1
|
Ornelas IM, Silva TM, Pereira MR, França GR, Ventura ALM. Cell cycle regulation by ADP and IGF-1 in cultured late developing glia progenitors of the avian retina. Purinergic Signal 2025; 21:179-191. [PMID: 38151691 PMCID: PMC11958921 DOI: 10.1007/s11302-023-09982-7] [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: 08/15/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023] Open
Abstract
In the avian retina, ADP induces the proliferation of late developing glia progenitors. Here, we show that in serum-containing retinal cell cultures, ADP-induced increase in [3H]-thymidine incorporation can be prevented by the IGF-1 receptor antagonists AG1024 and I-OMe-Tyrphostin AG 538, suggesting the participation of IGF-1 in ADP-mediated progenitor proliferation. In contrast, no increase in [3H]-thymidine incorporation is observed in retinal cultures treated only with IGF-1. Under serum starvation, while no increase in cell proliferation is detected in cultures treated only with ADP or IGF-1, a significant increase in [3H]-thymidine incorporation and number of PCNA expressing cells is observed in cultures treated concomitantly with ADP plus IGF-1, suggesting that both molecules are required to induce proliferation of retinal progenitors. In serum-starved cultures, although an increase in cell viability is detected by MTT assays in IGF-1-treated cultures, no significant increase in viability of [3H]-thymidine labeled progenitors is observed, suggesting that IGF-1 may contribute to survival of postmitotic cells in culture. While only ADP increases intracellular calcium, only IGF-1 induces the phosphorylation of Akt in the retinal cultures. IGF-1 through the PI3K/Akt pathway induces a significant increase in the transcription and expression of CDK1 with a decrease in phospho-histone H3 expression that is concomitant with an increase in the expression of cyclins D1 and E and CDK2. These findings suggest that IGF-1 stimulates CDK-1 mRNA and protein expression that enable progenitors to progress through the cell cycle. However, signaling of ADP in the presence IGF-I seems to be required for DNA synthesis.
Collapse
Affiliation(s)
- Isis Moraes Ornelas
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, 29047-105, Brazil
| | - Thayane Martins Silva
- Department of Neurobiology, Neuroscience Program, Federal Fluminense University, Rua Prof. M.W. de Freitas Reis, bloco M, sala 409, São Domingos, Niterói, Rio de Janeiro, CEP 24210-201, Brazil
| | - Mariana Rodrigues Pereira
- Department of Neurobiology, Neuroscience Program, Federal Fluminense University, Rua Prof. M.W. de Freitas Reis, bloco M, sala 409, São Domingos, Niterói, Rio de Janeiro, CEP 24210-201, Brazil
| | - Guilherme Rapozeiro França
- Department of Physiological Sciences, Federal University of the State of Rio de Janeiro, Rua Frei Caneca 94, Centro, Rio de Janeiro, RJ, CEP 20211-040, Brazil
| | - Ana Lucia Marques Ventura
- Department of Neurobiology, Neuroscience Program, Federal Fluminense University, Rua Prof. M.W. de Freitas Reis, bloco M, sala 409, São Domingos, Niterói, Rio de Janeiro, CEP 24210-201, Brazil.
| |
Collapse
|
2
|
Tempone MH, Borges-Martins VP, César F, Alexandrino-Mattos DP, de Figueiredo CS, Raony Í, dos Santos AA, Duarte-Silva AT, Dias MS, Freitas HR, de Araújo EG, Ribeiro-Resende VT, Cossenza M, P. Silva H, P. de Carvalho R, Ventura ALM, Calaza KC, Silveira MS, Kubrusly RCC, de Melo Reis RA. The Healthy and Diseased Retina Seen through Neuron-Glia Interactions. Int J Mol Sci 2024; 25:1120. [PMID: 38256192 PMCID: PMC10817105 DOI: 10.3390/ijms25021120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The retina is the sensory tissue responsible for the first stages of visual processing, with a conserved anatomy and functional architecture among vertebrates. To date, retinal eye diseases, such as diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, glaucoma, and others, affect nearly 170 million people worldwide, resulting in vision loss and blindness. To tackle retinal disorders, the developing retina has been explored as a versatile model to study intercellular signaling, as it presents a broad neurochemical repertoire that has been approached in the last decades in terms of signaling and diseases. Retina, dissociated and arranged as typical cultures, as mixed or neuron- and glia-enriched, and/or organized as neurospheres and/or as organoids, are valuable to understand both neuronal and glial compartments, which have contributed to revealing roles and mechanisms between transmitter systems as well as antioxidants, trophic factors, and extracellular matrix proteins. Overall, contributions in understanding neurogenesis, tissue development, differentiation, connectivity, plasticity, and cell death are widely described. A complete access to the genome of several vertebrates, as well as the recent transcriptome at the single cell level at different stages of development, also anticipates future advances in providing cues to target blinding diseases or retinal dysfunctions.
Collapse
Affiliation(s)
- Matheus H. Tempone
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Vladimir P. Borges-Martins
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Felipe César
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Dio Pablo Alexandrino-Mattos
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Camila S. de Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ícaro Raony
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Aline Araujo dos Santos
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Aline Teixeira Duarte-Silva
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana Santana Dias
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Hércules Rezende Freitas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Elisabeth G. de Araújo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Victor Tulio Ribeiro-Resende
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Hilda P. Silva
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Roberto P. de Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ana L. M. Ventura
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Karin C. Calaza
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana S. Silveira
- Laboratory for Investigation in Neuroregeneration and Development, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil;
| | - Regina C. C. Kubrusly
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Ricardo A. de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| |
Collapse
|
3
|
Markitantova YV, Simirskii VN. The Role of the Purinergic Signaling System in the Control of Histogenesis, Homeostasis, and Pathogenesis of the Vertebrate Retina. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
4
|
Cannabinoids Induce Cell Death and Promote P2X7 Receptor Signaling in Retinal Glial Progenitors in Culture. Mol Neurobiol 2019; 56:6472-6486. [DOI: 10.1007/s12035-019-1537-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022]
|
5
|
Ventura ALM, Dos Santos-Rodrigues A, Mitchell CH, Faillace MP. Purinergic signaling in the retina: From development to disease. Brain Res Bull 2018; 151:92-108. [PMID: 30458250 DOI: 10.1016/j.brainresbull.2018.10.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/14/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
Retinal injuries and diseases are major causes of human disability involving vision impairment by the progressive and permanent loss of retinal neurons. During development, assembly of this tissue entails a successive and overlapping, signal-regulated engagement of complex events that include proliferation of progenitors, neurogenesis, cell death, neurochemical differentiation and synaptogenesis. During retinal damage, several of these events are re-activated with both protective and detrimental consequences. Purines and pyrimidines, along with their metabolites are emerging as important molecules regulating both retinal development and the tissue's responses to damage. The present review provides an overview of the purinergic signaling in the developing and injured retina. Recent findings on the presence of vesicular and channel-mediated ATP release by retinal and retinal pigment epithelial cells, adenosine synthesis and release, expression of receptors and intracellular signaling pathways activated by purinergic signaling in retinal cells are reported. The pathways by which purinergic receptors modulate retinal cell proliferation, migration and death of retinal cells during development and injury are summarized. The contribution of nucleotides to the self-repair of the injured zebrafish retina is also discussed.
