Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 324(3): C674-C678, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36717106

RESUMO

Adropin is a highly conserved secreted peptide encoded by the Energy Homeostasis Associated gene (Enho). It is expressed in many tissues throughout the body, including the liver and brain, and plays a crucial role in maintaining lipid homeostasis and regulating insulin sensitivity. Adropin also participates in several other pathophysiological processes of multiple central nervous system (CNS) diseases. There is strong evidence of the protective effects of adropin in stroke, heart disease, aging, and other diseases. The peptide has been shown to reduce the risk of disease, attenuate histological alterations, and reduce cognitive decline associated with neurological disorders. Recent findings support its critical role in regulating endothelial cells and maintaining blood-brain barrier integrity through an endothelial nitric oxide synthase (eNOS)-dependent mechanism. Here we discuss current evidence of the protective effects of adropin in CNS diseases specifically involving the cerebrovasculature and highlight potential mechanisms through which the peptide exhibits these effects.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular , Doenças do Sistema Nervoso , Humanos , Envelhecimento , Células Endoteliais , Peptídeos e Proteínas de Sinalização Intercelular/genética , Doenças do Sistema Nervoso/genética , Peptídeos/genética
2.
Stroke ; 54(1): 234-244, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36305313

RESUMO

BACKGROUND: Adropin is a peptide encoded by the energy homeostasis-associated gene (Enho) that is highly expressed in the brain. Aging and stroke are associated with reduced adropin levels in the brain and plasma. We showed that treatment with synthetic adropin provides long-lasting neuroprotection in permanent ischemic stroke. However, it is unknown whether the protective effects of adropin are observed in aged animals following cerebral ischemia/reperfusion. We hypothesized that adropin provides neuroprotection in aged mice subjected to transient middle cerebral artery occlusion. METHODS: Aged (18-24 months old) male mice were subjected to 30 minutes of middle cerebral artery occlusion followed by 48 hours or 14 days of reperfusion. Sensorimotor (weight grip test and open field) and cognitive tests (Y-maze and novel object recognition) were performed at defined time points. Infarct volume was quantified by 2,3,5-triphenyltetrazolium chloride staining at 48 hours or Cresyl violet staining at 14 days post-middle cerebral artery occlusion. Blood-brain barrier damage, tight junction proteins, and MMP-9 (matrix metalloproteinase-9) were assessed 48 hours after middle cerebral artery occlusion by ELISA and Western blots. RESULTS: Genetic deletion of Enho significantly increased infarct volume and worsened neurological function, whereas overexpression of adropin dramatically reduced stroke volume compared to wild-type controls. Postischemic treatment with synthetic adropin peptide given at the onset of reperfusion markedly reduced infarct volume, brain edema, and significantly improved locomotor function and muscular strength at 48 hours. Delayed adropin treatment (4 hours after the stroke onset) reduced body weight loss, infarct volume, and muscular strength dysfunction, and improved long-term cognitive function. Postischemic adropin treatment significantly reduced blood-brain barrier damage. This effect was associated with reduced MMP-9 and preservation of tight junction proteins by adropin treatment. CONCLUSIONS: These data unveil a promising neuroprotective role of adropin in the aged brain after transient ischemic stroke via reducing neurovascular damage. These findings suggest that poststroke adropin therapy is a potential strategy to minimize brain injury and improve functional recovery in ischemic stroke patients.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Camundongos , Masculino , Animais , Barreira Hematoencefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , AVC Isquêmico/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Peptídeos/farmacologia , Peptídeos/genética , Peptídeos/metabolismo , Proteínas de Junções Íntimas/metabolismo
3.
J Neuroinflammation ; 20(1): 221, 2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37777791

RESUMO

BACKGROUND: Receptor-interacting protein kinase 2 (RIPK2) is a serine/threonine kinase whose activity propagates inflammatory signaling through its association with pattern recognition receptors (PRRs) and subsequent TAK1, NF-κB, and MAPK pathway activation. After stroke, dead and dying cells release a host of damage-associated molecular patterns (DAMPs) that activate PRRs and initiate a robust inflammatory response. We hypothesize that RIPK2 plays a damaging role in the progression of stroke injury by enhancing the neuroinflammatory response to stroke and that global genetic deletion or microglia-specific conditional deletion of Ripk2 will be protective following ischemic stroke. METHODS: Adult (3-6 months) male mice were subjected to 45 min of transient middle cerebral artery occlusion (tMCAO) followed by 24 h, 48 h, or 28 days of reperfusion. Aged male and female mice (18-24 months) were subjected to permanent ischemic stroke and sacrificed 48 h later. Infarct volumes were calculated using TTC staining (24-48 h) or Cresyl violet staining (28d). Sensorimotor tests (weight grip, vertical grid, and open field) were performed at indicated timepoints. Blood-brain barrier (BBB) damage, tight junction proteins, matrix metalloproteinase-9 (MMP-9), and neuroinflammatory markers were assessed via immunoblotting, ELISA, immunohistochemistry, and RT-qPCR. Differential gene expression profiles were generated through bulk RNA sequencing and nanoString®. RESULTS: Global genetic deletion of Ripk2 resulted in decreased infarct sizes and reduced neuroinflammatory markers 24 h after stroke compared to wild-type controls. Ripk2 global deletion also improved both acute and long-term behavioral outcomes with powerful effects on reducing infarct volume and mortality at 28d post-stroke. Conditional deletion of microglial Ripk2 (mKO) partially recapitulated our results in global Ripk2 deficient mice, showing reductive effects on infarct volume and improved behavioral outcomes within 48 h of injury. Finally, bulk transcriptomic profiling and nanoString data demonstrated that Ripk2 deficiency in microglia decreases genes associated with MAPK and NF-κB signaling, dampening the neuroinflammatory response after stroke injury by reducing immune cell activation and peripheral immune cell invasion. CONCLUSIONS: These results reveal a hitherto unknown role for RIPK2 in the pathogenesis of ischemic stroke injury, with microglia playing a distinct role. This study identifies RIPK2 as a potent propagator of neuroinflammatory signaling, highlighting its potential as a therapeutic target for post-stroke intervention.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Feminino , Camundongos , Masculino , Animais , Microglia/metabolismo , Doenças Neuroinflamatórias , NF-kappa B/metabolismo , Acidente Vascular Cerebral/patologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/metabolismo , Inflamação/metabolismo , Infarto , AVC Isquêmico/metabolismo , Proteínas Quinases/metabolismo , Isquemia Encefálica/metabolismo
4.
Stroke ; 53(5): 1473-1486, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35387495

RESUMO

Maintaining blood-brain barrier (BBB) integrity is crucial for the homeostasis of the central nervous system. Structurally comprising the BBB, brain endothelial cells interact with pericytes, astrocytes, neurons, microglia, and perivascular macrophages in the neurovascular unit. Brain ischemia unleashes a profound neuroinflammatory response to remove the damaged tissue and prepare the brain for repair. However, the intense neuroinflammation occurring during the acute phase of stroke is associated with BBB breakdown, neuronal injury, and worse neurological outcomes. Here, we critically discuss the role of neuroinflammation in ischemic stroke pathology, focusing on the BBB and the interactions between central nervous system and peripheral immune responses. We highlight inflammation-driven injury mechanisms in stroke, including oxidative stress, increased MMP (matrix metalloproteinase) production, microglial activation, and infiltration of peripheral immune cells into the ischemic tissue. We provide an updated overview of imaging techniques for in vivo detection of BBB permeability, leukocyte infiltration, microglial activation, and upregulation of cell adhesion molecules following ischemic brain injury. We discuss the possibility of clinical implementation of imaging modalities to assess stroke-associated neuroinflammation with the potential to provide image-guided diagnosis and treatment. We summarize the results from several clinical studies evaluating the efficacy of anti-inflammatory interventions in stroke. Although convincing preclinical evidence suggests that neuroinflammation is a promising target for ischemic stroke, thus far, translating these results into the clinical setting has proved difficult. Due to the dual role of inflammation in the progression of ischemic damage, more research is needed to mechanistically understand when the neuroinflammatory response begins the transition from injury to repair. This could have important implications for ischemic stroke treatment by informing time- and context-specific therapeutic interventions.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Humanos , Inflamação/patologia , Doenças Neuroinflamatórias , Acidente Vascular Cerebral/metabolismo
5.
Stroke ; 53(10): 3238-3242, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35904018

RESUMO

Poststroke infections are common complications of stroke and are highly associated with poor outcomes for patients. Stroke induces profound immunodepression coupled with alterations to autonomic signaling, which together render the body more susceptible to infection from without (nosocomial/community-acquired infection) and from within (commensal bacterial infection). Critical to the hypothesis of commensal infection is the phenomenon of poststroke gut permeability and gut dysbiosis. Few studies have provided adequate explanations for the mechanisms underlying the molecular alterations that produce a more permeable gut and perturbed gut microbiota after stroke. A dysregulation in the production of matrix MMP-7 (metalloproteinase-7) may play a critical role in the progression of gut permeability after stroke. By cleaving junctional and extracellular matrix proteins, MMP-7 is capable of compromising gut barrier integrity. Because of MMP-7's unique abundance in the small intestine and its capacity to be induced in states of bacterial invasion and inflammation, along with its unique degradative capability, MMP-7 may be crucially important to the progression of gut permeability after ischemic stroke.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Proteínas da Matriz Extracelular , Humanos , Metaloproteinase 7 da Matriz , Permeabilidade , Acidente Vascular Cerebral/complicações
6.
J Neuroinflammation ; 19(1): 168, 2022 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-35761277

RESUMO

Bromodomain-containing protein 4 (BRD4), a member of the bromodomain and extra-terminal domain (BET) protein family, plays a crucial role in regulating inflammation and oxidative stress that are tightly related to stroke development and progression. Consequently, BRD4 blockade has attracted increasing interest for associated neurological diseases, including stroke. dBET1 is a novel and effective BRD4 degrader through the proteolysis-targeting chimera (PROTAC) strategy. We hypothesized that dBET1 protects against brain damage and neurological deficits in a transient focal ischemic stroke mouse model by reducing inflammation and oxidative stress and preserving the blood-brain barrier (BBB) integrity. Post-ischemic dBET1 treatment starting 4 h after stroke onset significantly ameliorated severe neurological deficits and reduced infarct volume 48 h after stroke. dBET1 markedly reduced inflammation and oxidative stress after stroke, indicated by multiple pro-inflammatory cytokines and chemokines including IL-1ß, IL-6, TNF-α, CCL2, CXCL1 and CXCL10, and oxidative damage markers 4-hydroxynonenal (4-HNE) and gp91phox and antioxidative proteins SOD2 and GPx1. Meanwhile, stroke-induced BBB disruption, increased MMP-9 levels, neutrophil infiltration, and increased ICAM-1 were significantly attenuated by dBET1 treatment. Post-ischemic dBET1 administration also attenuated ischemia-induced reactive gliosis in microglia and astrocytes. Overall, these findings demonstrate that BRD4 degradation by dBET1 improves acute stroke outcomes, which is associated with reduced neuroinflammation and oxidative stress and preservation of BBB integrity. This study identifies a novel role of BET proteins in the mechanisms resulting in ischemic brain damage, which can be leveraged to develop novel therapies.


Assuntos
Barreira Hematoencefálica , Isquemia Encefálica , Proteínas Nucleares , Acidente Vascular Cerebral , Fatores de Transcrição , Animais , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Inflamação/metabolismo , Camundongos , Doenças Neuroinflamatórias , Proteínas Nucleares/metabolismo , Estresse Oxidativo , Proteólise , Acidente Vascular Cerebral/metabolismo , Fatores de Transcrição/metabolismo
7.
Diabetologia ; 64(10): 2279-2291, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34274990

RESUMO

AIMS/HYPOTHESIS: Normal cellular prion protein (PrPC) is a conserved mammalian glycoprotein found on the outer plasma membrane leaflet through a glycophosphatidylinositol anchor. Although PrPC is expressed by a wide range of tissues throughout the body, the complete repertoire of its functions has not been fully determined. The misfolded pathogenic isoform PrPSc (the scrapie form of PrP) is a causative agent of neurodegenerative prion diseases. The aim of this study is to evaluate PrPC localisation, expression and trafficking in pancreases from organ donors with and without type 1 diabetes and to infer PrPC function through studies on interacting protein partners. METHODS: In order to evaluate localisation and trafficking of PrPC in the human pancreas, 12 non-diabetic, 12 type 1 diabetic and 12 autoantibody-positive organ donor tissue samples were analysed using immunofluorescence analysis. Furthermore, total RNA was isolated from 29 non-diabetic, 29 type 1 diabetic and 24 autoantibody-positive donors to estimate PrPC expression in the human pancreas. Additionally, we performed PrPC-specific immunoblot analysis on total pancreatic protein from non-diabetic and type 1 diabetic organ donors to test whether changes in PrPC mRNA levels leads to a concomitant increase in PrPC protein levels in human pancreases. RESULTS: In non-diabetic and type 1 diabetic pancreases (the latter displaying both insulin-positive [INS(+)] and -negative [INS(-)] islets), we found PrPC in islets co-registering with beta cells in all INS(+) islets and, strikingly, unexpected activation of PrPC in alpha cells within diabetic INS(-) islets. We found PrPC localised to the plasma membrane and endoplasmic reticulum (ER) but not the Golgi, defining two cellular pools and an unconventional protein trafficking mechanism bypassing the Golgi. We demonstrate PrPC co-registration with established protein partners, neural cell adhesion molecule 1 (NCAM1) and stress-inducible phosphoprotein 1 (STI1; encoded by STIP1) on the plasma membrane and ER, respectively, linking PrPC function with cyto-protection, signalling, differentiation and morphogenesis. We demonstrate that both PRNP (encoding PrPC) and STIP1 gene expression are dramatically altered in type 1 diabetic and autoantibody-positive pancreases. CONCLUSIONS/INTERPRETATION: As the first study to address PrPC expression in non-diabetic and type 1 diabetic human pancreas, we provide new insights for PrPC in the pathogenesis of type 1 diabetes. We evaluated the cell-type specific expression of PrPC in the human pancreas and discovered possible connections with potential interacting proteins that we speculate might address mechanisms relevant to the role of PrPC in the human pancreas.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Pâncreas/metabolismo , Proteínas PrPC/metabolismo , Adolescente , Adulto , Autoanticorpos/sangue , Antígeno CD56/metabolismo , Membrana Celular/metabolismo , Criança , Retículo Endoplasmático/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Proteínas de Choque Térmico/metabolismo , Humanos , Imuno-Histoquímica , Anticorpos Anti-Insulina/imunologia , Masculino , Proteínas PrPC/genética , Proteínas Priônicas/genética , Proteínas Priônicas/metabolismo , Transporte Proteico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Doadores de Tecidos , Adulto Jovem
8.
Brain Behav Immun ; 93: 141-155, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33422638

RESUMO

The neuron-specific tyrosine phosphatase STEP is emerging as a key neuroprotectant against acute ischemic stroke. However, it remains unclear how STEP impacts the outcome of stroke. We find that the exacerbation of ischemic brain injury in STEP deficient mice involves an early onset and sustained activation of neuronal p38 mitogen activated protein kinase, a substrate of STEP. This leads to rapid increase in the expression of neuronal cyclooxygenase-2 and synthesis of prostaglandin E2, causing change in microglial morphology to an amoeboid activated state, activation of matrix metalloproteinase-9, cleavage of tight junction proteins and extravasation of IgG into the ischemic brain. Restoration of STEP signaling with intravenous administration of a STEP-derived peptide mimetic reduces the post-ischemic inflammatory response and attenuates brain injury. The findings identify a unique role of STEP in regulating post-ischemic neuroinflammation and further emphasizes the therapeutic potential of the STEP-mimetic in neurological disorders where inflammation contributes to brain damage.


Assuntos
Isquemia Encefálica , Proteínas Tirosina Fosfatases não Receptoras , Acidente Vascular Cerebral , Animais , Camundongos , Neurônios
9.
Am J Physiol Cell Physiol ; 316(2): C135-C153, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30379577

RESUMO

As part of the neurovascular unit, the blood-brain barrier (BBB) is a unique, dynamic regulatory boundary that limits and regulates the exchange of molecules, ions, and cells between the blood and the central nervous system. Disruption of the BBB plays an important role in the development of neurological dysfunction in ischemic stroke. Blood-borne substances and cells have restricted access to the brain due to the presence of tight junctions between the endothelial cells of the BBB. Following stroke, there is loss of BBB tight junction integrity, leading to increased paracellular permeability, which results in vasogenic edema, hemorrhagic transformation, and increased mortality. Thus, understanding principal mediators and molecular mechanisms involved in BBB disruption is critical for the development of novel therapeutics to treat ischemic stroke. This review discusses the current knowledge of how neuroinflammation contributes to BBB damage in ischemic stroke. Specifically, we provide an updated overview of the role of cytokines, chemokines, oxidative and nitrosative stress, adhesion molecules, matrix metalloproteinases, and vascular endothelial growth factor as well as the role of different cell types in the regulation of BBB permeability in ischemic stroke.


Assuntos
Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Mediadores da Inflamação/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Transporte Biológico/fisiologia , Barreira Hematoencefálica/imunologia , Isquemia Encefálica/imunologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Mediadores da Inflamação/imunologia , Estresse Oxidativo/fisiologia , Permeabilidade , Acidente Vascular Cerebral/imunologia , Junções Íntimas/imunologia , Junções Íntimas/metabolismo
10.
Int J Mol Sci ; 20(9)2019 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-31075861

RESUMO

A large body of experimental evidence suggests that neuroinflammation is a key pathological event triggering and perpetuating the neurodegenerative process associated with many neurological diseases. Therefore, different stimuli, such as lipopolysaccharide (LPS), are used to model neuroinflammation associated with neurodegeneration. By acting at its receptors, LPS activates various intracellular molecules, which alter the expression of a plethora of inflammatory mediators. These factors, in turn, initiate or contribute to the development of neurodegenerative processes. Therefore, LPS is an important tool for the study of neuroinflammation associated with neurodegenerative diseases. However, the serotype, route of administration, and number of injections of this toxin induce varied pathological responses. Thus, here, we review the use of LPS in various models of neurodegeneration as well as discuss the neuroinflammatory mechanisms induced by this toxin that could underpin the pathological events linked to the neurodegenerative process.


Assuntos
Inflamação/patologia , Degeneração Neural/patologia , Sistema Nervoso/patologia , Animais , Modelos Animais de Doenças , Humanos , Lipopolissacarídeos , Doenças Neurodegenerativas/patologia
11.
Biochem Biophys Res Commun ; 497(1): 410-415, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29448097

RESUMO

Bromodomain and extraterminal (BET) proteins are essential to pro-inflammatory gene transcription. The BET family proteins, BRD2, BRD3, BRD4, and testis-specific BRDT, couple chromatin remodeling to gene transcription, acting as histone acetyltransferases, scaffolds for transcription complexes, and markers of histone acetylation. To initiate an inflammatory response, cells undergo de novo gene transcription requiring histone-modifying proteins to make DNA wrapped around histones more or less readily available to transcription complexes. Because BET proteins are the gatekeepers of nuclear factor-κB (NF-κB)-dependent gene transcription, we hypothesized that degradation of BET proteins, particularly BRD2 and BRD4, with the proteolysis-targeting chimera (PROTAC) dBET1 would dampen the pro-inflammatory response in microglia subjected to lipopolysaccharide (LPS) challenge. Degradation of BRD2 and BRD4 was associated with significantly reduced expression of several pro-inflammatory genes: inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin (IL)-1ß, tumor necrosis factor-a (TNF-α), IL-6, chemokine (C-C motif) ligand 2 (CCL2), and matrix metalloproteinase-9 (MMP-9). This is the first study showing that dBET1-mediated targeted degradation of BET proteins robustly dampens pro-inflammatory responses in LPS-stimulated microglia. These data suggest that BET degradation with dBET1 will likely reduce expression of pro-inflammatory genes in in vivo neuroinflammatory models associated with microglial/immune cell activation.


Assuntos
Azepinas/administração & dosagem , Proteínas Cromossômicas não Histona/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Talidomida/análogos & derivados , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Lipopolissacarídeos , Camundongos , Microglia/metabolismo , Proteólise , Talidomida/administração & dosagem , Resultado do Tratamento
12.
Stroke ; 48(7): 1948-1956, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28588054

RESUMO

BACKGROUND AND PURPOSE: Targeting the prostaglandin I2 prostanoid (IP) receptor to reduce stroke injury has been hindered by the lack of selective drugs. MRE-269 is the active metabolite of selexipag showing a high selectivity toward the IP receptor. Selexipag has been recently approved for clinical use in pulmonary hypertension. We hypothesized that postischemic treatment with MRE-269 provides long-lasting neuroprotection with improved neurological outcomes in a clinically relevant rat stroke model. METHODS: Aged male Sprague-Dawley rats underwent transient middle cerebral artery occlusion and were randomly selected to receive either vehicle or MRE-269 (0.25 mg/kg) intravenously starting at 4.5 hours post ischemia. Accelerating rotarod and adhesive removal tests were conducted before and at 3, 7, 14, and 21 days after stroke. Infarct volume was quantified by magnetic resonance imaging at 48 hours and 21 days post middle cerebral artery occlusion. In parallel experiments, cerebral cortex samples from stroke and nonstroke sides from vehicle- and MRE-269-treated groups were collected at 18 hours post middle cerebral artery occlusion for molecular biology analyses. RESULTS: Quantitative magnetic resonance imaging data showed that postischemic MRE-269 treatment significantly reduced infarct volume compared with vehicle-treated rats at both 48 hours and 3 weeks after stroke. MRE-269 treatment resulted in a significant long-term recovery in both locomotor and somatosensory functions after middle cerebral artery occlusion, which was associated with a reduced weight loss in animals receiving the IP receptor agonist. Postischemic MRE-269 treatment reduced proinflammatory cytokines/chemokines and oxidative stress. Damage to the blood-brain barrier, as assessed by extravasation of immunoglobulin G to the ischemic brain, was significantly reduced by MRE-269, which was associated with a reduction in matrix metalloproteinase-9 activity in the brain of stroked aged rats given the IP agonist at 4.5 hours after ischemia onset. CONCLUSIONS: Our data suggest that targeting the IP receptor with MRE-269 is a novel strategy to reduce cerebral ischemia injury and promote long-term neurological recovery in ischemic stroke.


Assuntos
Acetatos/farmacologia , Isquemia Encefálica/tratamento farmacológico , Epoprostenol/análise , Pirazinas/farmacologia , Receptores de Prostaglandina/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Acetatos/administração & dosagem , Fatores Etários , Animais , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/imunologia , Infarto da Artéria Cerebral Média , Masculino , Pirazinas/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/imunologia
13.
J Neuroinflammation ; 14(1): 25, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28143498

RESUMO

BACKGROUND: The mammalian target of rapamycin (mTOR) is a kinase involved in a variety of physiological and pathological functions. However, the exact role of mTOR in excitotoxicity is poorly understood. Here, we investigated the effects of mTOR inhibition with rapamycin against neurodegeneration, and motor impairment, as well as inflammatory profile caused by an excitotoxic stimulus. METHODS: A single and unilateral striatal injection of quinolinic acid (QA) was used to induce excitotoxicity in mice. Rapamycin (250 nL of 0.2, 2, or 20 µM; intrastriatal route) was administered 15 min before QA injection. Forty-eight hours after QA administration, rotarod test was performed to evaluate motor coordination and balance. Fluoro-Jade C, Iba-1, and GFAP staining were used to evaluate neuronal cell death, microglia morphology, and astrocytes density, respectively, at this time point. Levels of cytokines and neurotrophic factors were measured by ELISA and Cytometric Bead Array 8 h after QA injection. Striatal synaptosomes were used to evaluate the release of glutamate. RESULTS: We first demonstrated that rapamycin prevented the motor impairment induced by QA. Moreover, mTOR inhibition also reduced the neurodegeneration and the production of interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α induced by excitotoxic stimulus. The lowest dose of rapamycin also increased the production of IL-10 and prevented the reduction of astrocyte density induced by QA. By using an in vitro approach, we demonstrated that rapamycin differently alters the release of glutamate from striatal synaptosomes induced by QA, reducing or enhancing the release of this neurotransmitter at low or high concentrations, respectively. CONCLUSION: Taken together, these data demonstrated a protective effect of rapamycin against an excitotoxic stimulus. Therefore, this study provides new evidence of the detrimental role of mTOR in neurodegeneration, which might represent an important target for the treatment of neurodegenerative diseases.


Assuntos
Corpo Estriado/efeitos dos fármacos , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Ácido Quinolínico/toxicidade , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Animais , Peso Corporal/efeitos dos fármacos , Corpo Estriado/fisiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/complicações , Equilíbrio Postural/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Sinaptossomos/ultraestrutura
14.
J Neurosci ; 33(45): 17814-26, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24198371

RESUMO

The striatal-enriched phosphatase (STEP) is a component of the NMDA-receptor-mediated excitotoxic signaling pathway, which plays a key role in ischemic brain injury. Using neuronal cultures and a rat model of ischemic stroke, we show that STEP plays an initial role in neuroprotection, during the insult, by disrupting the p38 MAPK pathway. Degradation of active STEP during reperfusion precedes ischemic brain damage and is associated with secondary activation of p38 MAPK. Application of a cell-permeable STEP-derived peptide that is resistant to degradation and binds to p38 MAPK protects cultured neurons from hypoxia-reoxygenation injury and reduces ischemic brain damage when injected up to 6 h after the insult. Conversely, genetic deletion of STEP in mice leads to sustained p38 MAPK activation and exacerbates brain injury and neurological deficits after ischemia. Administration of the STEP-derived peptide at the onset of reperfusion not only prevents the sustained p38 MAPK activation but also reduces ischemic brain damage in STEP KO mice. The findings indicate a neuroprotective role of STEP and suggest a potential role of the STEP-derived peptide in stroke therapy.


Assuntos
Isquemia Encefálica/metabolismo , Neurônios/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Regulação para Baixo , Masculino , Camundongos , Neurônios/citologia , Proteínas Tirosina Fosfatases não Receptoras/genética , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
J Neurochem ; 129(1): 130-42, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24225006

RESUMO

Resolution of inflammation is an emerging new strategy to reduce damage following ischemic stroke. Lipoxin A4 (LXA4 ) is an anti-inflammatory, pro-resolution lipid mediator with high affinity binding to ALX, the lipoxin A4 receptor. Since LXA4 is rapidly inactivated, potent analogs have been created, including the ALX agonist BML-111. We hypothesized that post-ischemic intravenous administration of BML-111 would provide protection to the neurovascular unit and reduce neuroinflammation in a rat stroke model. Animals were subjected to 90 min of middle cerebral artery occlusion (MCAO) and BML-111 was injected 100 min and 24 h after stroke onset and animals euthanized at 48 h. Post-ischemic treatment with BML-111 significantly reduced infarct size, decreased vasogenic edema, protected against blood-brain barrier disruption, and reduced hemorrhagic transformation. Matrix metalloproteinase-9 and matrix metalloproteinase-3 were significantly reduced following BML-111 treatment. Administration of BML-111 dramatically decreased microglial activation, as seen with CD68, and neutrophil infiltration and recruitment, as assessed by levels of myeloperoxidase and intracellular adhesion molecule-1. The tight junction protein zona occludens-1 was protected from degradation following treatment with BML-111. These results indicate that post-ischemic activation of ALX has pro-resolution effects that limit the inflammatory damage in the cerebral cortex and helps maintain blood-brain barrier integrity after ischemic stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Modelos Animais de Doenças , Ácidos Heptanoicos/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Receptores de Lipoxinas/agonistas , Acidente Vascular Cerebral/prevenção & controle , Animais , Isquemia Encefálica/patologia , Linhagem Celular Tumoral , Humanos , Injeções Intravenosas , Masculino , Ratos , Ratos Wistar , Acidente Vascular Cerebral/patologia , Fatores de Tempo
16.
Exp Neurol ; 377: 114812, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38729551

RESUMO

Ischemic stroke induces a debilitating neurological insult, where inflammatory processes contribute greatly to the expansion and growth of the injury. Receptor-interacting protein kinase 2 (RIPK2) is most well-known for its role as the obligate kinase for NOD1/2 pattern recognition receptor signaling and is implicated in the pathology of various inflammatory conditions. Compared to a sham-operated control, ischemic stroke resulted in a dramatic increase in the active, phosphorylated form of RIPK2, indicating that RIPK2 may be implicated in the response to stroke injury. Here, we assessed the effects of pharmacological inhibition of RIPK2 to improve post-stroke outcomes in mice subjected to experimental ischemic stroke. We found that treatment at the onset of reperfusion with a RIPK2 inhibitor, which inhibits the phosphorylation and activation of RIPK2, resulted in marked improvements in post-stroke behavioral outcomes compared to the vehicle-administered group assessed 24 h after stroke. RIPK2 inhibitor-treated mice exhibited dramatic reductions in infarct volume, concurrent with reduced damage to the blood-brain barrier, as evidenced by reduced levels of active matrix metalloproteinase-9 (MMP-9) and leakage of blood-borne albumin in the ipsilateral cortex. To explore the protective mechanism of RIPK2 inhibition, we next pretreated mice with RIPK2 inhibitor or vehicle and examined transcriptomic alterations occurring in the ischemic brain 6 h after stroke. We observed a dramatic reduction in neuroinflammatory markers in the ipsilateral cortex of the inhibitor-treated group while also attaining a comprehensive view of the vast transcriptomic alterations occurring in the brain with inhibitor treatment through bulk RNA-sequencing of the injured cortex. Overall, we provide significant novel evidence that RIPK2 may represent a viable target for post-stroke pharmacotherapy and potentially other neuroinflammatory conditions.


Assuntos
AVC Isquêmico , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Animais , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/antagonistas & inibidores , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Camundongos , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Masculino
17.
iScience ; 27(4): 109480, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38715940

RESUMO

Ischemic stroke is the second leading cause of death and disability worldwide, and efforts to prevent stroke, mitigate secondary neurological damage, and promote neurological recovery remain paramount. Recent findings highlight the critical importance of microbiome-related metabolites, including vitamin B12 (VB12), in alleviating toxic stroke-associated neuroinflammation. Here, we showed that VB12 tonically programmed genes supporting microglial cell division and activation and critically controlled cellular fatty acid metabolism in homeostasis. Intriguingly, VB12 promoted mitochondrial transcriptional and metabolic activities and significantly restricted stroke-associated gene alterations in microglia. Furthermore, VB12 differentially altered the functions of microglial subsets during the acute phase of ischemic stroke, resulting in reduced brain damage and improved neurological function. Pharmacological depletion of microglia before ischemic stroke abolished VB12-mediated neurological improvement. Thus, our preclinical studies highlight the relevance of VB12 in the functional programming of microglia to alleviate neuroinflammation, minimize ischemic injury, and improve host neurological recovery after ischemic stroke.

18.
J Neuroimmunol ; 385: 578239, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37976996

RESUMO

OBJECTIVES: Serum biomarkers for brain injury in neonates with congenital heart disease (CHD) provide a bedside tool for early identification and intervention. In this preliminary study, we aim to evaluate IL-18, Eotaxin-1 and Eotaxin-3 as biomarkers for the detection of brain injury in neonates with CHD. METHODS: We prospectively enrolled seven neonates diagnosed in-utero with CHD and obtained serum samples at birth, before and after surgery. Samples were analyzed using a human cytokine/chemokine multiplex assay. Brain injury was diagnosed on brain MRI before surgery. RESULTS: Samples from seven neonates at four time points before surgery and three time points after surgery were analyzed. A significant difference was found in neonates with brain injury compared to CHD neonates without. Elevations in interleukin (IL)-18 pre- and post-operative (p = 0.007), IL-18 pre-operative (p = 0.046), Eotaxin-1 pre-operative (p = 0.011), and Eotaxin-3 pre- and post-operative (p = 0.026) were found in CHD neonates with brain injury. CONCLUSION: This is the first published report on the use IL-18, Eotaxin-1, and Eotaxin-3 in the detection of brain injury for neonates with CHD. These biomarkers may provide an actionable target for neuroprotection through immunomodulation. Larger cohorts are needed to determine the significance and clinical utility of these biomarkers.


Assuntos
Lesões Encefálicas , Cardiopatias Congênitas , Recém-Nascido , Humanos , Interleucina-18 , Quimiocina CCL11 , Quimiocina CCL26 , Cardiopatias Congênitas/diagnóstico por imagem , Cardiopatias Congênitas/cirurgia , Biomarcadores
19.
Transl Stroke Res ; 14(6): 842-853, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36125734

RESUMO

Ischemic stroke is a leading cause of death and disability. However, very few neuroprotective agents have shown promise for treatment of ischemic stroke in clinical trials, despite showing efficacy in many successful preclinical studies. This may be attributed, at least in part, to the incongruency between experimental animal stroke models used in preclinical studies and the manifestation of ischemic stroke in humans. Most often the human population selected for clinical trials are more diverse than the experimental model used in a preclinical study. For successful translation, it is critical to develop clinical trial designs that match the experimental animal model used in the preclinical study. This review aims to provide a comprehensive summary of commonly used animal models with clear correlates between rodent models used to study ischemic stroke and the clinical stroke pathologies with which they most closely align. By improving the correlation between preclinical studies and clinical trials, new neuroprotective agents and stroke therapies may be more accurately and efficiently identified.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Fármacos Neuroprotetores , Acidente Vascular Cerebral , Animais , Humanos , AVC Isquêmico/complicações , Fármacos Neuroprotetores/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Modelos Animais de Doenças , Isquemia Encefálica/complicações
20.
Curr Neuropharmacol ; 21(2): 235-259, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36503452

RESUMO

The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.


Assuntos
Estimulantes do Sistema Nervoso Central , Cocaína , Transtornos Relacionados ao Uso de Substâncias , Humanos , Microglia , Cocaína/farmacologia , Anfetaminas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA