Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.018
Filtrar
1.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39273121

RESUMO

Traumatic spinal cord injury (tSCI) has complex pathophysiological events that begin after the initial trauma. One such event is fibroglial scar formation by fibroblasts and reactive astrocytes. A strong inhibition of axonal growth is caused by the activated astroglial cells as a component of fibroglial scarring through the production of inhibitory molecules, such as chondroitin sulfate proteoglycans or myelin-associated proteins. Here, we used neural precursor cells (aldynoglia) as promoters of axonal growth and a fibrin hydrogel gelled under alkaline conditions to support and guide neuronal cell growth, respectively. We added Tol-51 sulfoglycolipid as a synthetic inhibitor of astrocyte and microglia in order to test its effect on the axonal growth-promoting function of aldynoglia precursor cells. We obtained an increase in GFAP expression corresponding to the expected glial phenotype for aldynoglia cells cultured in alkaline fibrin. In co-cultures of dorsal root ganglia (DRG) and aldynoglia, the axonal growth promotion of DRG neurons by aldynoglia was not affected. We observed that the neural precursor cells first clustered together and then formed niches from which aldynoglia cells grew and connected to groups of adjacent cells. We conclude that the combination of alkaline fibrin with synthetic sulfoglycolipid Tol-51 increased cell adhesion, cell migration, fasciculation, and axonal growth capacity, promoted by aldynoglia cells. There was no negative effect on the behavior of aldynoglia cells after the addition of sulfoglycolipid Tol-51, suggesting that a combination of aldynoglia plus alkaline fibrin and Tol-51 compound could be useful as a therapeutic strategy for tSCI repair.


Assuntos
Axônios , Fibrina , Gânglios Espinais , Animais , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/citologia , Axônios/metabolismo , Axônios/efeitos dos fármacos , Fibrina/metabolismo , Hidrogéis/química , Hidrogéis/farmacologia , Ratos , Glicolipídeos/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Medula Espinal/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/citologia , Movimento Celular/efeitos dos fármacos
2.
BMC Neurosci ; 25(1): 44, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39278931

RESUMO

BACKGROUND: Peripheral neuropathy (PN) constitutes a dose-limiting side effect of oxaliplatin chemotherapy that often compromises the efficacy of antineoplastic treatments. Sensory neurons damage in dorsal root ganglia (DRG) are the cellular substrate of PN complex molecular origin. Dehydropeptidase-1 (DPEP1) inhibitors have shown to avoid platin-induced nephrotoxicity without compromising its anticancer efficiency. The objective of this study was to describe DPEP1 expression in rat DRG in health and in early stages of oxaliplatin toxicity. To this end, we produced and characterized anti-DPEP1 polyclonal antibodies and used them to define the expression, and cellular and subcellular localization of DPEP1 by immunohistochemical confocal microscopy studies in healthy controls and short term (six days) oxaliplatin treated rats. RESULTS: DPEP1 is expressed mostly in neurons and in glia, and to a lesser extent in endothelial cells. Rats undergoing oxaliplatin treatment developed allodynia. TNF-𝛼 expression in DRG revealed a pattern of focal and at different intensity levels of neural cell inflammatory damage, accompanied by slight variations in DPEP1 expression in endothelial cells and in nuclei of neurons. CONCLUSIONS: DPEP1 is expressed in neurons, glia and endothelial cells of DRG. Oxaliplatin caused allodynia in rats and increased TNF-α expression in DRG neurons. The expression of DPEP1 in neurons and other cells of DRG suggest this protein as a novel strategic molecular target in the prevention of oxaliplatin-induced acute neurotoxicity.


Assuntos
Antineoplásicos , Gânglios Espinais , Oxaliplatina , Doenças do Sistema Nervoso Periférico , Animais , Oxaliplatina/toxicidade , Gânglios Espinais/metabolismo , Gânglios Espinais/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/prevenção & controle , Doenças do Sistema Nervoso Periférico/patologia , Masculino , Antineoplásicos/toxicidade , Ratos , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Hiperalgesia/prevenção & controle , Fator de Necrose Tumoral alfa/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Ratos Sprague-Dawley , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Inflamação/metabolismo , Inflamação/induzido quimicamente
3.
J Neurophysiol ; 132(3): 1056-1073, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39110896

