Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1125962, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36967764

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, and current treatment options are unsatisfactory on the long term. Several studies suggest a potential neuroprotective action by female hormones, especially estrogens. The potential role of progestogens, however, is less defined, and no studies have investigated the potential involvement of membrane progesterone receptors (mPRs). In the present study, the putative neuroprotective role for mPRs was investigated in SH-SY5Y cells, using two established pharmacological treatments for cellular PD models, 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenylpyridinium (MPP+). Our results show that both the physiologic agonist progesterone and the specific mPR agonist Org OD 02-0 were effective in reducing SH-SY5Y cell death induced by 6-OHDA and MPP+, whereas the nuclear PR agonist promegestone (R5020) and the GABAA receptor agonist muscimol were ineffective. Experiments performed with gene silencing technology and selective pharmacological agonists showed that mPRα is the isoform responsible for the neuroprotective effects we observed. Further experiments showed that the PI3K-AKT and MAP kinase signaling pathways are involved in the mPRα-mediated progestogen neuroprotective action in SH-SY5Y cells. These findings suggest that mPRα could play a neuroprotective role in PD pathology and may be a promising target for the development of therapeutic strategies for PD prevention or management.


Assuntos
Doença de Parkinson , Receptores de Progesterona , Humanos , Oxidopamina , Doença de Parkinson/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Progesterona/farmacologia , Progestinas/farmacologia , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
2.
J Mol Neurosci ; 72(10): 2067-2080, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35974286

RESUMO

We recently showed that membrane progesterone receptor α (mPRα/PAQR7) promotes pro-regenerative effects in Schwann cell-like adipose stem cells (SCL-ASC), an alternative model to Schwann cells for the promotion of peripheral nerve regeneration. In this study, we investigated how mPRα activation with the mPR-specific agonist Org OD 02-0 in SCL-ASC affected regenerative parameters in two neuronal cell lines, IMR-32 and SH-SY-5Y. In a series of conditioned medium experiments, we found that mPR activation of SCL-ASC led to increased neurite outgrowth, protection from cell death and increased expression of peripheral nerve regeneration markers (CREB3, ATF3, GAP43) in neuronal cell lines. These effects were stronger than the ones observed with the conditioned medium from untreated SCL-ASC. The addition of Org OD 02-0 to the untreated cell medium mimicked the effects of mPR activation of SCL-ASC on cell death, but not on neurite outgrowth. Therefore, the effect of Org OD 02-0 on neurite outgrowth is SCL-ASC-dependent, while its effect on cell survivability is likely due to the direct activation of mPRs on neuronal cells. SCL-ASC transfection with mPRα siRNA showed that this isoform is responsible for the beneficial effect on neurite outgrowth. Further experiments showed that SCL-ASC-dependent outcomes likely involved the release of BDNF and IGF-2 from these cells. The beneficial mPRα effect on neurite outgrowth was confirmed in co-culture conditions. These findings strengthen the hypothesis that mPRα could play a pro-regenerative role in SCL-ASC and be a therapeutic target for the promotion of peripheral nerve regeneration.


Assuntos
Progesterona , Receptores de Progesterona , Humanos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Progesterona/farmacologia , Fator de Crescimento Insulin-Like II/metabolismo , Meios de Cultivo Condicionados/farmacologia , RNA Interferente Pequeno/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Crescimento Neuronal , Células de Schwann , Células-Tronco/metabolismo
3.
Endocrinology ; 163(11)2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36041040

RESUMO

Gender differences in a wide variety of physiological parameters have implicated the ovarian hormones, estrogens and progesterone, in the regulation of numerous nonreproductive tissue functions. Rapid, nongenomic (nonclassical) progesterone actions mediated by membrane progesterone receptors (mPRs), which belong to the progestin and adipoQ receptor family, have been extensively investigated in reproductive and nonreproductive tissues since their discovery in fish ovaries 20 years ago. The 5 mPR subtypes (α, ß, γ, δ, ε) are widely distributed in vertebrate tissues and are often expressed in the same cells as the nuclear progesterone receptor (PR) and progesterone receptor membrane component 1, thereby complicating investigations of mPR-specific functions. Nevertheless, mPR-mediated progesterone actions have been identified in a wide range of reproductive and nonreproductive tissues and distinguished from nuclear PR-mediated ones by knockdown of these receptors with siRNA in combination with a pharmacological approach using mPR- and PR-specific agonists. There are several recent reviews on the roles of the mPRs in vertebrate reproduction and cancer, but there have been no comprehensive assessments of mPR functions in nonreproductive tissues. Therefore, this article briefly reviews mPR functions in a broad range of nonreproductive tissues. The evidence that mPRs mediate progesterone and progestogen effects on neuroprotection, lordosis behavior, respiratory control of apnea, olfactory responses to pheromones, peripheral nerve regeneration, regulation of prolactin secretion in prolactinoma, immune functions, and protective functions in vascular endothelial and smooth muscle cells is critically reviewed. The ubiquitous expression of mPRs in vertebrate tissues suggests mPRs regulate many additional nonreproductive functions that remain to be identified.


Assuntos
Neoplasias Hipofisárias , Receptores de Progesterona , Animais , Membrana Celular/metabolismo , Estrogênios/farmacologia , Feromônios/metabolismo , Feromônios/farmacologia , Neoplasias Hipofisárias/metabolismo , Progesterona/metabolismo , Progesterona/farmacologia , Progestinas/metabolismo , Prolactina/metabolismo , RNA Interferente Pequeno , Receptores de Progesterona/metabolismo
4.
J Physiol ; 599(17): 4065-4084, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34174096

RESUMO

KEY POINTS: GABA depolarized sural nerve axons and increased the electrical excitability of C-fibres via GABAA receptor. Axonal excitability responses to GABA increased monotonically with the rate of action potential firing. Action potential activity in unmyelinated C-fibres is coupled to Na-K-Cl cotransporter type 1 (NKCC1) loading of axonal chloride. Activation of axonal GABAA receptor stabilized C-fibre excitability during prolonged low frequency (2.5 Hz) firing. NKCC1 maintains intra-axonal chloride to provide feed-forward stabilization of C-fibre excitability and thus support sustained firing. ABSTRACT: GABAA receptor (GABAA R)-mediated depolarization of dorsal root ganglia (DRG) axonal projections in the spinal dorsal horn is implicated in pre-synaptic inhibition. Inhibition, in this case, is predicated on an elevated intra-axonal chloride concentration and a depolarizing GABA response. In the present study, we report that the peripheral axons of DRG neurons are also depolarized by GABA and this results in an increase in the electrical excitability of unmyelinated C-fibre axons. GABAA R agonists increased axonal excitability, whereas GABA excitability responses were blocked by GABAA R antagonists and were absent in mice lacking the GABAA R ß3 subunit selectively in DRG neurons (AdvillinCre or snsCre ). Under control conditions, excitability responses to GABA became larger at higher rates of electrical stimulation (0.5-2.5 Hz). However, during Na-K-Cl cotransporter type 1 (NKCC1) blockade, the electrical stimulation rate did not affect GABA response size, suggesting that NKCC1 regulation of axonal chloride is coupled to action potential firing. To examine this, activity-dependent conduction velocity slowing (activity-dependent slowing; ADS) was used to quantify C-fibre excitability loss during a 2.5 Hz challenge. ADS was reduced by GABAA R agonists and exacerbated by either GABAA R antagonists, ß3 deletion or NKCC1 blockade. This illustrates that activation of GABAA R stabilizes C-fibre excitability during sustained firing. We posit that NKCC1 acts in a feed-forward manner to maintain an elevated intra-axonal chloride in C-fibres during ongoing firing. The resulting chloride gradient can be utilized by GABAA R to stabilize axonal excitability. The data imply that therapeutic strategies targeting axonal chloride regulation at peripheral loci of pain and itch may curtail aberrant firing in C-fibres.


Assuntos
Axônios , Fibras Nervosas Amielínicas , Animais , Camundongos , Membro 2 da Família 12 de Carreador de Soluto , Membro 3 da Família 12 de Carreador de Soluto , Simportadores , Ácido gama-Aminobutírico , Cotransportadores de K e Cl-
5.
Mol Cell Endocrinol ; 531: 111298, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33930460

RESUMO

Membrane progesterone receptors (mPRs) were recently found to be present and active in Schwann cells, where they have a potentially pro-regenerative activity. In this study, we investigated the role of mPRs in human adipose stem cells (ASC) differentiated into Schwann cell-like cells (SCL-ASC), which represent a promising alternative to Schwann cells for peripheral nerve regeneration. Our findings show that mPRs are present both in undifferentiated and differentiated ASC, and that the differentiation protocol upregulates mPR expression. Activation of mPRα promoted cell migration and differentiation in SCL-ASC, alongside with changes in cell morphology and mPRα localization. Moreover, it increased the expression and release of BDNF, a neurotrophin with pro-regenerative activity. Further analysis showed that Src and PI3K-Akt signaling pathways are involved in mPRα activity in SCL-ASC. These findings suggest that mPRα could play a pro-regenerative role in SCL-ASC and may be a promising target for the promotion of peripheral nerve regeneration.


Assuntos
Adipócitos/citologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Células de Schwann/citologia , Adipócitos/metabolismo , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Cultura Primária de Células , Regeneração , Células de Schwann/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Regulação para Cima
7.
Cells ; 9(8)2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32796542

RESUMO

Protein kinase type C-ε (PKCε) plays important roles in the sensitization of primary afferent nociceptors, such as ion channel phosphorylation, that in turn promotes mechanical hyperalgesia and pain chronification. In these neurons, PKCε is modulated through the local release of mediators by the surrounding Schwann cells (SCs). The progesterone metabolite allopregnanolone (ALLO) is endogenously synthesized by SCs, whereas it has proven to be a crucial mediator of neuron-glia interaction in peripheral nerve fibers. Biomolecular and pharmacological studies on rat primary SCs and dorsal root ganglia (DRG) neuronal cultures were aimed at investigating the hypothesis that ALLO modulates neuronal PKCε, playing a role in peripheral nociception. We found that SCs tonically release ALLO, which, in turn, autocrinally upregulated the synthesis of the growth factor brain-derived neurotrophic factor (BDNF). Subsequently, glial BDNF paracrinally activates PKCε via trkB in DRG sensory neurons. Herein, we report a novel mechanism of SCs-neuron cross-talk in the peripheral nervous system, highlighting a key role of ALLO and BDNF in nociceptor sensitization. These findings emphasize promising targets for inhibiting the development and chronification of neuropathic pain.


Assuntos
Comunicação Autócrina/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neuralgia/metabolismo , Comunicação Parácrina/fisiologia , Pregnanolona/metabolismo , Células de Schwann/metabolismo , Animais , Comunicação Autócrina/genética , Western Blotting , Células Cultivadas , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Gânglios Espinais/metabolismo , Humanos , Hiperalgesia/metabolismo , Comunicação Parácrina/genética , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo , Espectrometria de Massas em Tandem
8.
J Mol Neurosci ; 70(3): 433-448, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31749125

RESUMO

Several studies in the last decade demonstrated that progestogens play an important role in the biology of Schwann cells, the main neuroglial cells of the peripheral nervous system. Since a recent study showed that the S42 rat Schwann cell line expressed membrane progesterone receptors (mPRs), members of the PAQR family, in this study, we examined mPR expression in a more physiological model, primary rat Schwann cells. We demonstrated that primary rat Schwann cells show a different pattern of mPR expression compared to the previously studied model; mPRα (PAQR7) and ß (PAQR8) isoforms were the major mPR members identified, with different sub-cellular localizations. Activation of the nuclear progesterone receptor (PR) with the specific agonist R5020 upregulated mPR expression, while activation of mPRs with the specific agonist Org OD 02-0 changed their sub-cellular localization. An in-depth analysis revealed additional effects of mPR activation, such as AKT activation, reduced expression of the myelin-associated glycoprotein (MAG), morphological changes, altered expression of several Schwann cell differentiation markers, and increased Schwann cell migration and proliferation. In conclusion, we identified mPRα and mPRß in primary rat Schwann cells, and our findings suggest a putative role for mPRs in the regulation of Schwann cell migration, proliferation, and differentiation. Therefore, mPRs are a potential pharmacological target for Schwann cell-related disorders and neurodegenerative diseases, especially those in which Schwann cell-mediated axon remyelination is desirable.


Assuntos
Diferenciação Celular , Movimento Celular , Proliferação de Células , Receptores de Progesterona/metabolismo , Células de Schwann/metabolismo , Animais , Células Cultivadas , Feminino , Glicoproteína Associada a Mielina/genética , Glicoproteína Associada a Mielina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/genética , Células de Schwann/citologia , Células de Schwann/fisiologia
9.
Steroids ; 142: 6-13, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-28962850

RESUMO

The role played by progestogens in modulating Schwann cell pathophysiology is well established. Progestogens exert their effects in these cells through both classical genomic and non-genomic mechanisms, the latter mediated by the GABA-A receptor. However, there is evidence that other receptors may be involved. Membrane progesterone receptors (mPRs) are novel 7-transmembrane receptors coupled to G proteins that have been characterized in different tissues and cells, including the central nervous system (CNS). The mPRs were shown to mediate some of progestogens' neuroprotective effects in the CNS, and to be upregulated in glial cells after traumatic brain injury. Based on this evidence, this paper investigated the possible involvement of mPRs in mediating progestogen actions in S42 Schwann cells. All five mPR isoforms and progesterone receptor membrane component 1 (PGRMC1) were detected in Schwann cells, and were present on the cell membrane. Progesterone and the mPR-specific agonist, Org-OD-02-0 (02) bound to these membranes, indicating the presence of functional mPRs. The mPR agonist 02 rapidly increased cell migration in an in vitro assay, suggesting a putative role of mPRs in the nerve regeneration process. Treatment with pertussis toxin and 8-Br-cAMP blocked 02-induced cell migration, suggesting this progestogen action is mediated by activation of an inhibitory G protein, leading to a decrease in intracellular cAMP levels. In contrast, long-term mPR activation led to increased expression levels of myelin associated glycoprotein (MAG). Taken together, these findings show that mPRs are present and active in Schwann cells and have a role in modulating their physiological processes.


Assuntos
Membrana Celular/metabolismo , Movimento Celular , Glicoproteína Associada a Mielina/biossíntese , Neuroglia/citologia , Neuroglia/metabolismo , Receptores de Progesterona/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Neuroglia/efeitos dos fármacos , Toxina Pertussis/farmacologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Ratos , Receptores de Progesterona/agonistas , Receptores de Progesterona/análise , Tionucleotídeos/farmacologia , Células Tumorais Cultivadas
10.
Front Cell Neurosci ; 6: 68, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23335881

RESUMO

γ-aminobutyric acid type B (GABA-B) receptor mediates the inhibitory transmission of γ-aminobutyric acid in the mammalian nervous system, being present in neurons and also in glial cells. Recently the presence of GABA-B has been demonstrated in Schwann cells (SC) suggesting its contribution in regulating the cell fate, maturation, and plasticity. Here, we further support the functional presence of GABA-B receptor in SC plasma membrane. By confocal microscopy immunofluorescence we provide evidences that GABA-B localization on the cell elongated processes correlates with the morphological changes occurring in the differentiated SC. In vivo most of the GABA-B receptors seem to be present in non-myelinating SC, which are committed to ensheath the nociceptive fibers. Therefore, we argue that GABA-B receptors do not control exclusively the in vivo differentiation yielding the myelinating SC, but are also fundamental in regulating the SC plasticity versus the non-myelinating state. Data from the literature and our recent findings corroborate the role of the GABAergic system and GABA-B receptors in the peripheral nervous system, opening new perspectives on the mechanisms controlling the differentiation of SC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...