Collapse
Affiliation(s)
- Ana Lucia Marques Ventura
- Department of Neurobiology, Neuroscience Program, Fluminense Federal University, Niterói, RJ, Brazil.
| | | | - Claire H Mitchell
- Department of Anatomy and Cell Biology, Ophthalmology, and Physiology, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Maria Paula Faillace
- Instituto de Fisiología y Biofísica Prof. Bernardo Houssay (IFIBIO-Houssay), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Jacques FJ, Silva TM, da Silva FE, Ornelas IM, Ventura ALM. Nucleotide P2Y13-stimulated phosphorylation of CREB is required for ADP-induced proliferation of late developing retinal glial progenitors in culture. Cell Signal 2017; 35:95-106. [PMID: 28347874 DOI: 10.1016/j.cellsig.2017.03.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/23/2017] [Accepted: 03/24/2017] [Indexed: 12/28/2022]
Abstract
Nucleotides stimulate phosphorylation of CREB to induce cell proliferation and survival in diverse cell types. We report here that ADP induces the phosphorylation of CREB in a time- and concentration-dependent manner in chick embryo retinal progenitors in culture. ADP-induced increase in phospho-CREB is mediated by P2 receptors as it is blocked by PPADS but not by the adenosine antagonists DPCPX or ZM241385. Incubation of the cultures with the CREB inhibitor KG-501 prevents ADP-induced incorporation of [3H]-thymidine, indicating that CREB is involved in retinal cell proliferation. No effect of this compound is observed on the viability of retinal progenitors. While no significant increase in CREB phosphorylation is observed with the P2Y1 receptor agonist MRS2365, ADP-induced phosphorylation of CREB is blocked by the P2Y13 receptor selective antagonist MRS2211, but not by MRS2179 or PSB0739, two antagonists of the P2Y1 and P2Y12 receptors, respectively, suggesting that ADP-induced CREB phosphorylation is mediated by P2Y13 receptors. ADP-induced increase in phospho-CREB is attenuated by the PI3K inhibitor LY294002 and completely prevented by the MEK inhibitor U0126, suggesting that at least ERK is involved in ADP-induced CREB phosphorylation. A pharmacological profile similar to the activation and inhibition of CREB phosphorylation is observed in the phosphorylation of ERK, suggesting that P2Y13 receptors mediate ADP induced ERK/CREB pathway in the cultures. While no increase in [3H]-thymidine incorporation is observed with the P2Y1 receptor agonist MRS2365, both MRS2179 and MRS2211 prevent ADP-mediated increase in [3H]-thymidine incorporation, but not progenitor's survival, suggesting that both P2Y1 and P2Y13 receptor subtypes are involved in ADP-induced cell proliferation. P2Y1 receptor-mediated increase in [Ca2+]i is observed in glial cells only when cultures maintained for 9days are used. In glia from cultures cultivated for only 2days, no increase in [Ca2+]i is detected with MRS2365 and no inhibition of ADP-mediated calcium response is observed with MRS2179. In contrast, MRS2211 attenuates ADP-mediated increase in [Ca2+]i in glial cells from cultures at both stages, suggesting the presence of P2Y13 receptors coupled to calcium mobilization in proliferating retinal glial progenitors in culture.
Collapse
Affiliation(s)
- Flavia Jesus Jacques
- Department of Neurobiology, Neuroscience Program, Fluminense Federal University, Outeiro de São João Batista s/n, Centro, Niterói, Rio de Janeiro CEP 24020-141, Brazil
| | - Thayane Martins Silva
- Department of Neurobiology, Neuroscience Program, Fluminense Federal University, Outeiro de São João Batista s/n, Centro, Niterói, Rio de Janeiro CEP 24020-141, Brazil
| | - Flavia Emenegilda da Silva
- Department of Neurobiology, Neuroscience Program, Fluminense Federal University, Outeiro de São João Batista s/n, Centro, Niterói, Rio de Janeiro CEP 24020-141, Brazil
| | - Isis Moraes Ornelas
- Department of Neurobiology, Neuroscience Program, Fluminense Federal University, Outeiro de São João Batista s/n, Centro, Niterói, Rio de Janeiro CEP 24020-141, Brazil
| | - Ana Lucia Marques Ventura
- Department of Neurobiology, Neuroscience Program, Fluminense Federal University, Outeiro de São João Batista s/n, Centro, Niterói, Rio de Janeiro CEP 24020-141, Brazil.
| |
Collapse
|
7
|
Tang Y, Illes P. Regulation of adult neural progenitor cell functions by purinergic signaling. Glia 2016; 65:213-230. [PMID: 27629990 DOI: 10.1002/glia.23056] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/20/2016] [Accepted: 08/23/2016] [Indexed: 01/30/2023]
Abstract
Extracellular purines are signaling molecules in the neurogenic niches of the brain and spinal cord, where they activate cell surface purinoceptors at embryonic neural stem cells (NSCs) and adult neural progenitor cells (NPCs). Although mRNA and protein are expressed at NSCs/NPCs for almost all subtypes of the nucleotide-sensitive P2X/P2Y, and the nucleoside-sensitive adenosine receptors, only a few of those have acquired functional significance. ATP is sequentially degraded by ecto-nucleotidases to ADP, AMP, and adenosine with agonistic properties for distinct receptor-classes. Nucleotides/nucleosides facilitate or inhibit NSC/NPC proliferation, migration and differentiation. The most ubiquitous effect of all agonists (especially of ATP and ADP) appears to be the facilitation of cell proliferation, usually through P2Y1Rs and sometimes through P2X7Rs. However, usually P2X7R activation causes necrosis/apoptosis of NPCs. Differentiation can be initiated by P2Y2R-activation or P2X7R-blockade. A key element in the transduction mechanism of either receptor is the increase of the intracellular free Ca2+ concentration, which may arise due to its release from intracellular storage sites (G protein-coupling; P2Y) or due to its passage through the receptor-channel itself from the extracellular space (ATP-gated ion channel; P2X). Further research is needed to clarify how purinergic signaling controls NSC/NPC fate and how the balance between the quiescent and activated states is established with fine and dynamic regulation. GLIA 2017;65:213-230.
