1016/bs.ctm.2019.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Plasma membrane disruption occurs frequently in many animal tissues. Cell membrane disruption induces not only a rapid and massive influx of Ca2+ into the cytosol but also an efflux or release of various signaling molecules, such as ATP, from the cytosol; in turn, these signaling molecules stimulate a variety of pathways in both wounded and non-wounded neighboring cells. These signals first trigger cell membrane repair responses in the wounded cell but then induce an adaptive response, which results in faster membrane repair in the event of future wounds in both wounded and non-wounded neighboring cells. In addition, signaling pathways stimulated by membrane disruption induce other adaptive responses, including cell survival, regeneration, migration, and proliferation. This chapter summarizes the role of intra- and intercellular signaling pathways in adaptive responses triggered by cell membrane disruption.
Collapse
|
41
|
Hevia MJ, Castro P, Pinto K, Reyna-Jeldes M, Rodríguez-Tirado F, Robles-Planells C, Ramírez-Rivera S, Madariaga JA, Gutierrez F, López J, Barra M, De la Fuente-Ortega E, Bernal G, Coddou C. Differential Effects of Purinergic Signaling in Gastric Cancer-Derived Cells Through P2Y and P2X Receptors. Front Pharmacol 2019; 10:612. [PMID: 31249523 PMCID: PMC6584115 DOI: 10.3389/fphar.2019.00612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/15/2019] [Indexed: 01/04/2023] Open
Abstract
Gastric cancer (GC) is the one of the most prevalent cancers and one of the leading causes of cancer-induced deaths. Previously, we found that the expression of purinergic P2Y2 receptor (P2Y2R) is increased in GC samples as compared to adjacent healthy mucosa taken from GC-diagnosed patients. In this work, we studied in detail purinergic signaling in the gastric adenocarcinoma-derived cell lines: AGS, MKN-45, and MKN-74, and compared them to a nontumoral epithelial cell line: GES-1. In GC-derived cells, we detected the expression of several purinergic receptors, and found important differences as compared to GES-1 cells. Functional studies revealed a strong contribution of P2Y2Rs in intracellular calcium increases, elicited by adenosine-triphosphate (ATP), uridine-triphosphate (UTP), and the P2Y2R agonist MRS2768. Responses were preserved in the absence of extracellular calcium and inhibited by P2Y2R antagonists. In GES-1 cells, ATP and UTP induced similar responses and the combination of P2X and P2Y receptor antagonists was able to block them. Proliferation studies showed that ATP regulates AGS and MKN-74 cells in a biphasic manner, increasing cell proliferation at 10–100 μM, but inhibiting at 300 μM ATP. On the other hand, 1–300 μM UTP, a P2Y2R agonist, increased concentration-dependent cell proliferation. The effects of UTP and ATP were prevented by both wide-range and specific purinergic antagonists. In contrast, in GES-1 cells ATP only decreased cell proliferation in a concentration-dependent manner, and UTP had no effect. Notably, the isolated application of purinergic antagonists was sufficient to change the basal proliferation of AGS cells, indicating that nucleotides released by the cells can act as paracrine/autocrine signals. Finally, in tumor-derived biopsies, we found an increase of P2Y2R and a decrease in P2X4R expression; however, we found high variability between seven different biopsies and their respective adjacent healthy gastric mucosa. Even so, we found a correlation between the expression levels of P2Y2R and P2X4R and survival rates of GC patients. Taken together, these results demonstrate the involvement of different purinergic receptors and signaling in GC, and the pattern of expression changes in tumoral cells, and this change likely directs ATP and nucleotide signaling from antiproliferative effects in healthy tissues to proliferative effects in cancer.
Collapse
Affiliation(s)
- María José Hevia
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Patricio Castro
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Katherine Pinto
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Mauricio Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | | | | | - Sebastián Ramírez-Rivera
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Juan Andrés Madariaga
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Hospital San Pablo, Coquimbo, Chile
| | | | - Javier López
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Hospital San Pablo, Coquimbo, Chile
| | - Marcelo Barra
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.,Hospital San Pablo, Coquimbo, Chile
| | - Erwin De la Fuente-Ortega
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Giuliano Bernal
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Claudio Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| |
Collapse
|
42
|
Gong D, Zhang J, Chen Y, Xu Y, Ma J, Hu G, Huang Y, Zheng J, Zhai W, Xue W. The m 6A-suppressed P2RX6 activation promotes renal cancer cells migration and invasion through ATP-induced Ca 2+ influx modulating ERK1/2 phosphorylation and MMP9 signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:233. [PMID: 31159832 PMCID: PMC6547495 DOI: 10.1186/s13046-019-1223-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/09/2019] [Indexed: 12/24/2022]
Abstract
Background Previous study demonstrated that extracellular ATP could promote cell migration and invasion in multiple human cancers. Till now, the pro-invasive mechanisms of ATP and P2RX6, a preferred receptor for ATP, are still poorly studied in RCC. Methods Bioinformatics analysis was performed to identify the differentially expressed genes during RCC different stages. Tissue microarray, IHC staining and survival analysis was respectively used to evaluate potential clinical function. In vitro and in vivo assays were performed to explore the P2RX6 biological effects in RCC progression. Results We found that ATP might increase RCC cells migration and invasion through P2RX6. Mechanism dissection revealed that ATP-P2RX6 might modulate the Ca2+-mediated p-ERK1/2/MMP9 signaling to increase the RCC cells migration and invasion. Furthermore, METTL14 implicated m6A modification in RCC and down-regulated P2RX6 protein translation. In addition, human clinical survey also indicated the positive correlation of this newly identified signaling in RCC progression and prognosis. Conclusions Our findings revealed that the newly identified ATP-P2RX6-Ca2+-p-ERK1/2-MMP9 signaling facilitates RCC cell invasion and metastasis. Targeting this novel signaling pathway with small molecules might help us to develop a new approach to better suppress RCC progression. Electronic supplementary material The online version of this article (10.1186/s13046-019-1223-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dongkui Gong
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yunfei Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China
| | - Junjie Ma
- Department of Urology, Pudong Hospital, School of Medicine in Fudan University, Shanghai, 201300, China
| | - Guanghui Hu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Junhua Zheng
- Department of Urology, Shanghai First People's Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
43
|
Fan F, Yang M, Geng X, Ma X, Sun H. Effects of Restraint Water-Immersion Stress-Induced Gastric Mucosal Damage on Astrocytes and Neurons in the Nucleus Raphe Magnus of Rats via the ERK1/2 Signaling Pathway. Neurochem Res 2019; 44:1841-1850. [PMID: 31119435 DOI: 10.1007/s11064-019-02818-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/03/2019] [Accepted: 05/20/2019] [Indexed: 01/31/2023]
Abstract
Restraint water-immersion stress (RWIS) consists of psychological and physical stimulation, and it has been utilized in the research of gastric mucosal damage. It has been shown by previous studies that the nucleus raphe magnus (NRM) is closely involved in the gastrointestinal function, but its functions on the stress-induced gastric mucosal injury (SGMI) have not been thoroughly elucidated to date. Consequently, in this research, we aim to measure the expression of astrocytic glial fibrillary acidic protein (GFAP), neuronal c-Fos, and phosphorylation extracellular signal regulated kinase 1/2 (p-ERK1/2) in the process of RWIS with immunohistochemistry and western blot methods. What is more, we detect the relation between astrocytes and neurons throughout the stress procedure and explore the regulation of the ERK1/2 signaling pathway on the activity of astrocytes and neurons after RWIS. The results indicated that all three proteins expression multiplied following peaked 3 h substantially. The SMGI, astrocyte and neuron activity were affected after the astrocytotoxin L-A-aminohexanedioic acid (L-AA) and c-fos antisense oligonucleotide (ASO) injections. After the injection of PD98059, the gastric mucosal injury, astrocyte and neuron activity significantly fell off. These results suggested that RWIS-induced activity of astrocytes and neurons in the NRM may play a significant part in gastric mucosa damage via the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Fangcheng Fan
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, 250014, China
| | - Mengzhu Yang
- Qingdao No. 31 Middle School, Qingdao, 266041, China
| | - Xiwen Geng
- Shandong Traditional Chinese Medicine University, Jinan, 250014, China
| | - Xiaoli Ma
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, China.
| | - Haiji Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
44
|
Di Virgilio F, Sarti AC, Falzoni S, De Marchi E, Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer 2018; 18:601-618. [PMID: 30006588 DOI: 10.1038/s41568-018-0037-0] [Citation(s) in RCA: 470] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Modulation of the biochemical composition of the tumour microenvironment is a new frontier of cancer therapy. Several immunosuppressive mechanisms operate in the milieu of most tumours, a condition that makes antitumour immunity ineffective. One of the most potent immunosuppressive factors is adenosine, which is generated in the tumour microenvironment owing to degradation of extracellular ATP. Accruing evidence over the past few years shows that ATP is one of the major biochemical constituents of the tumour microenvironment, where it acts at P2 purinergic receptors expressed on both tumour and host cells. Stimulation of P2 receptors has different effects depending on the extracellular ATP concentration, the P2 receptor subtype engaged and the target cell type. Among P2 receptors, the P2X purinergic receptor 7 (P2X7R) subtype appears to be a main player in host-tumour cell interactions. Preclinical studies in several tumour models have shown that P2X7R targeting is potentially a very effective anticancer treatment, and many pharmaceutical companies have now developed potent and selective small molecule inhibitors of P2X7R. In this Review, we report on the multiple mechanisms by which extracellular ATP shapes the tumour microenvironment and how its stimulation of host and tumour cell P2 receptors contributes to determining tumour fate.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Adinolfi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
45
|
Gendron FP, Placet M, Arguin G. P2Y 2 Receptor Functions in Cancer: A Perspective in the Context of Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1051:91-106. [PMID: 28815512 DOI: 10.1007/5584_2017_90] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Purinergic signaling has recently emerged as a network of signaling molecules, enzymes and receptors that coordinates the action and behavior of cancerous cells. Extracellular adenosine 5' triphosphate activates a plethora of P2 nucleotide receptors that can putatively modulate cancer cell proliferation, survival and dissemination. In this context, the G protein-coupled P2Y2 receptor was identified as one of the entities coordinating the cellular and molecular events that characterize cancerous cells. In this chapter, we will look at the contribution of the P2Y2 receptor in cancer outcomes and use this information to demonstrate that the P2Y2 receptor represents a drug target of interest in the setting of colorectal cancer, for which the role and function of this receptor is poorly defined. More particularly, we will review how the P2Y2 receptor modulates cancer cell proliferation and survival, while promoting cell dissemination and formation of metastases. Finally, we will investigate how the P2Y2 receptor can contribute to the detrimental development of drug resistance that is often observed in cancerous cells.
Collapse
Affiliation(s)
- Fernand-Pierre Gendron
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Pavillon de Recherche Appliquée sur le Cancer, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Morgane Placet
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Pavillon de Recherche Appliquée sur le Cancer, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Guillaume Arguin
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Pavillon de Recherche Appliquée sur le Cancer, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
46
|
Khalid M, Brisson L, Tariq M, Hao Y, Guibon R, Fromont G, Mortadza SAS, Mousawi F, Manzoor S, Roger S, Jiang LH. Carcinoma-specific expression of P2Y11 receptor and its contribution in ATP-induced purinergic signalling and cell migration in human hepatocellular carcinoma cells. Oncotarget 2018; 8:37278-37290. [PMID: 28418839 PMCID: PMC5514908 DOI: 10.18632/oncotarget.16191] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 02/15/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular ATP-induced Ca2+ signalling is critical in regulating diverse physiological and disease processes. Emerging evidence suggests high concentrations of extracellular ATP in tumour tissues. In this study, we examined the P2 receptor for ATP-induced Ca2+ signalling in human hepatocellular carcinoma (HCC) cells. Fura-2-based measurements of the intracellular Ca2+ concentration ([Ca2+]i) showed that extracellular ATP induced an increase in the [Ca2+]i in human HCC Huh-7 and HepG2 cells. NF546, a P2Y11 receptor agonist was equally effective in inducing an increase in the [Ca2+]i. In contrast, agonists for the P2X receptors (αβmeATP and BzATP), P2Y1 receptor (MRS2365) or P2Y2 receptor (MRS2768) were ineffective. In addition, ATP/NF546-induced increases in the [Ca2+]i were strongly inhibited by treatment with NF340, a P2Y11 receptor antagonist. Immunofluorescent confocal imaging and western blotting analysis consistently demonstrated the P2Y11 receptor expression in Huh-7 and HepG2 cells. Transfection with P2Y11-specific siRNA attenuated the P2Y11 receptor protein expression level and also reduced NF546-induced increase in the [Ca2+]i. Importantly, immunohistochemistry revealed that the P2Y11 receptor was expressed at very high level in human HCC tissues and, by contrast, it was barely detected in normal liver tissues. Trans-well cell migration assay demonstrated that ATP and NF546 induced concentration-dependent stimulation of Huh-7 cell migration. Treatment with NF340 prevented ATP-induced stimulation of cell migration. Taken together, our results show carcinoma-specific expression of the P2Y11 receptor and its critical role in mediating ATP-inducing Ca2+ signalling and regulating cell migration in human HCC cells.
Collapse
Affiliation(s)
- Madiha Khalid
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.,Atta-ur-Rahman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
| | - Lucie Brisson
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, Tours, France
| | - Menahil Tariq
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Yunjie Hao
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | | | | | | | - Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Sobia Manzoor
- Atta-ur-Rahman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
| | - Sébastien Roger
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, Tours, France.,Institut Universitaire de France, Paris Cedex 05, France
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.,Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours, Tours, France.,Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, P. R. China.,Sino-UK Joint Laboratory of Brain Function and Injury, Xinxiang Medical University, Xinxiang, P. R. China
| |
Collapse
|
47
|
Qiu Y, Liu Y, Li WH, Zhang HQ, Tian XX, Fang WG. P2Y2 receptor promotes the migration and invasion of breast cancer cells via EMT-related genes Snail and E-cadherin. Oncol Rep 2018; 39:138-150. [PMID: 29115551 PMCID: PMC5783596 DOI: 10.3892/or.2017.6081] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Adenosine 5'-triphosphate (ATP) is one of the most abundant biochemical constituents within the tumor microenvironment and is postulated to play critical roles in the progression of a number of types of tumors via interaction with the P2Y2 receptor. In the present study, we demonstrated that the P2Y2 receptor was highly expressed in MCF7 and Hs578T breast cancer cells. Downregulation of the P2Y2 receptor by small interfering RNA (siRNA) significantly attenuated ATP- or UTP-driven migration and invasion of the breast cancer cells as well as expression of EMT-related genes Snail and E-cadherin. Consistent with the observations in vitro, the P2Y2 receptor was found to be abundantly expressed at the invasive edge of the tumor, in infiltrating tumor cells in breast adipose tissues and/or the cancer embolus in the lymphatic sinuses compared with the tumor core areas. Furthermore, high Snail expression and weak or negative expression of E-cadherin were observed at the invasive edge of tumors. Taken together, these data indicate that the P2Y2 receptor promoted cell migration and invasion in breast cancer cells via EMT-related genes Snail and E-cadherin.
Collapse
Affiliation(s)
- Ying Qiu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
- Department of Pathology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yan Liu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Wei-Hua Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Hong-Quan Zhang
- Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Xin-Xia Tian
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Wei-Gang Fang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| |
Collapse
|
48
|
|
49
|
Dou L, Chen YF, Cowan PJ, Chen XP. Extracellular ATP signaling and clinical relevance. Clin Immunol 2017; 188:67-73. [PMID: 29274390 DOI: 10.1016/j.clim.2017.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022]
Abstract
Since purinergic signaling was discovered in the early 1970s, it has been shown that extracellular nucleotides, and their derivative nucleosides, are released in a regulated or unregulated manner by cells in various challenging settings and then bind defined purinergic receptors to activate intricate signaling networks. Extracellular ATP plays a role based on different P2 receptor subtypes expressed on specific cell types. Sequential hydrolysis of extracellular ATP catalyzed by ectonucleotidases (e.g. CD39, CD73) is the main pathway for the generation of adenosine, which in turn activates P1 receptors. Many studies have demonstrated that extracellular ATP signaling functions as an important dynamic regulatory pathway to coordinate appropriate immune responses in various pathological processes, including intracellular infection, host-tumor interaction, pro-inflammation vascular injury, and transplant immunity. ATP receptors and CD39 also participate in related clinical settings. Here, we review the latest research in to the development of promising clinical treatment strategies.
Collapse
Affiliation(s)
- Lei Dou
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Fa Chen
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peter J Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Australia.
| | - Xiao-Ping Chen
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
de Andrade Mello P, Coutinho-Silva R, Savio LEB. Multifaceted Effects of Extracellular Adenosine Triphosphate and Adenosine in the Tumor-Host Interaction and Therapeutic Perspectives. Front Immunol 2017; 8:1526. [PMID: 29184552 PMCID: PMC5694450 DOI: 10.3389/fimmu.2017.01526] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 10/27/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer is still one of the world's most pressing health-care challenges, leading to a high number of deaths worldwide. Immunotherapy is a new developing therapy that boosts patient's immune system to fight cancer by modifying tumor-immune cells interaction in the tumor microenvironment (TME). Extracellular adenosine triphosphate (eATP) and adenosine (Ado) are signaling molecules released in the TME that act as modulators of both immune and tumor cell responses. Extracellular adenosine triphosphate and Ado activate purinergic type 2 (P2) and type 1 (P1) receptors, respectively, triggering the so-called purinergic signaling. The concentration of eATP and Ado within the TME is tightly controlled by several cell-surface ectonucleotidases, such as CD39 and CD73, the major ecto-enzymes expressed in cancer cells, immune cells, stromal cells, and vasculature, being CD73 also expressed on tumor-associated fibroblasts. Once accumulated in the TME, eATP boosts antitumor immune response, while Ado attenuates or suppresses immunity against the tumor. In addition, both molecules can mediate growth stimulation or inhibition of the tumor, depending on the specific receptor activated. Therefore, purinergic signaling is able to modulate both tumor and immune cells behavior and, consequently, the tumor-host interaction and disease progression. In this review, we discuss the role of purinergic signaling in the host-tumor interaction detailing the multifaceted effects of eATP and Ado in the inflammatory TME. Moreover, we present recent findings into the application of purinergic-targeting therapy as a potential novel option to boost antitumor immune responses in cancer.
Collapse
Affiliation(s)
- Paola de Andrade Mello
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|