RESUMO

Besides having high potency and efficacy at the µ-opioid (MOR) and other opioid receptor types, fentanyl has some affinity for some adrenergic receptor types, which may underlie its unique pathophysiological differences from typical opioids. To better understand the unique actions of fentanyl, we assessed the extent to which fentanyl alters striatal medium spiny neuron (MSN) activity via opioid receptors or α1-adrenoceptors in dopamine type 1 or type 2 receptor (D1 or D2)-expressing MSNs. In neuronal and mixed-glial cocultures from the striatum, acute fentanyl (100 nM) exposure decreased the frequency of spontaneous action potentials. Overnight exposure of cocultures to 100 nM fentanyl severely reduced the proportion of MSNs with spontaneous action potentials, which was unaffected by coexposure to the opioid receptor antagonist naloxone (10 µM) but fully negated by coadministering the pan-α1-adrenoceptor inverse agonist prazosin (100 nM) and partially reversed by the selective α1A-adrenoceptor antagonist RS 100329 (300 nM). Acute fentanyl (100 nM) exposure modestly reduced the frequency of action potentials and caused firing rate adaptations in D2, but not D1, MSNs. Prolonged (2-5 h) fentanyl (100 nM) application dramatically attenuated firing rates in both D1 and D2 MSNs. To identify possible cellular sites of α1-adrenoceptor action, α1-adrenoceptors were localized in subpopulations of striatal astroglia and neurons by immunocytochemistry and Adra1a mRNA by in situ hybridization in astrocytes. Thus, sustained fentanyl exposure can inhibit striatal MSN activity via a nonopioid receptor-dependent pathway, which may be modulated via complex actions in α1-adrenoceptor-expressing striatal neurons and/or glia.NEW & NOTEWORTHY Acute fentanyl exposure attenuated the activity of striatal medium spiny neurons (MSNs) in vitro and in dopamine D2, but not D1, receptor-expressing MSNs in ex vivo slices. By contrast, sustained fentanyl exposure suppressed the spontaneous activity of MSNs cocultured with glia through a nonopioid receptor-dependent mechanism modulated, in part, by α1-adrenoceptors. Fentanyl exposure can affect striatal function via a nonopioid receptor mechanism of action that appears mediated by α1-adrenoreceptor-expressing striatal neurons and/or astroglia.


Assuntos
Potenciais de Ação , Analgésicos Opioides , Técnicas de Cocultura , Corpo Estriado , Fentanila , Neuroglia , Neurônios , Animais , Fentanila/farmacologia , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Camundongos , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/fisiologia , Analgésicos Opioides/farmacologia , Potenciais de Ação/efeitos dos fármacos , Receptores Opioides/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D2/efeitos dos fármacos , Masculino , Antagonistas de Entorpecentes/farmacologia , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/antagonistas & inibidores , Células Cultivadas
4.
Chemosphere ; 363: 142993, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39097108

RESUMO

Due to their extensive use, the release of zinc oxide nanoparticles (ZnO NP) into the environment is increasing and may lead to unintended risk to both human health and ecosystems. Access of ZnO NP to the brain has been demonstrated, so their potential toxicity on the nervous system is a matter of particular concern. Although evaluation of ZnO NP toxicity has been reported in several previous studies, the specific effects on the nervous system are not completely understood and, particularly, effects on genetic material and on organism behaviour are poorly addressed. We evaluated the potential toxic effects of ZnO NP in vitro and in vivo, and the role of zinc ions (Zn2+) in these effects. In vitro, the ability of ZnO NP to be internalized by A172 glial cells was verified, and the cytotoxic and genotoxic effects of ZnO NP or the released Zn2+ ions were addressed by means of vital dye exclusion and comet assay, respectively. In vivo, behavioural alterations were evaluated in zebrafish embryos using a total locomotion assay. ZnO NP induced decreases in viability of A172 cells after 24 h of exposure and genetic damage after 3 and 24 h. The involvement of the Zn2+ ions released from the NP in genotoxicity was confirmed. ZnO NP exposure also resulted in decreased locomotor activity of zebrafish embryos, with a clear role of released Zn2+ ions in this effect. These findings support the toxic potential of ZnO NP showing, for the first time, genetic effects on glial cells and proving the intervention of Zn2+ ions.


Assuntos
Peixe-Zebra , Óxido de Zinco , Óxido de Zinco/toxicidade , Animais , Humanos , Nanopartículas Metálicas/toxicidade , Dano ao DNA , Sobrevivência Celular/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Ensaio Cometa , Neuroglia/efeitos dos fármacos , Nanopartículas/toxicidade
5.
Int J Mol Sci ; 25(16)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39201378

RESUMO

Endoplasmic reticulum (ER) stress is a significant player in the pathophysiology of various neurodegenerative and neuropsychiatric disorders. Despite the established link between ER stress and inflammatory pathways, there remains a need for deeper exploration of the specific cellular mechanisms underlying ER stress-mediated neuroinflammation. This study aimed to investigate how the severity of ER stress (triggered by different concentrations of tunicamycin) can impact the release of proinflammatory cytokines IL-6 and IL-8 from astrocytes and microglia, comparing the effects with those induced by well-known immunostimulants-tumor necrosis factor alpha (TNF-α) or lipopolysaccharide (LPS). Mild ER stress has a distinct effect on the cytokine release compared to more intense stress levels, i.e., diminished IL-6 production was accompanied by an increase in IL-8 level, which was significantly more pronounced in astrocytes than in microglia. On the contrary, prolonged or more severe ER stress induced inflammation in glial cells, leading to a time- and concentration-dependent buildup of proinflammatory IL-6, but unlike inflammatory agents, an ER stress inducer diminished IL-8 secretions by glial cells. The differences could hold importance in identifying ER stress markers as potential drug targets for the treatment of neurodegenerative diseases or mood disorders, yet this requires confirmation in more complex animal studies.


