1
|
Dong J, Zhao W, Zhao J, Chen J, Liu P, Zheng X, Li D, Xue Y, Zhou H. ALPL regulates pro-angiogenic capacity of mesenchymal stem cells through ATP-P2X7 axis controlled exosomes secretion. J Nanobiotechnology 2024; 22:172. [PMID: 38609899 PMCID: PMC11015668 DOI: 10.1186/s12951-024-02396-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Early-onset bone dysplasia is a common manifestation of hypophosphatasia (HPP), an autosomal inherited disease caused by ALPL mutation. ALPL ablation induces prototypical premature bone ageing characteristics, resulting in impaired osteogenic differentiation capacity of human bone marrow mesenchymal stem cells (hBMMSCs). As angiogenesis is tightly coupled with osteogenesis, it also plays a necessary role in sustaining bone homeostasis. We have previously observed a decrease in expression of angiogenesis marker gene CD31 in the metaphysis of long bone in Alpl+/- mice. However, the role of ALPL in regulation of angiogenesis in bone has remained largely unknown. METHODS Exosomes derived from Normal and HPP hBMMSCs were isolated and identified by ultracentrifugation, transmission electron microscopy, and nanoparticle size measurement. The effects of ALPL on the angiogenic capacity of hBMMSCs from HPP patients were assessed by immunofluorescence, tube formation, wound healing and migration assay. exo-ELISA and Western Blot were used to evaluate the exosomes secretion of hBMMSCs from HPP, and the protein expression of VEGF, PDGFBB, Angiostatin and Endostatin in exosomes respectively. RESULTS We verified that ALPL ablation resulted in impaired pro-angiogenic capacity of hBMMSCs, accounting for reduced migration and tube formation of human umbilical vein endothelial cells, as the quantities and proteins composition of exosomes varied with ALPL expression. Mechanistically, loss of function of ALPL enhanced ATP release. Additional ATP, in turn, led to markedly elevated level of ATP receptor P2X7, which consequently promoted exosomes secretion, resulting in a decreased capacity to promote angiogenesis. Conversely, inhibition of P2X7 increased the angiogenic induction capacity by preventing excessive release of anti-angiogenic exosomes in ALPL deficient-hBMMSCs. CONCLUSION The ALPL-ATP axis regulates the pro-angiogenic ability of hBMMSCs by controlling exosomes secretion through the P2X7 receptor. Thus, P2X7 may be proved as an effective therapeutic target for accelerating neovascularization in ALPL-deficient bone defects.
Collapse
Affiliation(s)
- Jiayi Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Wanmin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jiangdong Zhao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, China
| | - Ji Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Ping Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xueni Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Dehua Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| | - Yang Xue
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| | - Hongzhi Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Acuña-Castillo C, Escobar A, García-Gómez M, Bachelet VC, Huidobro-Toro JP, Sauma D, Barrera-Avalos C. P2X7 Receptor in Dendritic Cells and Macrophages: Implications in Antigen Presentation and T Lymphocyte Activation. Int J Mol Sci 2024; 25:2495. [PMID: 38473744 DOI: 10.3390/ijms25052495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
The P2X7 receptor, a member of the P2X purinergic receptor family, is a non-selective ion channel. Over the years, it has been associated with various biological functions, from modulating to regulating inflammation. However, its emerging role in antigen presentation has captured the scientific community's attention. This function is essential for the immune system to identify and respond to external threats, such as pathogens and tumor cells, through T lymphocytes. New studies show that the P2X7 receptor is crucial for controlling how antigens are presented and how T cells are activated. These studies focus on antigen-presenting cells, like dendritic cells and macrophages. This review examines how the P2X7 receptor interferes with effective antigen presentation and activates T cells and discusses the fundamental mechanisms that can affect the immune response. Understanding these P2X7-mediated processes in great detail opens up exciting opportunities to create new immunological therapies.
Collapse
Affiliation(s)
- Claudio Acuña-Castillo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Alejandro Escobar
- Laboratorio Biología Celular y Molecular, Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380000, Chile
| | - Moira García-Gómez
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| | - Vivienne C Bachelet
- Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Juan Pablo Huidobro-Toro
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
- Centro Ciencia & Vida, Av. Del Valle Norte 725, Huechuraba 8580000, Chile
| | - Carlos Barrera-Avalos
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| |
Collapse
|
3
|
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: 10] [Impact Index Per Article: 5.0] [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
|
4
|
Mg.ATP-decorated ultrafine magnetic nanofibers: A bone scaffold with high osteogenic and antibacterial properties in the presence of an electromagnetic field. Colloids Surf B Biointerfaces 2021; 210:112256. [PMID: 34875469 DOI: 10.1016/j.colsurfb.2021.112256] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 12/27/2022]
Abstract
In this study, ultrafine magnetic nanofibers were developed for bone regeneration purposes. Nanofibers were acquired by electrospinning using a two-component nanofiber matrix (CP: chitosan (Cs) and polyvinyl alcohol (PVA)) containing different concentrations of succinate conjugated-magnetic hydroxyapatite nanocomposites (SMHA). Hybrid nanofibers (CP&SMHA) containing 5 mg ml-1 of SMHA nanocomposite showed well-defined properties in terms of physicochemical properties and cell behavior. Then, they were modified with adenosine 5'-triphosphate (ATP) and Mg2+ ions. The initial adhesion of mesenchymal stem cells and their proliferation rate on the surface of modified nanofibers (Mg.ATP.CP&SMHA) were significantly increased as compared to those of bare nanofibers. Analysis of common osteogenic markers such as alkaline phosphatase activity and the expression of Runt-related transcription factor 2 and osteocalcin confirmed the osteogenic efficacy enhancement of CP&SMHA nanofibers when they were functionalized with ATP and Mg2+. The utilization of the antagonist of purine receptor, P2X7, revealed that this receptor has a major role in the osteogenesis process induced by Mg.ATP.CP&SMHA. Moreover, the results showed that cell adhesion, proliferation, and differentiation improved as nanofibers were under the influence of the electromagnetic field (EMF), displaying synergistic effects in the process of bone formation. Mg.ATP.CP&SMHA also showed an antibacterial effect against gram-negative and gram-positive bacteria, Escherichia coli and Staphylococcus aureus, respectively. Considering the high osteogenic potential and antibacterial activity of Mg.ATP.CP&SMHA nanofibers particularly in combination with EMF, it can serve as a great candidate for use in bone tissue engineering applications.
Collapse
|
5
|
P2X7 promotes metastatic spreading and triggers release of miRNA-containing exosomes and microvesicles from melanoma cells. Cell Death Dis 2021; 12:1088. [PMID: 34789738 PMCID: PMC8599616 DOI: 10.1038/s41419-021-04378-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/14/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022]
Abstract
Tumor growth and metastatic spreading are heavily affected by the P2X7 receptor as well as microvesicles and exosomes release into the tumor microenvironment. P2X7 receptor stimulation is known to trigger vesicular release from immune and central nervous system cells. However, P2X7 role in microvesicles and exosomes delivery from tumor cells was never analyzed in depth. Here we show that P2X7 is overexpressed in patients affected by metastatic malignant melanoma and that its expression closely correlates with reduced overall survival. Antagonism of melanoma cell-expressed P2X7 receptor inhibited in vitro anchorage-independent growth and migration and in vivo dissemination and lung metastasis formation. P2X7 stimulation triggered the release of miRNA-containing microvesicles and exosomes from melanoma cells, profoundly altering the nature of their miRNA content, as well as their dimensions and quantity. Among the more than 200 miRNAs that we found up-or-down-modulated for each vesicular fraction tested, we identified three miRNAs, miR-495-3p, miR-376c-3p, and miR-6730-3p, that were enriched in both the exosome and microvesicle fraction in a P2X7-dependent fashion. Interestingly, upon transfection, these miRNAs promoted melanoma cell growth or migration, and their vesicular release was minimized by P2X7 antagonism. Our data unveil an exosome/microvesicle and miRNA-dependent mechanism for the pro-metastatic activity of the P2X7 receptor and highlight this receptor as a suitable prognostic biomarker and therapeutic target in malignant melanoma.
Collapse
|
6
|
Zuccarini M, Giuliani P, Caciagli F, Ciccarelli R, Di Iorio P. In Search of a Role for Extracellular Purine Enzymes in Bone Function. Biomolecules 2021; 11:biom11050679. [PMID: 33946568 PMCID: PMC8147220 DOI: 10.3390/biom11050679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is one of the major tissues that undergoes continuous remodeling throughout life, thus ensuring both organic body growth during development and protection of internal organs as well as repair of trauma during adulthood. Many endogenous substances contribute to bone homeostasis, including purines. Their role has increasingly emerged in recent decades as compounds which, by interacting with specific receptors, can help determine adequate responses of bone cells to physiological or pathological stimuli. Equally, it is recognized that the activity of purines is closely dependent on their interconversion or metabolic degradation ensured by a series of enzymes present at extracellular level as predominantly bound to the cell membrane or, also, as soluble isoforms. While the effects of purines mediated by their receptor interactions have sufficiently, even though not entirely, been characterized in many tissues including bone, those promoted by the extracellular enzymes providing for purine metabolism have not been. In this review, we will try to circumstantiate the presence and the role of these enzymes in bone to define their close relationship with purine activities in maintaining bone homeostasis in normal or pathological conditions.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| | - Francesco Caciagli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
- StemTeCh Group, Via L. Polacchi, 66100 Chieti, Italy
- Correspondence:
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (P.G.); (P.D.I.)
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100 Chieti, Italy;
| |
Collapse
|
7
|
Abstract
The P2X7 receptor for extracellular ATP is a well-established mediator of tumoral development and progression both in solid cancers and hematological malignancies. The human P2X7 gene is highly polymorphic, and several splice variants of the receptor have been identified in time. P2X7 single-nucleotide polymorphisms (SNPs) have been broadly analyzed by studies relating them to pathologies as different as infectious, inflammatory, nervous, and bone diseases, among which cancer is included. Moreover, in the last years, an increasing number of reports concentrated on P2X7 splice variants’ different roles and their implications in pathological conditions, including oncogenesis. Here, we give an overview of established and recent literature demonstrating a role for human P2X7 gene products in oncological conditions, mainly focusing on current data emerging on P2X7 isoform B and nfP2X7. We explored the role of these and other genetic variants of P2X7 in cancer insurgence, dissemination, and progression, as well as the effect of chemotherapy on isoforms expression. The described literature strongly suggests that P2X7 variants are potential new biomarkers and therapeutical targets in oncological conditions and that their study in carcinogenesis deserves to be further pursued.
Collapse
|
8
|
Pegoraro A, Orioli E, De Marchi E, Salvestrini V, Milani A, Di Virgilio F, Curti A, Adinolfi E. Differential sensitivity of acute myeloid leukemia cells to daunorubicin depends on P2X7A versus P2X7B receptor expression. Cell Death Dis 2020; 11:876. [PMID: 33071281 PMCID: PMC7569086 DOI: 10.1038/s41419-020-03058-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/08/2023]
Abstract
Acute myeloid leukemia (AML) is a common adult leukemia often arising from a preexistent myelodysplastic syndrome (MDS). High mortality rates of AML are caused by relapse and chemoresistance; therefore, we analyzed the role of P2X7 receptor (P2X7R) splice variants A and B in AML progression and response to chemotherapy. The expression of P2X7RA and P2X7RB was investigated in samples obtained from MDS and AML untreated subjects or AML patients in relapse or remission after chemotherapy. Both P2X7RA and P2X7RB were overexpressed in AML versus MDS suggesting a disease-promoting function. However, in relapsing patients, P2X7RA was downmodulated, while P2X7RB was upmodulated. Treatment with daunorubicin (DNR), one of the main chemotherapeutics for AML, upregulated P2X7RB expression while reducing P2X7RA mRNA in AML blasts. Interestingly, DNR administration also caused ATP release from AML blasts suggesting that, following chemotherapy, activation of the receptor isoforms via their agonist will be responsible for the differential survival of blasts overexpressing P2X7RA versus P2X7RB. Indeed, AML blasts expressing high levels of P2X7RA were more prone to cell death if exposed to DNR, while those overexpressing P2X7RB were more vital and even protected against DNR toxicity. These data were reproducible also in HEK-293 cells separately expressing P2X7RA and B. P2X7RA facilitation of DNR toxicity was in part due to increased uptake of the drug inside the cell that was lost upon P2X7RB expression. Finally, in an AML xenograft model administration of DNR or the P2X7R antagonist, AZ10606120 significantly reduced leukemic growth and coadministration of the drugs proved more efficacious than single treatment as it reduced both P2X7RA and P2X7RB levels and downmodulated c-myc oncogene. Taken together, our data suggest P2X7RA and P2X7RB as potential prognostic markers for AML and P2X7RB as a therapeutic target to overcome chemoresistance in AML relapsing patients.
Collapse
Affiliation(s)
- Anna Pegoraro
- Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Elisa Orioli
- Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Elena De Marchi
- Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Valentina Salvestrini
- Department of Haematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Haematology "L. and A. Seràgnoli", 40138, Bologna, Italy
| | - Asia Milani
- Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | | | - Antonio Curti
- Department of Haematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Haematology "L. and A. Seràgnoli", 40138, Bologna, Italy
| | - Elena Adinolfi
- Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy.
| |
Collapse
|
9
|
Ollà I, Santos-Galindo M, Elorza A, Lucas JJ. P2X7 Receptor Upregulation in Huntington's Disease Brains. Front Mol Neurosci 2020; 13:567430. [PMID: 33122998 PMCID: PMC7573237 DOI: 10.3389/fnmol.2020.567430] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/28/2020] [Indexed: 01/02/2023] Open
Abstract
Huntington’s disease (HD) is a fatal degenerative disorder affecting the nervous system. It is characterized by motor, cognitive, and psychiatric dysfunctions, with a late onset and an autosomal dominant pattern of inheritance. HD-causing mutation consists in an expansion of repeated CAG triplets in the huntingtin gene (HTT), encoding for an expanded polyglutamine (polyQ) stretch in the huntingtin protein (htt). The mutation causes neuronal dysfunction and loss through multiple mechanisms, affecting both the nucleus and cytoplasm. P2X7 receptor (P2X7R) emerged as a major player in neuroinflammation, since ATP – its endogenous ligand – is massively released under this condition. Indeed, P2X7R stimulation in the central nervous system (CNS) is known to enhance the release of pro-inflammatory cytokines from microglia and of neurotransmitters from neuronal presynaptic terminals, as well as to promote apoptosis. Previous experiments performed with neurons expressing the mutant huntingtin and exploiting HD mouse models demonstrated a role of P2X7R in HD. On the basis of those results, here, we explore for the first time the status of P2X7R in HD patients’ brain. We report that in HD postmortem striatum, as earlier observed in HD mice, the protein levels of the full-length form of P2X7R, also named P2X7R-A, are upregulated. In addition, the exclusively human naturally occurring variant lacking the C-terminus region, P2X7R-B, is upregulated as well. As we show here, this augmented protein levels can be explained by elevated mRNA levels. Furthermore, in HD patients’ striatum, P2X7R shows not only an augmented total transcript level but also an alteration of its splicing. Remarkably, P2X7R introns 10 and 11 are more retained in HD patients when compared with controls. Taken together, our data confirm that P2X7R is altered in brains of HD subjects and strengthen the notion that P2X7R may represent a potential therapeutic target for HD.
Collapse
Affiliation(s)
- Ivana Ollà
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO) CSIC/UAM, Madrid, Spain.,Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - María Santos-Galindo
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO) CSIC/UAM, Madrid, Spain.,Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ainara Elorza
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO) CSIC/UAM, Madrid, Spain.,Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - José J Lucas
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO) CSIC/UAM, Madrid, Spain.,Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
10
|
de Oliveira M, Mathias LS, de Sibio MT, Noronha-Matos JB, Costa MA, Nogueira CR, Correia-de-Sá P. Pitfalls and challenges of the purinergic signaling cascade in obesity. Biochem Pharmacol 2020; 182:114214. [PMID: 32905795 DOI: 10.1016/j.bcp.2020.114214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/15/2022]
Abstract
Obesity is a worldwide health problem which have reached pandemic proportions, now also including low and middle-income countries. Excessive or abnormal fat deposition in the abdomen especially in the visceral compartment is tightly associated with a high metabolic risk for arterial hypertension, type II diabetes, cardiovascular diseases, musculoskeletal disorders (especially articular degeneration) and some cancers. Contrariwise, accumulation of fat in the subcutaneous compartment has been associated with a neutral metabolic impact, favoring a lower risk of insulin resistance. Obesity results more often from an avoidable imbalance between food consumption and energy expenditure. There are several recommended strategies for dealing with obesity, including pharmacological therapies, but their success remains incomplete and may not compensate the associated adverse effects. Purinergic signaling operated by ATP and its metabolite, adenosine, has attracted increasing attention in obesity. The extracellular levels of purines often reflect the energy status of a given cell population. Adenine nucleotides and nucleosides fine tuning control adipogenesis and mature adipocytes function via the activation of P2 and P1 purinoceptors, respectively. These features make the purinergic signaling cascade a putative target for therapeutic intervention in obesity and related metabolic syndromes. There are, however, gaps in our knowledge regarding the role of purines in adipocyte precursors differentiation and mature adipocytes functions, as well as their impact among distinct adipose tissue deposits (e.g. white vs. brown, visceral vs. subcutaneous), which warrants further investigations before translation to clinical trials can be made.
Collapse
Affiliation(s)
- Miriane de Oliveira
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Lucas Solla Mathias
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Maria Teresa de Sibio
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Maria Adelina Costa
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Departamento de Química, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Célia Regina Nogueira
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
11
|
Benzaquen J, Dit Hreich SJ, Heeke S, Juhel T, Lalvee S, Bauwens S, Saccani S, Lenormand P, Hofman V, Butori M, Leroy S, Berthet JP, Marquette CH, Hofman P, Vouret-Craviari V. P2RX7B is a new theranostic marker for lung adenocarcinoma patients. Theranostics 2020; 10:10849-10860. [PMID: 33042257 PMCID: PMC7532666 DOI: 10.7150/thno.48229] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/21/2020] [Indexed: 12/25/2022] Open
Abstract
Rationale: The characterization of new theranostic biomarkers is crucial to improving the clinical outcome of patients with advanced lung cancer. Here, we aimed at characterizing the P2RX7 receptor, a positive modulator of the anti-tumor immune response, in patients with lung adenocarcinoma. Methods: The expression of P2RX7 and its splice variants was analyzed by RT-qPCR using areas of tumor and non-tumor lung adenocarcinoma (LUAD) tissues on both immune and non-immune cells. The biological activity of P2RX7 was studied by flow cytometry using fluorescent dyes. Bi-molecular fluorescence complementation and confocal microscopy were used to assess the oligomerization of P2RX7. Tumor immune infiltrates were characterized by immunohistochemistry. Results: Fifty-three patients with LUAD were evaluated. P2RX7A, and 3 alternative splice variants were expressed in LUAD tissues and expression was down regulated in tumor versus adjacent non-tumor tissues. The protein retained biological activity only in immune cells. The P2RX7B splice variant was differentially upregulated in immune cells (P < 0.001) of the tumor and strong evidence of oligomerization of P2RX7A and B was observed in the HEK expression model, which correlated with a default in the activity of P2RX7. Finally, LUAD patients with a high level of P2RX7B had non-inflamed tumors (P = 0.001). Conclusion: Our findings identified P2RX7B as a new theranostic tool to restore functional P2RX7 activity and open alternative therapeutic opportunities to improve LUAD patient outcome.
Collapse
|
12
|
Carluccio M, Ziberi S, Zuccarini M, Giuliani P, Caciagli F, Di Iorio P, Ciccarelli R. Adult mesenchymal stem cells: is there a role for purine receptors in their osteogenic differentiation? Purinergic Signal 2020; 16:263-287. [PMID: 32500422 DOI: 10.1007/s11302-020-09703-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
The role played by mesenchymal stem cells (MSCs) in contributing to adult tissue homeostasis and damage repair thanks to their differentiation capabilities has raised a great interest, mainly in bone regenerative medicine. The growth/function of these undifferentiated cells of mesodermal origin, located in specialized structures (niches) of differentiated organs is influenced by substances present in this microenvironment. Among them, ancestral and ubiquitous molecules such as adenine-based purines, i.e., ATP and adenosine, may be included. Notably, extracellular purine concentrations greatly increase during tissue injury; thus, MSCs are exposed to effects mediated by these agents interacting with their own receptors when they act/migrate in vivo or are transplanted into a damaged tissue. Here, we reported that ATP modulates MSC osteogenic differentiation via different P2Y and P2X receptors, but data are often inconclusive/contradictory so that the ATP receptor importance for MSC physiology/differentiation into osteoblasts is yet undetermined. An exception is represented by P2X7 receptors, whose expression was shown at various differentiation stages of bone cells resulting essential for differentiation/survival of both osteoclasts and osteoblasts. As well, adenosine, usually derived from extracellular ATP metabolism, can promote osteogenesis, likely via A2B receptors, even though findings from human MSCs should be implemented and confirmed in preclinical models. Therefore, although many data have revealed possible effects caused by extracellular purines in bone healing/remodeling, further studies, hopefully performed in in vivo models, are necessary to identify defined roles for these compounds in favoring/increasing the pro-osteogenic properties of MSCs and thereby their usefulness in bone regenerative medicine.
Collapse
Affiliation(s)
- Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy.,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy
| | - Sihana Ziberi
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy.,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Francesco Caciagli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy. .,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy. .,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy.
| |
Collapse
|
13
|
Di Virgilio F, Jiang LH, Roger S, Falzoni S, Sarti AC, Vultaggio-Poma V, Chiozzi P, Adinolfi E. Structure, function and techniques of investigation of the P2X7 receptor (P2X7R) in mammalian cells. Methods Enzymol 2019; 629:115-150. [PMID: 31727237 DOI: 10.1016/bs.mie.2019.07.043] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The P2X7 receptor [P2X7R or P2RX7 in National Center for Biotechnology Information (NCBI) gene nomenclature] is a member of the P2X receptor (P2XR) subfamily of P2 receptors (P2Rs). The P2X7R is an extracellular ATP-gated ion channel with peculiar permeability properties expressed by most cell types, mainly in the immune system, where it has a leading role in cytokine release, oxygen radical generation, T lymphocyte differentiation and proliferation. A role in cancer cell growth and tumor progression has also been demonstrated. These features make the P2X7R an appealing target for drug development in inflammation and cancer. The functional P2X7R, recently (partially) crystallized and 3-D solved, is formed by the assembly of three identical subunits (homotrimer). The P2X7R is preferentially permeable to small cations (Ca2+, Na+, K+), and in most (but not all) cell types also to large positively charged molecules of molecular mass up to 900Da. Permeability to negatively charged species of comparable molecular mass (e.g., Lucifer yellow) is debated. Several highly selective P2X7R pharmacological blockers have been developed over the years, thus providing powerful tools for P2X7R studies. Biophysical properties and coupling to several different physiological responses make the P2X7R amenable to investigation by electrophysiology and cell biology techniques, which allow its identification and characterization in many different cell types and tissues. A careful description of the physiological features of the P2X7R is a prerequisite for an effective therapeutic development. Here we describe the most common techniques to asses P2X7R functions, including patch-clamp, intracellular calcium measurements, and membrane permeabilization to large fluorescent dyes in a selection of different cell types. In addition, we also describe common toxicity assays used to verify the effects of P2X7R stimulation on cell viability.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Sébastien Roger
- EA4245 Transplantation, Immunology and Inflammation, University of Tours, Tours, France
| | - Simonetta Falzoni
- 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
| | - Valentina Vultaggio-Poma
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Chiozzi
- 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
|