Collapse
Affiliation(s)
- Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, 04107, Germany
| |
Collapse
|
8
|
de Almeida-Pereira L, Magalhães CF, Repossi MG, Thorstenberg MLP, Sholl-Franco A, Coutinho-Silva R, Ventura ALM, Fragel-Madeira L. Adenine Nucleotides Control Proliferation In Vivo of Rat Retinal Progenitors by P2Y 1 Receptor. Mol Neurobiol 2016; 54:5142-5155. [PMID: 27558237 DOI: 10.1007/s12035-016-0059-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/15/2016] [Indexed: 11/30/2022]
Abstract
Previous studies demonstrated that exogenous ATP is able to regulate proliferation of retinal progenitor cells (RPCs) in vitro possibly via P2Y1 receptor, a G protein-coupled receptor. Here, we evaluated the function of adenine nucleotides in vivo during retinal development of newborn rats. Intravitreal injection of apyrase, an enzyme that hydrolyzes nucleotides, reduced cell proliferation in retinas at postnatal day 2 (P2). This decrease was reversed when retinas were treated together with ATPγ-S or ADPβ-S, two hydrolysis-resistant analogs of ATP and ADP, respectively. During early postnatal days (P0 to P5), an increase in ectonucleotidase (E-NTPDase) activity was observed in the retina, suggesting a decrease in the availability of adenine nucleotides, coinciding with the end of proliferation. Interestingly, intravitreal injection of the E-NTPDase inhibitor ARL67156 increased proliferation by around 60 % at P5 rats. Furthermore, immunolabeling against P2Y1 receptor was observed overall in retina layers from P2 rats, including proliferating Ki-67-positive cells in the neuroblastic layer (NBL), suggesting that this receptor could be responsible for the action of adenine nucleotides upon proliferation of RPCs. Accordingly, intravitreal injection of MRS2179, a selective antagonist of P2Y1 receptors, reduced cell proliferation by approximately 20 % in P2 rats. Moreover, treatment with MRS 2179 caused an increase in p57KIP2 and cyclin D1 expression, a reduction in cyclin E and Rb phosphorylated expression and in BrdU-positive cell number. These data suggest that the adenine nucleotides modulate the proliferation of rat RPCs via activation of P2Y1 receptors regulating transition from G1 to S phase of the cell cycle.
Collapse
Affiliation(s)
- Luana de Almeida-Pereira
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Camila Feitosa Magalhães
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Marinna Garcia Repossi
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | | | - Alfred Sholl-Franco
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Lucianne Fragel-Madeira
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil.
- Laboratório de Desenvolvimento e Regeneração Neural, Departmento de Neurobiologia, Universidade Federal Fluminense, Cx. Postal 100180, Niterói, RJ, 24020-141, Brazil.
| |
Collapse
|
9
|
Oliveira Á, Illes P, Ulrich H. Purinergic receptors in embryonic and adult neurogenesis. Neuropharmacology 2015; 104:272-81. [PMID: 26456352 DOI: 10.1016/j.neuropharm.2015.10.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/01/2015] [Accepted: 10/04/2015] [Indexed: 01/14/2023]
Abstract
ATP (adenosine 5'-triphosphate), one of the most ancient neurotransmitters, exerts essential functions in the brain, including neurotransmission and modulation of synaptic activity. Moreover, this nucleotide has been attributed with trophic properties and experimental evidence points to the participation of ATP-activated P2X and P2Y purinergic receptors in embryonic brain development as well as in adult neurogenesis for maintenance of normal brain functions and neuroregeneration upon brain injury. We discuss here the available data on purinergic P2 receptor expression and function during brain development and in the neurogenic zones of the adult brain, as well as the insights based on the use of in vitro stem cell cultures. While several P2 receptor subtypes were shown to be expressed during in vitro and in vivo neurogenesis, specific functions have been proposed for P2Y1, P2Y2 metabotropic as well as P2X2 ionotropic receptors to promote neurogenesis. Further, the P2X7 receptor is suggested to function in the maintenance of pools of neural stem and progenitor cells through induction of proliferation or cell death, depending on the microenvironment. Pathophysiological actions have been proposed for this receptor in worsening damage in brain disease. The P2X7 receptor and possibly additional P2 receptor subtypes have been implicated in pathophysiology of neurological diseases including Parkinson's disease, Alzheimer's disease and epilepsy. New strategies in cell therapy could involve modulation of purinergic signaling, either in the achievement of more effective protocols to obtain viable and homogeneous cell populations or in the process of functional engraftment of transplanted cells into the damaged brain. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Ágatha Oliveira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-900, Av. Prof. Lineu Prestes, 748, Brazil
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie der Universität Leipzig, Haertelstrasse 16-18, 04107 Leipzig, Germany.
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-900, Av. Prof. Lineu Prestes, 748, Brazil.
| |
Collapse
|
10
|
Burnstock G, Dale N. Purinergic signalling during development and ageing. Purinergic Signal 2015; 11:277-305. [PMID: 25989750 PMCID: PMC4529855 DOI: 10.1007/s11302-015-9452-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 01/28/2023] Open
Abstract
Extracellular purines and pyrimidines play major roles during embryogenesis, organogenesis, postnatal development and ageing in vertebrates, including humans. Pluripotent stem cells can differentiate into three primary germ layers of the embryo but may also be involved in plasticity and repair of the adult brain. These cells express the molecular components necessary for purinergic signalling, and their developmental fates can be manipulated via this signalling pathway. Functional P1, P2Y and P2X receptor subtypes and ectonucleotidases are involved in the development of different organ systems, including heart, blood vessels, skeletal muscle, urinary bladder, central and peripheral neurons, retina, inner ear, gut, lung and vas deferens. The importance of purinergic signalling in the ageing process is suggested by changes in expression of A1 and A2 receptors in old rat brains and reduction of P2X receptor expression in ageing mouse brain. By contrast, in the periphery, increases in expression of P2X3 and P2X4 receptors are seen in bladder and pancreas.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | |
Collapse
|
11
|
|
12
|
Silva TM, França GR, Ornelas IM, Loiola EC, Ulrich H, Ventura ALM. Involvement of nucleotides in glial growth following scratch injury in avian retinal cell monolayer cultures. Purinergic Signal 2015; 11:183-201. [PMID: 25663277 DOI: 10.1007/s11302-015-9444-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 01/21/2015] [Indexed: 10/24/2022] Open
Abstract
When retinal cell cultures were mechanically scratched, cell growth over the empty area was observed. Only dividing and migrating, 2 M6-positive glial cells were detected. Incubation of cultures with apyrase (APY), suramin, or Reactive Blue 2 (RB-2), but not MRS 2179, significantly attenuated the growth of glial cells, suggesting that nucleotide receptors other than P2Y1 are involved in the growth of glial cells. UTPγS but not ADPβS antagonized apyrase-induced growth inhibition in scratched cultures, suggesting the participation of UTP-sensitive receptors. No decrease in proliferating cell nuclear antigen (PCNA(+)) cells was observed at the border of the scratch in apyrase-treated cultures, suggesting that glial proliferation was not affected. In apyrase-treated cultures, glial cytoplasm protrusions were smaller and unstable. Actin filaments were less organized and alfa-tubulin-labeled microtubules were mainly parallel to scratch. In contrast to control cultures, very few vinculin-labeled adhesion sites could be noticed in these cultures. Increased Akt and ERK phosphorylation was observed in UTP-treated cultures, effect that was inhibited by SRC inhibitor 1 and PI3K blocker LY294002. These inhibitors and the FAK inhibitor PF573228 also decreased glial growth over the scratch, suggesting participation of SRC, PI3K, and FAK in UTP-induced growth of glial cells in scratched cultures. RB-2 decreased dissociated glial cell attachment to fibronectin-coated dishes and migration through transwell membranes, suggesting that nucleotides regulated adhesion and migration of glial cells. In conclusion, mechanical scratch of retinal cell cultures induces growth of glial cells over the empty area through a mechanism that is dependent on activation of UTP-sensitive receptors, SRC, PI3K, and FAK.
Collapse
Affiliation(s)
- Thayane Martins Silva
- Programa de Neurociências, Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, RJ, 24020-141, Brazil
| | | | | | | | | | | |
Collapse
|
13
|
Ornelas IM, Silva TM, Fragel-Madeira L, Ventura ALM. Inhibition of PI3K/Akt pathway impairs G2/M transition of cell cycle in late developing progenitors of the avian embryo retina. PLoS One 2013; 8:e53517. [PMID: 23301080 PMCID: PMC3534656 DOI: 10.1371/journal.pone.0053517] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/29/2012] [Indexed: 01/30/2023] Open
Abstract
PI3K/Akt is an important pathway implicated in the proliferation and survival of cells in the CNS. Here we investigated the participation of the PI3K/Akt signal pathway in cell cycle of developing retinal progenitors. Immunofluorescence assays performed in cultures of chick embryo retinal cells and intact tissues revealed the presence of phosphorylated Akt and 4E-BP1 in cells with typical mitotic profiles. Blockade of PI3K activity with the chemical inhibitor LY 294002 (LY) in retinal explants blocked the progression of proliferating cells through G2/M transition, indicated by an expressive increase in the number of cells labeled for phosphorylated histone H3 in the ventricular margin of the retina. No significant level of cell death could be detected at this region. Retinal explants treated with LY for 24 h also showed a significant decrease in the expression of phospho-Akt, phospho-GSK-3 and the hyperphosphorylated form of 4E-BP1. Although no change in the expression of cyclin B1 was detected, a significant decrease in CDK1 expression was noticed after 24 h of LY treatment both in retinal explants and monolayer cultures. Our results suggest that PI3K/Akt is an active pathway during proliferation of retinal progenitors and its activity appears to be required for proper CDK1 expression levels and mitosis progression of these cells.
Collapse
Affiliation(s)
- Isis Moraes Ornelas
- Department of Neurobiology, Neuroscience Program, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Thayane Martins Silva
- Department of Neurobiology, Neuroscience Program, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Lucianne Fragel-Madeira
- Department of Neurobiology, Neuroscience Program, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Ana Lucia Marques Ventura
- Department of Neurobiology, Neuroscience Program, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
14
|
Ulrich H, Abbracchio MP, Burnstock G. Extrinsic purinergic regulation of neural stem/progenitor cells: implications for CNS development and repair. Stem Cell Rev Rep 2012; 8:755-67. [PMID: 22544361 DOI: 10.1007/s12015-012-9372-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There has been tremendous progress in understanding neural stem cell (NSC) biology, with genetic and cell biological methods identifying sequential gene expression and molecular interactions guiding NSC specification into distinct neuronal and glial populations during development. Data has emerged on the possible exploitation of NSC-based strategies to repair adult diseased brain. However, despite increased information on lineage specific transcription factors, cell-cycle regulators and epigenetic factors involved in the fate and plasticity of NSCs, understanding of extracellular cues driving the behavior of embryonic and adult NSCs is still very limited. Knowledge of factors regulating brain development is crucial in understanding the pathogenetic mechanisms of brain dysfunction. Since injury-activated repair mechanisms in adult brain often recapitulate ontogenetic events, the identification of these players will also reveal novel regenerative strategies. Here, we highlight the purinergic system as a key emerging player in the endogenous control of NSCs. Purinergic signalling molecules (ATP, UTP and adenosine) act with growth factors in regulating the synchronized proliferation, migration, differentiation and death of NSCs during brain and spinal cord development. At early stages of development, transient and time-specific release of ATP is critical for initiating eye formation; once anatomical CNS structures are defined, purinergic molecules participate in calcium-dependent neuron-glia communication controlling NSC behaviour. When development is complete, some purinergic mechanisms are silenced, but can be re-activated in adult brain after injury, suggesting a role in regeneration and self-repair. Targeting the purinergic system to develop new strategies for neurodevelopmental disorders and neurodegenerative diseases will be also discussed.
Collapse
Affiliation(s)
- Henning Ulrich
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, São Paulo 05508-900, SP, Brazil.
| | | | | |
Collapse
|
15
|
Yamashita M. From neuroepithelial cells to neurons: changes in the physiological properties of neuroepithelial stem cells. Arch Biochem Biophys 2012; 534:64-70. [PMID: 22892549 DOI: 10.1016/j.abb.2012.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 07/04/2012] [Accepted: 07/27/2012] [Indexed: 10/28/2022]
Abstract
The central nervous system, which includes the spinal cord, retina, and brain, is derived from the neural tube. The neural tube is formed of a sheet of cells called the neuroepithelium. During embryonic development, neuroepithelial cells function as neural stem cells: they renew themselves while undergoing interkinetic nuclear movements along the apico-basal axis during the cell cycle, and they produce postmitotic cells that function as newborn neurons. Neuroepithelial cells exhibit a robust increase in nucleoplasmic [Ca(2+)] in response to G protein-coupled receptor activation during S-phase when the nucleus is located in the basal region of the cell. This Ca(2+) rise is caused by the release of Ca(2+) from intracellular Ca(2+) stores, and the Ca(2+) release in turn activates Ca(2+) entry from the extracellular space, which is called capacitative (or store-operated) Ca(2+) entry. The Ca(2+) release and store-operated Ca(2+) entry are essential for DNA synthesis during S-phase. The activity of this store-operated Ca(2+) signaling system declines in parallel with the decreasing proliferative activity of neuroepithelial cells. When exiting the cell cycle, the cells lose the apical process where gap junctions are located. Following the loss of gap junction coupling, the postmitotic cells show a high input resistance, which allows them to be readily depolarized. The Ca(2+) response to the excitatory neurotransmitter glutamate appears and develops during neuronal differentiation. The glutamate-induced Ca(2+) rise increases transiently during natural cell death (apoptosis). The rise in Ca(2+) levels mediated by voltage-gated Ca(2+) channels also develops during neuronal differentiation. Thus, when neuroepithelial cells differentiate into neurons, a transition from a store-operated system to a voltage-operated system occurs in the main Ca(2+) signaling system. This transition may reflect a change in the mode of intercellular communication from a stored Ca(2+)-dependent mode to a plasma membrane potential-dependent mode.
Collapse
Affiliation(s)
- Masayuki Yamashita
- Department of Physiology 1, Nara Medical University, Shijo-cho 840, Kashihara, Japan.
| |
Collapse
|
16
|
Suyama S, Sunabori T, Kanki H, Sawamoto K, Gachet C, Koizumi S, Okano H. Purinergic signaling promotes proliferation of adult mouse subventricular zone cells. J Neurosci 2012; 32:9238-47. [PMID: 22764232 PMCID: PMC6622243 DOI: 10.1523/jneurosci.4001-11.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 05/18/2012] [Accepted: 05/23/2012] [Indexed: 02/04/2023] Open
Abstract
In adult mammalian brains, neural stem cells (NSCs) exist in the subventricular zone (SVZ), where persistent neurogenesis continues throughout life. Those NSCs produce neuroblasts that migrate into the olfactory bulb via formation of transit-amplifying cells, which are committed precursor cells of the neuronal lineage. In this SVZ niche, cell-cell communications conducted by diffusible factors as well as physical cell-cell contacts are important for the regulation of the proliferation and fate determination of NSCs. Previous studies have suggested that extracellular purinergic signaling, which is mediated by purine compounds such as ATP, plays important roles in cell-cell communication in the CNS. Purinergic signaling also promotes the proliferation of adult NSCs in vitro. However, the in vivo roles of purinergic signaling in the neurogenic niche still remain unknown. In this study, ATP infusion into the lateral ventricle of the mouse brain resulted in an increase in the numbers of rapidly dividing cells and Mash1-positive transit-amplifying cells (Type C cells) in the SVZ. Mash1-positive cells express the P2Y1 purinergic signaling receptor and infusion of the P2Y1 receptor-specific antagonist MRS2179 decreased the number of rapidly dividing bromodeoxyuridine (BrdU)-positive cells and Type C cells. Moreover, a 17% reduction of rapidly dividing BrdU-positive cells and a 19% reduction of Mash1-positive cells were observed in P2Y1 knock-out mice. Together, these results suggest that purinergic signaling promotes the proliferation of rapidly dividing cells and transit-amplifying cells, in the SVZ niche through the P2Y1 receptor.
Collapse
Affiliation(s)
- Satoshi Suyama
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takehiko Sunabori
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan
| | - Hiroaki Kanki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Christian Gachet
- UMR_S949 INSERM, Université de Strasbourg, Etablissement Français du Sang-Alsace, 67065, Strasbourg Cedex, France, and
| | - Schuichi Koizumi
- Department of Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
17
|
ATP induces the death of developing avian retinal neurons in culture via activation of P2X7 and glutamate receptors. Purinergic Signal 2012; 9:15-29. [PMID: 22733428 DOI: 10.1007/s11302-012-9324-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 06/12/2012] [Indexed: 10/28/2022] Open
Abstract
Previous data suggest that nucleotides are important mitogens in the developing retina. Here, the effect of ATP on the death of cultured chick embryo retina cells was investigated. In cultures obtained from retinas of 7-day-old chick embryos (E7) that were cultivated for 2 days (E7C2), both ATP and BzATP induced a ∼30 % decrease in cell viability that was time- and dose-dependent and that could be blocked by 0.2 mM oxidized ATP or 0.3 μM KN-62. An increase in cleaved caspase-3 levels and in the number of TUNEL-positive cells was observed when cultures were incubated with 3 mM ATP and immunolabeling for cleaved-caspase 3 was observed over neurons but not over glial cells. ATP-dependent cell death was developmentally regulated, the maximal levels being detected by E7C2-3. Nucleotides were able to increase neuronal ethidium bromide and sulforhodamine B uptake in mixed and purified neuronal cultures, an effect that was blocked by the antagonists Brilliant Blue G and oxidized ATP. In contrast, nucleotide-induced cell death was observed only in mixed cultures, but not in purified cultures of neurons or glia. ATP-induced neuronal death was blocked by the glutamatergic antagonists MK801 and DNQX and activation of P2X7 receptors by ATP decreased the uptake of [(3)H]-D-aspartate by cultured glial cells with a concomitant accumulation of it in the extracellular medium. These results suggest that ATP induces apoptosis of chick embryo retinal neurons in culture through activation of P2X7 and glutamate ionotropic receptors. Involvement of a P2X7 receptor-mediated inhibition of the glial uptake of glutamate is suggested.
Collapse
|
18
|
Wurm A, Pannicke T, Iandiev I, Francke M, Hollborn M, Wiedemann P, Reichenbach A, Osborne NN, Bringmann A. Purinergic signaling involved in Müller cell function in the mammalian retina. Prog Retin Eye Res 2011; 30:324-42. [DOI: 10.1016/j.preteyeres.2011.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/06/2011] [Accepted: 06/06/2011] [Indexed: 10/18/2022]
|
19
|
Leecharoenkiat A, Wannatung T, Lithanatudom P, Svasti S, Fucharoen S, Chokchaichamnankit D, Srisomsap C, Smith DR. Increased oxidative metabolism is associated with erythroid precursor expansion in β0-thalassaemia/Hb E disease. Blood Cells Mol Dis 2011; 47:143-57. [PMID: 21783389 DOI: 10.1016/j.bcmd.2011.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 06/22/2011] [Accepted: 06/25/2011] [Indexed: 10/18/2022]
Abstract
Erythropoiesis in β0-thalassaemia/Hb E patients, the most common variant form of β-thalassaemia in Southeast Asia, is characterized by accelerated differentiation and over-expansion of erythroid precursor cells. The mechanism driving this accelerated expansion and differentiation remain unknown. To address this issue a proteomic analysis was undertaken to firstly identify proteins differentially expressed during erythroblast differentiation and a second analysis was undertaken to identify proteins differentially expressed between β0-thalassaemia/Hb E erythroblasts and control erythroblasts. The majority of proteins identified as being differentially expressed between β0-thalassaemia/Hb E and control erythroblasts were constituents of the glycolysis/TCA pathway and levels of oxidative stress correlated with the degree of erythroid expansion. A model was constructed linking these observations with previous studies showing increased phosphorylation of ERK1/2 in thalassemic erythroblasts which predicted the increased activation of PKA, PKB and PKC which Western analysis confirmed. Inhibition of PKA or PKC reduced β0-thalassaemia/Hb E erythroblast differentiation and/or expansion. We propose that increased expansion and differentiation of β0-thalassaemia/Hb E erythroblasts occur as a result of feedback loops acting through increased oxidative metabolism.
Collapse
Affiliation(s)
- Amporn Leecharoenkiat
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Zimmermann H. Purinergic signaling in neural development. Semin Cell Dev Biol 2011; 22:194-204. [DOI: 10.1016/j.semcdb.2011.02.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/07/2011] [Accepted: 02/07/2011] [Indexed: 02/07/2023]
|
21
|
Zhang L, Zhang Y, Liu XY, Qin ZH, Yang JM. Expression of elongation factor-2 kinase contributes to anoikis resistance and invasion of human glioma cells. Acta Pharmacol Sin 2011; 32:361-7. [PMID: 21278783 PMCID: PMC3586188 DOI: 10.1038/aps.2010.213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 11/29/2010] [Indexed: 12/29/2022]
Abstract
AIM To determine whether elongation factor-2 kinase (eEF-2 kinase) contributes to the malignant phenotype of glioblastoma multiforme by promoting the migration and invasion of glioma cells. The mechanism involved was also explored. METHODS Human glioma cell lines T98G and LN-229 were used. The expression of eEF-2 kinase was silenced using siRNA, and the invasive potential of tumor cells was assessed using a wound-healing assay and a Matrigel invasion assay. Apoptosis was determined using propidium iodide (PI) staining and Western blot analysis of cleaved caspase-3. RESULTS Silencing the expression of eEF-2 kinase by siRNA significantly suppressed both the migration and invasion of human glioma cells. Silencing eEF-2 kinase expression also sensitized glioma cells to anoikis, thereby decreasing tumor cell viability in the absence of attachment. Treatment of tumor cells with the caspase inhibitor z-VAD-fmk down-regulated Bim accumulation and abolished glioma cell sensitivity to anoikis. CONCLUSION The results suggest that the expression of eEF-2 kinase contributes to migration and invasion of human glioma cells by protecting them from anoikis. eEF-2 kinase expression may serve as a prognostic marker and a novel target for cancer therapy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Yi Zhang
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Xiao-yuan Liu
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Zheng-hong Qin
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Jin-ming Yang
- Department of Pharmacology and Penn State Hershey Cancer Institute, the Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
22
|
Loiola EC, Ventura ALM. Release of ATP from avian Müller glia cells in culture. Neurochem Int 2010; 58:414-22. [PMID: 21193002 DOI: 10.1016/j.neuint.2010.12.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 12/17/2010] [Accepted: 12/20/2010] [Indexed: 01/07/2023]
Abstract
ATP can be released from neurons and act as a neuromodulator in the nervous system. Besides neurons, cortical astrocytes also are capable of releasing ATP from acidic vesicles in a Ca(2+)-dependent way. In the present work, we investigated the release of ATP from Müller glia cells of the chick embryo retina by examining quinacrine staining and by measuring the extracellular levels of ATP in purified Müller glia cultures. Our data revealed that glial cells could be labeled with quinacrine, a reaction that was prevented by incubation of the cells with 1μM bafilomycin A1 or 2μM Evans blue, potent inhibitors of vacuolar ATPases and of the vesicular nucleotide transporter, respectively. Either 50mM KCl or 1mM glutamate was able to decrease quinacrine staining of the cells, as well as to increase the levels of ATP in the extracellular medium by 77% and 89.5%, respectively, after a 5min incubation of the cells. Glutamate-induced rise in extracellular ATP could be mimicked by 100μM kainate (81.5%) but not by 100μM NMDA in medium without MgCl(2) but with 2mM glycine. However, both glutamate- and kainate-induced increase in extracellular ATP levels were blocked by 50μM of the glutamatergic antagonists DNQX and MK-801, suggesting the involvement of both NMDA and non-NMDA receptors. Extracellular ATP accumulation induced by glutamate was also blocked by incubation of the cells with 30μM BAPTA-AM or 1μM bafilomycin A1. These results suggest that glutamate, through activation of both NMDA and non-NMDA receptors, induces the release of ATP from retinal Müller cells through a calcium-dependent exocytotic mechanism.
Collapse
Affiliation(s)
- Erick Correia Loiola
- Department of Neurobiology, Neuroscience Program, Institute of Biology, Federal Fluminense University, Niterói, RJ, Brazil
| | | |
Collapse
|
23
|
Song D, Liu X, Liu R, Yang L, Zuo J, Liu W. Connexin 43 hemichannel regulates H9c2 cell proliferation by modulating intracellular ATP and [Ca2+]. Acta Biochim Biophys Sin (Shanghai) 2010; 42:472-82. [PMID: 20705586 DOI: 10.1093/abbs/gmq047] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Connexin 43 (Cx43), known to be the main protein building blocks of gap junctions and hemichannels in mammalian heart, plays an important role in cardiocytes proliferation. Gap junctional intercellular communication has been suggested to be necessary for cellular proliferation and differentiation. However, the effect of Cx43 hemichannel on cardiocytes proliferation and the mechanism remain unclear. In this study, rat heart cell line H9c2 was used. The Cx43 location, the proliferation rate and hemichannel activity of H9c2 cells and Wnt-3a(+)-H9c2 cells were investigated and the changes of intracellular ATP and [Ca(2+)] were determined. Results showed that the inhibited hemichannel induced by 18beta-glycyrrhetinic acid (GA) evoked intracellular ATP and [Ca(2+)] increase and enhanced H9c2 cell proliferation. Wnt-3a(+)-H9c2 cells displayed enhanced hemichannel activity and proliferation rate. Inhibited hemichannel of Wnt-3a(+)-H9c2 cells induced by 18beta-GA decreased intracellular ATP, increased [Ca(2+)], and enhanced the proliferation of H9c2 cells. This study validated the role of hemichannel in H9c2 cell proliferation regulation, and showed a mechanism involved in the regulation of H9c2 cell proliferation. The proliferation could be enhanced by Cx43 hemichannel-mediated ATP release accompanying intracellular [Ca(2+)] change. However, different changes of ATP were observed in Wnt-3a(+)-H9c2 cells. These findings provided new insights into the molecular mechanisms of proliferation regulation in H9c2 cells and the effect of Wnt-3a on intracellular ATP.
Collapse
Affiliation(s)
- Dongli Song
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
24
|
Involvement of the PI3K/AKT pathway in ATP-induced proliferation of developing retinal cells in culture. Int J Dev Neurosci 2010; 28:503-11. [PMID: 20542106 DOI: 10.1016/j.ijdevneu.2010.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 06/04/2010] [Accepted: 06/06/2010] [Indexed: 01/23/2023] Open
Abstract
ATP induces the proliferation of chick retinal cells in culture through the activation of P2Y1 receptors, PKC and MAP kinases. Together with MAP kinases, the PI3K/AKT pathway has also been implicated as an important mediator in proliferative events during development. Here we investigated the participation of the PI3K/AKT signal pathway on ATP-induced proliferation of chick embryo retinal cells in culture. When retinal cultures obtained from 7-day-old embryos were cultivated for 1 day and treated with ATP, a transient and dose-dependent phosphorylation of both ERK and AKT was observed, an effect that could be mimicked by 500 microM ADP and blocked by 100 microM PPADS, a P2 receptor antagonist. Maximal stimulation of both enzymes was obtained with 100 microM ATP in 5 min, decreasing thereafter. Activation of these pathways by ATP seemed to be independent, since LY294002 and U0126, inhibitors of PI3K and MEK, did not block the activation of ERK and AKT, respectively, although each compound blocked its respective target. Moreover, when the cultures were incubated with ATP in the presence of LY294002, a decreased incorporation of [(3)H]-thymidine was observed, as compared to cultures treated only with ATP, a decline that was also obtained by incubating the cells with ATP plus 0.5 microM API-59CJ-Ome, an inhibitor of AKT. No decrease in cell viability was observed with this concentration of API-59CJ-Ome. An increase in cyclin D1 expression, that could be inhibited by 10 microM LY 294002 or 20 microM U0126, was observed when cells were incubated with 500 microM ADP. No effect of PI3K and MEK inhibitors was observed in the expression of p27kip1 in the cultures. These results suggest that, besides the involvement of the MAP kinases pathway, ATP-induced cell cycling of late developing retinal progenitors in culture also involves the activation of the PI3K/AKT pathway.
Collapse
|
25
|
Sholl-Franco A, Fragel-Madeira L, Macama ADCC, Linden R, Ventura ALM. ATP controls cell cycle and induces proliferation in the mouse developing retina. Int J Dev Neurosci 2009; 28:63-73. [PMID: 19799993 DOI: 10.1016/j.ijdevneu.2009.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 06/30/2009] [Accepted: 09/20/2009] [Indexed: 01/20/2023] Open
Abstract
Previous data suggest that nucleotides are important mitogens in the developing chick retina. Here, we extended the study on the mitogenic effect of ATP to newborn mouse retinal explants. Our results showed that P2Y(1) receptors were widely distributed in C57bl/6 mice retina and that the majority of PCNA positive cells co-localized with P2Y(1) receptor. To evaluate proliferation, retinal explants obtained from newborn mice were incubated with 0.5 microCi [(3)H]-thymidine or 3 microM BrDU 1h before the end of culture. Our data showed that ATP induced a dose-dependent increase in [(3)H]-thymidine incorporation, an effect that was mimicked by ADP but not by UTP and was blocked by the P2 antagonist PPADS in a dose-dependent manner. The increase in [(3)H]-thymidine incorporation induced by ATP was only observed in explants cultured for 3 days or less and was mimicked by the ectoapyrase inhibitor ARL 67156. It corresponded to an increase in the number of BrdU(+) cells in the neuroblastic layer (NL) of the tissue, suggesting that ATP, through activation of P2Y(1) receptors, induced proliferation of late developing progenitors in retinal explants of newborn mice. The increase in the number of BrdU(+) cells was observed across the whole NL when explants were incubated with ATP for 24h and no increase in the number of p-histone H3 labeled cells could be noticed at this time point. In longer incubations of 48h with ATP or 24h with ATP followed by a period of 24h in fresh medium, an increase in the number of BrdU(+) cells promoted by ATP was observed only in the middle and outer, but not in the inner NL. In these conditions, an increase in the number of p-histone H3 labeled cells was detected in the outer NL, suggesting that ATP induced cells to enter S and progress to G2 phase of the cell cycle in the first 24h period of incubation. ATP also induced an increase and a decrease in the expression of cyclin D1 and p27(kip1), respectively, in retinal progenitors of the NL. While the increase in the expression of cyclin D1 was observed when retinal explants were incubated for 3h or longer periods of time, the decrease in the expression of p27(kip1) was noticed only after 6h incubation with ATP. Both effects were blocked by the P2 receptor antagonist PPADS. These data suggest that ATP induces cell proliferation in retinal explants by inducing late developing progenitors to progress from G1 to S phase of cell cycle.
Collapse
Affiliation(s)
- Alfred Sholl-Franco
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21949-900, Brazil.
| | | | | | | | | |
Collapse
|
26
|
Battista AG, Ricatti MJ, Pafundo DE, Gautier MA, Faillace MP. Extracellular ADP regulates lesion-induced in vivo cell proliferation and death in the zebrafish retina. J Neurochem 2009; 111:600-13. [PMID: 19694906 DOI: 10.1111/j.1471-4159.2009.06352.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Regeneration and growth that occur in the adult teleost retina by neurogenesis have been helpful in identifying molecular and cellular mechanisms underlying cell proliferation and differentiation. In this report, we demonstrate that endogenous purinergic signals regulate cell proliferation induced by a cytotoxic injury of the adult zebrafish retina which mainly damages inner retinal layers. Particularly, we found that ADP but not ATP or adenosine significantly enhanced cell division as assessed by 5-bromo-2'-deoxyuridine incorporation following injury, during the degenerative and proliferative phase of the regeneration process. This effect of ADP occurs via P2Y1 metabotropic receptors as shown by intra-ocular injection of selective antagonists. Additionally, we describe a role for purinergic signals in regulating cell death induced by injury. Scavenging of extracellular nucleotides significantly increased cell death principally seen in the inner retinal layers. This effect is partially reproduced by blocking P2Y1 receptors suggesting a neuroprotective function for ADP, which is derived from extracellular ATP probably released by dying cells as a consequence of the ouabain treatment. This study demonstrates a crucial role for ADP as a paracrine signal in the repair of retinal tissue following injury.
Collapse
Affiliation(s)
- Ariadna G Battista
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
27
|
Bringmann A, Iandiev I, Pannicke T, Wurm A, Hollborn M, Wiedemann P, Osborne NN, Reichenbach A. Cellular signaling and factors involved in Müller cell gliosis: neuroprotective and detrimental effects. Prog Retin Eye Res 2009; 28:423-51. [PMID: 19660572 DOI: 10.1016/j.preteyeres.2009.07.001] [Citation(s) in RCA: 559] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Müller cells are active players in normal retinal function and in virtually all forms of retinal injury and disease. Reactive Müller cells protect the tissue from further damage and preserve tissue function by the release of antioxidants and neurotrophic factors, and may contribute to retinal regeneration by the generation of neural progenitor/stem cells. However, Müller cell gliosis can also contribute to neurodegeneration and impedes regenerative processes in the retinal tissue by the formation of glial scars. This article provides an overview of the neuroprotective and detrimental effects of Müller cell gliosis, with accounts on the cellular signal transduction mechanisms and factors which are implicated in Müller cell-mediated neuroprotection, immunomodulation, regulation of Müller cell proliferation, upregulation of intermediate filaments, glial scar formation, and the generation of neural progenitor/stem cells. A proper understanding of the signaling mechanisms implicated in gliotic alterations of Müller cells is essential for the development of efficient therapeutic strategies that increase the supportive/protective and decrease the destructive roles of gliosis.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Liebigstrasse 10-14, D-04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Housley GD, Bringmann A, Reichenbach A. Purinergic signaling in special senses. Trends Neurosci 2009; 32:128-41. [DOI: 10.1016/j.tins.2009.01.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 12/22/2008] [Accepted: 01/05/2009] [Indexed: 02/06/2023]
|
29
|
Abbracchio MP, Burnstock G, Verkhratsky A, Zimmermann H. Purinergic signalling in the nervous system: an overview. Trends Neurosci 2008; 32:19-29. [PMID: 19008000 DOI: 10.1016/j.tins.2008.10.001] [Citation(s) in RCA: 624] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/02/2008] [Accepted: 10/03/2008] [Indexed: 12/15/2022]
Abstract
Purinergic receptors, represented by several families, are arguably the most abundant receptors in living organisms and appeared early in evolution. After slow acceptance, purinergic signalling in both peripheral and central nervous systems is a rapidly expanding field. Here, we emphasize purinergic co-transmission, mechanisms of release and breakdown of ATP, ion channel and G-protein-coupled-receptor subtypes for purines and pyrimidines, the role of purines and pyrimidines in neuron-glial communication and interactions of this system with other transmitter systems. We also highlight recent data involving purinergic signalling in pathological conditions, including pain, trauma, ischaemia, epilepsy, migraine, psychiatric disorders and drug addiction, which we expect will lead to the development of therapeutic strategies for these disorders with novel mechanisms of action.
Collapse
Affiliation(s)
- Maria P Abbracchio
- Department of Pharmacological Sciences, Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, via Balzaretti 9, University of Milan, 20133-Milan, Italy
| | | | | | | |
Collapse
|
30
|
Abstract
Purinergic signalling plays a major role in the function of every organ including the brain. A growing body of evidence also suggests that purinergic signalling is important in the development of the retina, cochlea and neocortex. In these three contexts release of ATP through the spontaneous gating of connexin hemichannels in cells, respectively, of the retinal pigment epithelium, Köllicker's organ, and the radial glia triggers waves of intracellular Ca(2+) release. In the case of the developing retina and cortex, the released ATP acts to control proliferation of neuronal precursor cells, while in the cochlea it coordinates the spontaneous activity of adjacent hair cells to refine the tonotopic maps in the cochlear nucleus. Recently ATP-derived ADP signalling has been implicated at the very earliest stages of development, notably in triggering the gene expression necessary for formation of the eye. It is now timely to test the extent to which connexin hemichannel-mediated ATP release and accompanying Ca(2+) waves contribute to all stages of development.
Collapse
Affiliation(s)
- Nicholas Dale
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
31
|
Müller glia as an active compartment modulating nervous activity in the vertebrate retina: neurotransmitters and trophic factors. Neurochem Res 2008; 33:1466-74. [PMID: 18273703 DOI: 10.1007/s11064-008-9604-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 01/23/2008] [Indexed: 01/13/2023]
Abstract
Müller cells represent the main type of glia present in the retina interacting with most, if not all neurons in this tissue. Müller cells have been claimed to function as optic fibers in the retina delivering light to photoreceptors with minimal distortion and low loss [Franze et al (2007) Proc Natl Acad Sci 104:8287-8292]. Most of the mediators found in the brain are also detected in the retinal tissue, and glia cells are active players in the synthesis, release, signaling and uptake of major mediators of synaptic function. Müller glia trophic factors may regulate many different aspects of neuronal circuitry during synaptogenesis, differentiation, neuroprotection and survival of photoreceptors, Retinal Ganglion Cells (RGCs) and other targets in the retina. Here we review the role of several transmitters and trophic factors that participate in the neuron-glia loop in the retina.
Collapse
|