Assuntos
Astrócitos , Estresse do Retículo Endoplasmático , Interleucina-6 , Interleucina-8 , Neuroglia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Interleucina-8/metabolismo , Interleucina-6/metabolismo , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Tunicamicina/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas
6.
Nat Commun ; 15(1): 7028, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39147742

RESUMO

Alzheimer's disease (AD) is the leading cause of dementia in older adults, and the need for effective, sustainable therapeutic targets is imperative. The complement pathway has been proposed as a therapeutic target. C5aR1 inhibition reduces plaque load, gliosis, and memory deficits in animal models, however, the cellular bases underlying this neuroprotection were unclear. Here, we show that the C5aR1 antagonist PMX205 improves outcomes in the Arctic48 mouse model of AD. A combination of single cell and single nucleus RNA-seq analysis of hippocampi derived from males and females identified neurotoxic disease-associated microglia clusters in Arctic mice that are C5aR1-dependent, while microglial genes associated with synapse organization and transmission and learning were overrepresented in PMX205-treated mice. PMX205 also reduced neurotoxic astrocyte gene expression, but clusters associated with protective responses to injury were unchanged. C5aR1 inhibition promoted mRNA-predicted signaling pathways between brain cell types associated with cell growth and repair, while suppressing inflammatory pathways. Finally, although hippocampal plaque load was unaffected, PMX205 prevented deficits in short-term memory in female Arctic mice. In conclusion, C5aR1 inhibition prevents cognitive loss, limits detrimental glial polarization while permitting neuroprotective responses, as well as leaving most protective functions of complement intact, making C5aR1 antagonism an attractive therapeutic strategy for AD.


Assuntos
Doença de Alzheimer , Modelos Animais de Doenças , Hipocampo , Microglia , Receptor da Anafilatoxina C5a , Transdução de Sinais , Animais , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Receptor da Anafilatoxina C5a/metabolismo , Receptor da Anafilatoxina C5a/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Camundongos , Feminino , Masculino , Transdução de Sinais/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Camundongos Transgênicos , Humanos , Camundongos Endogâmicos C57BL
7.
Biomed Pharmacother ; 179: 117290, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39153433

RESUMO

Hydrogen sulfide (H2S) is a gaseous signaling molecule that influences digestive and nervous system functions. Enteric glial cells (EGCs) are integral to the enteric nervous system and play a role in regulating gastrointestinal motility. This study explored the dual effects of exogenous H2S on EGCs and the influence of apoptosis-related pathways and ion channels in EGCs. We also administered honokiol for further interventional studies. The results revealed that low-concentration H2S increased the mitochondrial membrane potential (MMP) of EGCs, decreased the whole-cell membrane potential, downregulated BAX and caspase-3, upregulated Bcl2 expression, reduced apoptosis, and promoted cell proliferation. The Ca2+ concentration, Cx43 mRNA, and protein expression were also increased. A high concentration of H2S had the opposite effect. In addition, GFAP mRNA expression was upregulated in the test-low group, downregulated in the test-high group, and upregulated in the test-high + Hon group. Honokiol treatment increased MMP, reduced whole-cell membrane potential, inhibited BAX and caspase-3 expression, increased Bcl2 expression, decreased cell apoptosis, and increased cell proliferation. The Ca2+ concentration, Cx43 mRNA, and protein expression were also upregulated. In conclusion, our study showed that exogenous H2S can bidirectionally regulate EGC proliferation and apoptosis by affecting MMP and cell membrane potential via the Bcl2/BAX/caspase-3 pathway and modulate Cx43-mediated Ca2+ responses in EGCs to regulate colonic motility bidirectionally. Honokiol can ameliorate the damage to EGCs induced by high H2S concentrations through the Bcl2/BAX/caspase-3 pathway and improve colon motility by increasing Cx43 expression and Ca2+ concentration.


Assuntos
Apoptose , Compostos de Bifenilo , Sinalização do Cálcio , Proliferação de Células , Conexina 43 , Sulfeto de Hidrogênio , Lignanas , Neuroglia , Ratos Sprague-Dawley , Sulfeto de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Lignanas/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Ratos , Conexina 43/metabolismo , Conexina 43/genética , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Cálcio/metabolismo , Sistema Nervoso Entérico/efeitos dos fármacos , Sistema Nervoso Entérico/metabolismo , Células Cultivadas , Compostos Alílicos , Fenóis
8.
Mol Med ; 30(1): 113, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095693

RESUMO

BACKGROUND: To explore whether nobiletin has a protective effect on high-fat diet (HFD)-induced enteric nerve injury and its underlying mechanism. METHODS: An obesity model was induced by a HFD. Nobiletin (100 mg/kg and 200 mg/kg) and vehicle were administered by gastric gavage for 4 weeks. Lee's index, body weight, OGTT and intestinal propulsion assays were performed before sacrifice. After sampling, lipids were detected using Bodipy 493/503; lipid peroxidation was detected using MDA and SOD kits and the expression of PGP 9.5, Trem2, GFAP, ß-tubulin 3, Bax, Bcl2, Nestin, P75 NTR, SOX10 and EDU was detected using immunofluorescence. The GDNF, p-AKT, AKT, p-FOXO3a, FOXO3a and P21 proteins were detected using western blotting. The relative mRNA expression levels of NOS2 were detected via qPCR. Primary enteric neural stem cells (ENSCs) were cultured. After ENSCs were treated with palmitic acid (PA) and nobiletin, CCK-8 and caspase-3/7 activity assays were performed to evaluate proliferation and apoptosis. RESULTS: HFD consumption caused colon lipid accumulation and peroxidation, induced enteric nerve damage and caused intestinal motor dysfunction. However, nobiletin reduced lipid accumulation and peroxidation in the colon; promoted Trem2, ß-tubulin 3, Nestin, P75NTR, SOX10 and Bcl2 expression; inhibited Bax and GFAP expression; reduced NOS2 mRNA transcription; and regulated the GDNF/AKT/FOXO3a/P21 pathway. Nobiletin also promoted PA-induced impairment of ENSCs. CONCLUSIONS: Nobiletin restored HFD-induced enteric nerve injury, which may be associated with inhibiting enteric nerve apoptosis, promoting enteric nerve survival and regulating the GDNF/AKT/FOXO3a/P21 pathway.


Assuntos
Dieta Hiperlipídica , Sistema Nervoso Entérico , Flavonas , Proteína Forkhead Box O3 , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Proteína Forkhead Box O3/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Dieta Hiperlipídica/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Masculino , Flavonas/farmacologia , Flavonas/uso terapêutico , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/efeitos dos fármacos , Camundongos , Modelos Animais de Doenças , Ratos , Obesidade/metabolismo , Obesidade/tratamento farmacológico , Apoptose/efeitos dos fármacos
9.
Neurotox Res ; 42(4): 37, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39102123

RESUMO

Amyloid-peptide (Aß) monomeric forms (ABM) occurring in presymptomatic Alzheimer's disease (AD) brain are thought to be devoid of neurotoxicity while the transition/aggregation of ABM into oligomers is determinant for Aß-induced toxicity since Aß is predominantly monomeric up to 3 µM and aggregates over this concentration. However, recent imaging and/or histopathological investigations revealed alterations of myelin in prodromal AD brain in absence of aggregated Aß oligomers, suggesting that ABM may induce toxicity in myelin-producing cells in early AD-stages. To check this hypothesis, here we studied ABM effects on the viability of the Human oligodendrocyte cell line (HOG), a reliable oligodendrocyte model producing myelin proteins. Furthermore, to mimic closely interactions between oligodendrocytes and other glial cells regulating myelination, we investigated also ABM effects on mouse brain primary mixed-glial cell cultures. Various methods were combined to show that ABM concentrations (600 nM-1 µM), extremely lower than 3 µM, significantly decreased HOG cell and mouse brain primary mixed-glial cell survival. Interestingly, flow-cytometry studies using specific cell-type markers demonstrated that oligodendrocytes represent the most vulnerable glial cell population affected by ABM toxicity. Our work also shows that the neurosteroid 3α-O-allyl-allopregnanolone BR351 (250 and 500 nM) efficiently prevented ABM-induced HOG and brain primary glial cell toxicity. Bicuculline (50-100 nM), the GABA-A-receptor antagonist, was unable to block/reduce BR351 effect against ABM-induced HOG and primary glial cell toxicity, suggesting that BR351-evoked neuroprotection of these cells may not depend on GABA-A-receptor allosterically modulated by neurosteroids. Altogether, our results suggest that further exploration of BR351 therapeutic potential may offer interesting perspectives to develop effective neuroprotective strategies.


Assuntos
Peptídeos beta-Amiloides , Fármacos Neuroprotetores , Oligodendroglia , Pregnanolona , Animais , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Humanos , Peptídeos beta-Amiloides/toxicidade , Fármacos Neuroprotetores/farmacologia , Pregnanolona/farmacologia , Camundongos , Linhagem Celular , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/toxicidade , Células Cultivadas , Relação Dose-Resposta a Droga
10.
Int Immunopharmacol ; 139: 112654, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-38996777

RESUMO

Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.


Assuntos
Enzima de Conversão de Angiotensina 2 , Ligante de CD40 , Diminazena , Modelos Animais de Doenças , Hipertensão , Peptidil Dipeptidase A , Transdução de Sinais , Animais , Diminazena/análogos & derivados , Diminazena/farmacologia , Diminazena/uso terapêutico , Enzima de Conversão de Angiotensina 2/metabolismo , Masculino , Transdução de Sinais/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Ligante de CD40/metabolismo , Peptidil Dipeptidase A/metabolismo , Ratos , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Proto-Oncogene Mas , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Fragmentos de Peptídeos , Angiotensina I , Células Cultivadas , Microglia/efeitos dos fármacos , Microglia/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ratos Wistar , Sistema Renina-Angiotensina/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Antígenos CD40/metabolismo , Humanos , Ativação Plaquetária/efeitos dos fármacos , Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico
11.
Adv Sci (Weinh) ; 11(35): e2403461, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38992955

RESUMO

The precise mechanism underlying the therapeutic effects of dihydroartemisinin (DHA) in alleviating colitis remains incompletely understood. A strong correlation existed between the elevation of glial fibrillary acidic protein (GFAP)+/S100 calcium binding protein B (S100ß)+ enteric glial cells (EGCs) in inflamed colonic tissues and the disruption of the intestinal epithelial barrier (IEB) and gut vascular barrier (GVB) observed in chronic colitis. DHA demonstrated efficacy in restoring the functionality of the dual gut barrier while concurrently attenuating intestinal inflammation. Mechanistically, DHA inhibited the transformation of GFAP+ EGCs into GFAP+/S100ß+ EGCs while promoting the differentiation of GFAP+/S100ß+ EGCs back into GFAP+ EGCs. Furthermore, DHA induced apoptosis in GFAP+/S100ß+ EGCs by inducing cell cycle arrest at the G0/G1 phase. The initial mechanism is further validated that DHA regulates EGC heterogeneity by improving dysbiosis in colitis. These findings underscore the multifaceted therapeutic potential of DHA in ameliorating colitis by improving dysbiosis, modulating EGC heterogeneity, and preserving gut barrier integrity, thus offering promising avenues for novel therapeutic strategies for inflammatory bowel diseases.


Assuntos
Artemisininas , Colite , Modelos Animais de Doenças , Neuroglia , Animais , Artemisininas/farmacologia , Artemisininas/uso terapêutico , Colite/tratamento farmacológico , Colite/metabolismo , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Camundongos Endogâmicos C57BL , Masculino
12.
Eur J Pharmacol ; 979: 176818, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39029779

RESUMO

Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most debilitating adverse effects caused by chemotherapy drugs such as paclitaxel, oxaliplatin and vincristine. It is untreatable and often leads to the discontinuation of cancer therapy and a decrease in the quality of life of cancer patients. It is well-established that neuroinflammation and the activation of immune and glial cells are among the major drivers of CIPN. However, these processes are still poorly understood, and while many chemotherapy drugs alone can drive the activation of these cells and consequent neuroinflammation, it remains elusive to what extent the gut microbiome influences these processes. In this review, we focus on the peripheral mechanisms driving CIPN, and we address the bidirectional pathways by which the gut microbiome communicates with the immune and nervous systems. Additionally, we critically evaluate literature addressing how chemotherapy-induced dysbiosis and the consequent imbalance in bacterial products may contribute to the activation of immune and glial cells, both of which drive neuroinflammation and possibly CIPN development, and how we could use this knowledge for the development of effective treatment strategies.


Assuntos
Antineoplásicos , Microbioma Gastrointestinal , Doenças Neuroinflamatórias , Doenças do Sistema Nervoso Periférico , Humanos , Microbioma Gastrointestinal/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/microbiologia , Animais , Antineoplásicos/efeitos adversos , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/induzido quimicamente , Disbiose/induzido quimicamente , Disbiose/microbiologia , Neuroglia/efeitos dos fármacos , Neuroglia/imunologia
13.
J Ethnopharmacol ; 334: 118571, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38996953

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Jiawei Bai-Hu-Decoction (JWBHD), a prescription formulated with seven traditional Chinese medicinal material has demonstrated clinical efficacy in mitigating brain injury among heat stroke (HS) patients. AIM OF THE STUDY: This study aimed to evaluate the therapeutic efficacy of JWBHD on rat model of HS and to explore its therapeutic mechanisms by integrating network pharmacology and pharmacodynamic methodologies, which major components were analyzed by using UPLC-MS/MS. MATERIALS AND METHODS: The network pharmacology analysis was firstly conducted to predict the potential active ingredients and therapeutic targets of JWBHD. The anti-HS effectiveness of JWBHD was then evaluated on rats experienced HS. Rat brain tissues were harvested for a comprehensive array of experiments, including Western blot, PCR, H&E staining, Nissl staining, ELISA, transmission electron microscope, flow cytometry and immunofluorescence to validate the protective effects of JWBHD against HS-induced brain damage. Furthermore, the inhibitory effects of JWBHD on TLR4/NF-κB signal and mitophagy of glial were further verified on HS-challenged F98 cell line. Finally, the chemical compositions of the water extract of JWBHD were analyzed by using UPLC-MS/MS. RESULTS: Network pharmacology has identified fifty core targets and numerous HS-related signaling pathways as potential therapeutic targets of JWBHD. Analysis of protein-protein interaction (PPI) and GO suggests that JWBHD may suppress HS-induced inflammatory signals. In experiments conducted on HS-rats, JWBHD significantly reduced the core temperature, restored blood pressure and alleviated neurological defect. Furthermore, JWBHD downregulated the counts of white blood cells and monocytes, decreased the levels of inflammatory cytokines such as IL-1ß, IL-6 and TNF-α in peripheral blood, and suppressed the expression of TLR4 and NF-κB in the cerebral cortex of HS-rats. Besides, JWBHD inhibited the apoptosis of cortical cells and mitigated the damage to the cerebral cortex in HS group. Conversely, overactive mitophagy was observed in the cerebral cortex of HS-rats. However, JWBHD restored the mitochondrial membrane potential and downregulated expressions of mitophagic proteins including Pink1, Parkin, LC3B and Tom20. JWBHD reduced the co-localization of Pink1 and GFAP, a specific marker of astrocytes in the cerebral cortex of HS-rats. In addition, the inhibitory effect of JWBHD on TLR4/NF-κB signaling and overactive mitophagy were further confirmed in F98 cells. Finally, UPLC-MS/MS analysis showed that the main components of JWBHD include isoliquiritigenin, liquiritin, dipotassium glycyrrhizinate, ginsenoside Rb1, ginsenoside Re, etc. CONCLUSIONS: JWBHD protected rats from HS and prevented HS-induced damage in the cerebral cortex by suppressing TLR4/NF-κB signaling and mitophagy of glial.


Assuntos
Medicamentos de Ervas Chinesas , Golpe de Calor , Mitofagia , NF-kappa B , Neuroglia , Ratos Sprague-Dawley , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Receptor 4 Toll-Like/metabolismo , Mitofagia/efeitos dos fármacos , NF-kappa B/metabolismo , Masculino , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Transdução de Sinais/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Ratos , Golpe de Calor/tratamento farmacológico , Golpe de Calor/complicações , Fármacos Neuroprotetores/farmacologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/prevenção & controle , Farmacologia em Rede , Modelos Animais de Doenças
14.
J Med Chem ; 67(15): 13217-13230, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39031092

RESUMO

Pro-inflammatory polarization of microglia and astrocytes results in neuroinflammation and blood-brain barrier (BBB) disruption after a primary traumatic brain injury (TBI). Herein, we demonstrate that the dual-ligand functionalized lipid nanoparticles (AM31 LNPs) were actively and specifically internalized by microglia and astrocytes via mannose receptor (MR)- and adenosine receptor (AR)-mediated endocytosis, respectively, in a mouse model of TBI. Systemic administration of AM31 LNPs carrying siRNA against p65 resulted in internalization by the glial cells in the peri-infarct region and a robust knockdown of p65 at both mRNA and protein levels in these cells, leading to significant down-regulation of key pro-inflammatory cytokines and up-regulation of key anti-inflammatory cytokines. AM31 LNP-mediated silencing of p65 ameliorated TBI-induced BBB disruption. Our data proved that AM 31 LNP is a promising vehicle for RNA therapeutics for targeting microglia and astrocytes in neural disorder.


Assuntos
Barreira Hematoencefálica , Lipídeos , Nanopartículas , Animais , Barreira Hematoencefálica/metabolismo , Nanopartículas/química , Ligantes , Camundongos , Lipídeos/química , RNA Interferente Pequeno/metabolismo , Camundongos Endogâmicos C57BL , Interferência de RNA , Microglia/metabolismo , Microglia/efeitos dos fármacos , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/terapia , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Masculino , Neuroglia/metabolismo , Neuroglia/efeitos dos fármacos
15.
Cells ; 13(13)2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38994956

RESUMO

Clostridioides difficile (C. difficile) is responsible for a spectrum of nosocomial/antibiotic-associated gastrointestinal diseases that are increasing in global incidence and mortality rates. The C. difficile pathogenesis is due to toxin A and B (TcdA/TcdB), both causing cytopathic and cytotoxic effects and inflammation. Recently, we demonstrated that TcdB induces cytopathic and cytotoxic (apoptosis and necrosis) effects in enteric glial cells (EGCs) in a dose/time-dependent manner and described the underlying signaling. Despite the role played by lipids in host processes activated by pathogens, to counter infection and/or induce cell death, to date no studies have investigated lipid changes induced by TcdB/TcdA. Here, we evaluated the modification of lipid composition in our in vitro model of TcdB infection. Apoptosis, cell cycle, cell viability, and lipidomic profiles were evaluated in EGCs treated for 24 h with two concentrations of TcdB (0.1 ng/mL; 10 ng/mL). In EGCs treated with the highest concentration of TcdB, not only an increased content of total lipids was observed, but also lipidome changes, allowing the separation of TcdB-treated cells and controls into different clusters. The statistical analyses also allowed us to ascertain which lipid classes and lipid molecular species determine the clusterization. Changes in lipid species containing inositol as polar head and plasmalogen phosphatidylethanolamine emerged as key indicators of altered lipid metabolism in TcdB-treated EGCs. These results not only provide a picture of the phospholipid profile changes but also give information regarding the lipid metabolism pathways altered by TcdB, and this might represent an important step for developing strategies against C. difficile infection.


Assuntos
Proteínas de Bactérias , Toxinas Bacterianas , Neuroglia , Fosfolipídeos , Neuroglia/metabolismo , Neuroglia/efeitos dos fármacos , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Toxinas Bacterianas/farmacologia , Fosfolipídeos/metabolismo , Proteínas de Bactérias/metabolismo , Clostridioides difficile/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Lipidômica , Humanos
16.
Nutrients ; 16(14)2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39064769

RESUMO

Vitamin D deficiency (VDD) is widespread around the world and has been extensively documented to affect various health conditions, including the cognitive functioning of the brain. Serum 25-hydroxylated forms of vitamin D are traditionally used to determine vitamin D status. However, there is now evidence that cholecalciferol activation can occur and be controlled by locally expressed enzymes in the brain. This study aimed to investigate the effects of cholecalciferol supplementation on cognitive function in rats who underwent transient VDD in adulthood. Thirty-six adult Wistar rats were administered paricalcitol (seven doses of 32 ng injected every other day) along with a "vitamin D-free" diet to induce VDD, which was confirmed using a LC-MS/MS serum analysis of the cholecalciferol and 25-hydroxyvitamin D3 levels. Treatment was performed by including 1000 IU/kg and 10,000 IU/kg cholecalciferol in the diet. Cognitive performance was evaluated using the novel object recognition (NOR), Morris water maze (MWM), and radial arm maze (RAM) tests. An immunohistochemical analysis of the brain regions involved in learning and memory was performed by quantifying the neurons, astrocytes, and microglia labelled with anti-neuronal nuclei (NeuN), glial fibrillary acidic protein (GFAP), and ionized calcium-binding adaptor molecule 1 (Iba-1) antibodies, respectively. The vitamin D deficient group showed the lowest performance in both the MWM and RAM tests. In contrast, the cholecalciferol-treated groups exhibited a faster learning curve. However, no difference was detected between the groups in the NOR test. On the other hand, differences in the cellular organization of the hippocampus and amygdala were observed between the groups. Cholecalciferol supplementation decreased the density of the Iba-1- and GFAP-labeled cells in the hilus and cornu Ammonis 3 (CA3) regions of the hippocampus and in the amygdala. These results support vitamin D's substantial role in learning and memory. They also highlight that subtle changes of cognitive function induced by transient VDD could be reversed by cholecalciferol supplementation. Further studies are needed to better understand VDD and cholecalciferol's effects on the brain structure and function.


Assuntos
Colecalciferol , Suplementos Nutricionais , Hipocampo , Neuroglia , Ratos Wistar , Deficiência de Vitamina D , Animais , Colecalciferol/farmacologia , Deficiência de Vitamina D/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Masculino , Ratos , Cognição/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Ergocalciferóis/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Modelos Animais de Doenças , Vitamina D/farmacologia , Vitamina D/sangue
17.
Neurotoxicol Teratol ; 104: 107370, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38964664

RESUMO

The brain is especially vulnerable to environmental influences during the perinatal period. While the effects of environmental factors are usually studied in isolation, it is more typical to be exposed to multiple influences during early development, necessitating study of synergistic actions on the developing brain. Both maternal infection and endocrine disrupting phthalates can decrease cell number in the medial prefrontal cortex (mPFC), a region critical for executive functioning. In the present study, groups of pregnant Long Evans rats were treated with either (1) 100 µg/kg (i.p.) lipopolysaccharide (LPS) on embryonic days 15 and 16 combined with a low-dose (1 mg/kg) phthalate mixture throughout gestation and the neonatal period, (2) LPS alone, (3) phthalates alone, or (4) neither phthalates nor LPS (control). Neurons and glial cells were stereologically quantified in the mPFC. The adult offspring previously exposed to LPS or phthalates alone had reduced mPFC neuron number in exposed males, but not females, while the combination treatment did not produce significant effects. In males, LPS alone also reduced the number of glia in the mPFC. Additionally, the combination of LPS and phthalates resulted in fewer pregnancies to term and decreased litter size. These results provide insight into how common environmental factors can interact to alter the developmental trajectory of the mPFC.


Assuntos
Lipopolissacarídeos , Neurônios , Ácidos Ftálicos , Córtex Pré-Frontal , Efeitos Tardios da Exposição Pré-Natal , Ratos Long-Evans , Animais , Córtex Pré-Frontal/efeitos dos fármacos , Feminino , Gravidez , Lipopolissacarídeos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/patologia , Masculino , Ratos , Neurônios/efeitos dos fármacos , Ácidos Ftálicos/toxicidade , Contagem de Células , Neuroglia/efeitos dos fármacos , Exposição Materna/efeitos adversos
18.
Prog Orthod ; 25(1): 33, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39034361

RESUMO

BACKGROUND: Orthodontic pain affects the physical and mental health of patients. The spinal trigeminal subnucleus caudalis (SPVC) contributes to the transmission of pain information and serves as a relay station for integrating orofacial damage information. Recently, glial cells have been found to be crucial for both acute and maintenance phases of pain. It has also been demonstrated that rho kinase (ROCK) inhibitors can manage different pain models by inhibiting glial cell activation. Here, we hypothesized that orthodontic pain is related to glial cells in the SPVC, and Fasudil, a representative rho/rock kinase inhibitor, can relieve orthodontic pain by regulating the function of glial cells and the related inflammatory factors. In this study, we constructed a rat model of tooth movement pain and used immunofluorescence staining to evaluate the activation of microglia and astrocytes. Quantitative real-time PCR was used to detect the release of related cytokines and the expression of pain-related genes in the SPVC. Simultaneously, we investigated the effect of Fasudil on the aforementioned indicators. RESULTS: In the SPVC, the expression of c-Fos peaked on day 1 along with the expression of OX42 (related to microglial activation), CD16 (a pro-inflammatory factor), and CD206 (an anti-inflammatory factor) on day 3 after tooth movement, followed by a gradual decrease. GFAP-staining showed that the number of activated astrocytes was the highest on day 5 and that cell morphology became complex. After Fasudil treatment, the expression of these proteins showed a downward trend. The mRNA levels of pro-inflammatory factors (IL-1ß and TNF-α) peaked on day 3, and the mRNA expression of the anti-inflammatory factor TGF-ß was the lowest 3 days after tooth movement. Fasudil inhibited the mRNA expression of pain-related genes encoding CSF-1, t-PA, CTSS, and BDNF. CONCLUSION: This study shows that tooth movement can cause the activation of glial cells in SPVC, and ROCK inhibitor Fasudil can inhibit the activation of glial cells and reduce the expression of the related inflammatory factors. This study presents for the first time the potential application of Fasudil in othodontic pain.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina , Neuroglia , Técnicas de Movimentação Dentária , Animais , Técnicas de Movimentação Dentária/métodos , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/uso terapêutico , Ratos , Neuroglia/efeitos dos fármacos , Ratos Sprague-Dawley , Masculino , Microglia/efeitos dos fármacos , Núcleo Inferior Caudal do Nervo Trigêmeo/efeitos dos fármacos , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Modelos Animais de Doenças , Citocinas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Astrócitos/efeitos dos fármacos
19.
Biomed Pharmacother ; 178: 117157, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39042964

RESUMO

Although the potent anti-inflammatory effects of irisin have been documented in various inflammatory disorders, its efficacy against inflammatory pain remains unexplored. Herein, we examined the therapeutic effects of irisin in a mouse model of inflammatory pain induced by complete Freund's adjuvant (CFA). Mice were divided into three groups: normal control, CFA-injected (CFA), and CFA plus irisin-treated (CFA+Irisin). The irisin-treated group exhibited a gradual reduction in mechanical allodynia and thermal hyperalgesia when compared with the CFA group. Moreover, treatment with irisin significantly upregulated the expression of M2 macrophage markers (interleukin [IL]-4 and IL-10) and downregulated M1 macrophage markers (IL-1ß, IL-6, and tumor necrosis factor-α) in the local paw tissue, dorsal root ganglion, and spinal cord tissue. However, there was no significant difference in the total number of F4/80+ macrophages in the paw tissue and dorsal root ganglion, indicating phenotypic exchange. Treatment with irisin also downregulated the expression of the glial cell activation-related markers Iba-1 and GFAP in the spinal cord tissue. To elucidate the underlying mechanisms, we detected the expression of Toll-like receptor 4 (TLR4), MyD88, and interferon regulatory factor 5 (IRF5) in paw tissues, dorsal root ganglion, and spinal tissues, revealing that irisin could downregulate the expression of these proteins. Irisin alleviated inflammatory pain by modulating local tissue inflammation and peripheral and central neuroinflammation and reducing glial cell activation and M2 macrophage polarization by modulating the TLR4-MyD88-IRF5 signaling pathway. Accordingly, irisin is a promising candidate for treating inflammatory pain in various diseases.


Assuntos
Fibronectinas , Adjuvante de Freund , Inflamação , Macrófagos , Neuroglia , Medula Espinal , Animais , Fibronectinas/metabolismo , Masculino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Camundongos Endogâmicos C57BL , Dor/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Receptor 4 Toll-Like/metabolismo , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Modelos Animais de Doenças , Transdução de Sinais/efeitos dos fármacos
20.
Neuroscience ; 553: 145-159, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-38992567

RESUMO

Glutamate excitotoxicity is involved in retinal ganglion cell (RGC) death in various retinal degenerative diseases, including ischemia-reperfusion injury and glaucoma. Excitotoxic RGC death is caused by both direct damage to RGCs and indirect damage through neuroinflammation of retinal glial cells. Omidenepag (OMD), a novel E prostanoid receptor 2 (EP2) agonist, is a recently approved intraocular pressure-lowering drug. The second messenger of EP2 is cyclic adenosine monophosphate (cAMP), which activates protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac). In this study, we investigated the neuroprotective effects of OMD on excitotoxic RGC death by focusing on differences in cAMP downstream signaling from the perspective of glia-neuron interactions. We established a glutamate excitotoxicity model in vitro and NMDA intravitreal injection model in vivo. In vitro, rat primary RGCs were used in an RGC survival rate assay. MG5 cells (mouse microglial cell line) and A1 cells (astrocyte cell line) were used for immunocytochemistry and Western blotting to evaluate the expressions of COX-1/2, PKA, Epac1/2, pCREB, cleaved caspase-3, inflammatory cytokines, and neurotrophic factors. Mouse retinal specimens underwent hematoxylin and eosin staining, flat-mounted retina examination, and immunohistochemistry. OMD significantly suppressed excitotoxic RGC death, cleaved caspase-3 expression, and activated glia both in vitro and in vivo. Moreover, it inhibited Epac1 and inflammatory cytokine expression and promoted COX-2, pCREB, and neurotrophic factor expression. OMD may have neuroprotective effects through inhibition of the Epac pathway and promotion of the COX-2-EP2-cAMP-PKA pathway by modulating glia-neuron interaction.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico , AMP Cíclico , Ciclo-Oxigenase 2 , Neuroglia , Fármacos Neuroprotetores , Células Ganglionares da Retina , Animais , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Fármacos Neuroprotetores/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/metabolismo , AMP Cíclico/metabolismo , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/agonistas , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Ratos Sprague-Dawley , Ratos , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Camundongos Endogâmicos C57BL , Masculino , N-Metilaspartato/farmacologia , N-Metilaspartato/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA