Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 29(13)2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38998988

RESUMO

Naturally occurring homoisoflavonoids isolated from some Liliaceae plants have been reported to have diverse biological activities (e.g., antioxidant, anti-inflammatory, and anti-angiogenic effects). The exact mechanism by which homoisoflavonones exert anti-neuroinflammatory effects against activated microglia-induced inflammatory cascades has not been well studied. Here, we aimed to explore the mechanism of homoisoflavonoid SH66 having a potential anti-inflammatory effect in lipopolysaccharide (LPS)-primed BV2 murine microglial cells. Microglia cells were pre-treated with SH66 followed by LPS (100 ng/mL) activation. SH66 treatment attenuated the production of inflammatory mediators, including nitric oxide and proinflammatory cytokines, by down-regulating mitogen-activated protein kinase signaling in LPS-activated microglia. The SH66-mediated inhibition of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome complex and the respective inflammatory biomarker-like active interleukin (IL)-1ß were noted to be one of the key pathways of the anti-inflammatory effect. In addition, SH66 increased the neurite length in the N2a neuronal cell and the level of nerve growth factor in the C6 astrocyte cell. Our results demonstrated the anti-neuroinflammatory effect of SH66 against LPS-activated microglia-mediated inflammatory events by down-regulating the NLRP3 inflammasome complex, with respect to its neuroprotective effect. SH66 could be an interesting candidate for further research and development regarding prophylactics and therapeutics for inflammation-mediated neurological complications.


Assuntos
Anti-Inflamatórios , Lipopolissacarídeos , Microglia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Lipopolissacarídeos/farmacologia , Animais , Camundongos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Linhagem Celular , Isoflavonas/farmacologia , Isoflavonas/química , Citocinas/metabolismo , Óxido Nítrico/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo
2.
Cell Mol Neurobiol ; 42(8): 2505-2525, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34460037

RESUMO

Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.


Assuntos
Isquemia Encefálica , Microglia , Anti-Inflamatórios/farmacologia , Isquemia Encefálica/metabolismo , Citocinas/metabolismo , Humanos , Microglia/metabolismo , Neuroproteção
3.
Pharmacol Res ; 165: 105419, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33450385

RESUMO

The search for novel therapeutic agents for the management of cerebral ischemia/stroke has become an appealing research interest in the recent past. Neuroprotective phytochemicals as novel stroke drug candidates have recently drawn significant interests from stroke scientists due to their strong brain protective effects in animal stroke models. The underlying mechanism of action is likely owing to their anti-inflammatory properties, even though other mechanisms such as anti-oxidation and vasculoprotection have also been proposed. It is generally held that the early proinflammatory responses after stroke can lead to a secondary brain injury, mainly due to the damaging effect exerted by over-activation of brain resident microglial cells and infiltration of circulating monocytes and macrophages. This review focuses on the anti-inflammatory properties of bioactive phytochemicals, including activation and polarization of microglia/macrophages in the post-ischemic brain. The latest studies in animal stroke models demonstrate that this group of bioactive phytochemicals exerts their anti-inflammatory effects via attenuation of brain proinflammatory microglia and macrophages M1 polarization while promoting anti-inflammatory microglial and macrophages M2 polarization. As a result, stroked animals treated with brain protective phytochemicals have significantly fewer brain active M1 microglia and macrophages, smaller brain infarct volume, better functional recovery, and better survival rate. Therefore, this review provides insights into a new category of drug candidates for stroke drug development by employing neuroprotective phytochemicals.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Compostos Fitoquímicos/uso terapêutico , Animais , Humanos , Doenças Neuroinflamatórias/tratamento farmacológico
4.
Pharmacol Res ; 169: 105661, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33971269

RESUMO

Tanshinones, lipophilic diterpenes isolated from the rhizome of Salvia miltiorrhiza, have diverse pharmacological activities against human ailments including neurological diseases. In fact, tanshinones have been used to treat heart diseases, stroke, and vascular diseases in traditional Chinese medicine. During the last decade, tanshinones have been the most widely studied phytochemicals for their neuroprotective effects against experimental models of cerebral ischemia and Alzheimer's diseases. Importantly, tanshinone IIA, mostly studied tanshinone for biological activities, is recently reported to attenuate blood-brain barrier permeability among stroke patients, suggesting tanshinone IIA as an appealing therapeutic candidate for neurological diseases. Tanshinone I and IIA are also effective in experimental models of Parkinson's disease, Multiple sclerosis, and other neuroinflammatory diseases. In addition, several experimental studies suggested the pleiotropic neuroprotective effects of tanshinones such as anti-inflammatory, antioxidant, anti-apoptotic, and BBB protectant further value aiding to tanshinone as an appealing therapeutic strategy in neurological diseases. Therefore, in this review, we aimed to compile the recent updates and cellular and molecular mechanisms of neuroprotection of tanshinone IIA in diverse neurological diseases.


Assuntos
Abietanos/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Animais , Isquemia Encefálica/tratamento farmacológico , Humanos , Esclerose Múltipla/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico
5.
Int J Mol Sci ; 22(15)2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34360625

RESUMO

Activation of microglia and/or astrocytes often releases proinflammatory molecules as critical pathogenic mediators that can promote neuroinflammation and secondary brain damages in diverse diseases of the central nervous system (CNS). Therefore, controlling the activation of glial cells and their neuroinflammatory responses has been considered as a potential therapeutic strategy for treating neuroinflammatory diseases. Recently, receptor-mediated lysophospholipid signaling, sphingosine 1-phosphate (S1P) receptor- and lysophosphatidic acid (LPA) receptor-mediated signaling in particular, has drawn scientific interest because of its critical roles in pathogenies of diverse neurological diseases such as neuropathic pain, systemic sclerosis, spinal cord injury, multiple sclerosis, cerebral ischemia, traumatic brain injury, hypoxia, hydrocephalus, and neuropsychiatric disorders. Activation of microglia and/or astrocytes is a common pathogenic event shared by most of these CNS disorders, indicating that lysophospholipid receptors could influence glial activation. In fact, many studies have reported that several S1P and LPA receptors can influence glial activation during the pathogenesis of cerebral ischemia and multiple sclerosis. This review aims to provide a comprehensive framework about the roles of S1P and LPA receptors in the activation of microglia and/or astrocytes and their neuroinflammatory responses in CNS diseases.


Assuntos
Astrócitos/metabolismo , Doenças do Sistema Nervoso Central/metabolismo , Neuroglia/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Humanos
6.
Int J Mol Sci ; 22(9)2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946349

RESUMO

Nitric oxide (NO) is a neurotransmitter that mediates the activation and inhibition of inflammatory cascades. Even though physiological NO is required for defense against various pathogens, excessive NO can trigger inflammatory signaling and cell death through reactive nitrogen species-induced oxidative stress. Excessive NO production by activated microglial cells is specifically associated with neuroinflammatory and neurodegenerative conditions, such as Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, ischemia, hypoxia, multiple sclerosis, and other afflictions of the central nervous system (CNS). Therefore, controlling excessive NO production is a desirable therapeutic strategy for managing various neuroinflammatory disorders. Recently, phytochemicals have attracted considerable attention because of their potential to counteract excessive NO production in CNS disorders. Moreover, phytochemicals and nutraceuticals are typically safe and effective. In this review, we discuss the mechanisms of NO production and its involvement in various neurological disorders, and we revisit a number of recently identified phytochemicals which may act as NO inhibitors. This review may help identify novel potent anti-inflammatory agents that can downregulate NO, specifically during neuroinflammation and neurodegeneration.


Assuntos
Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/metabolismo , Compostos Fitoquímicos/farmacologia , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Descoberta de Drogas , Humanos , Inflamação/metabolismo , Terapia de Alvo Molecular , Doenças Neurodegenerativas/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/uso terapêutico , Óxido Nítrico/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Compostos Fitoquímicos/química , Compostos Fitoquímicos/uso terapêutico , Espécies Reativas de Nitrogênio/metabolismo
7.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-33669456

RESUMO

The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2/COVID-19), is a worldwide pandemic, as declared by the World Health Organization (WHO). It is a respiratory virus that infects people of all ages. Although it may present with mild to no symptoms in most patients, those who are older, immunocompromised, or with multiple comorbidities may present with severe and life-threatening infections. Throughout history, nutraceuticals, such as a variety of phytochemicals from medicinal plants and dietary supplements, have been used as adjunct therapies for many disease conditions, including viral infections. Appropriate use of these adjunct therapies with antiviral proprieties may be beneficial in the treatment and/or prophylaxis of COVID-19. In this review, we provide a comprehensive summary of nutraceuticals, such as vitamins C, D, E, zinc, melatonin, and other phytochemicals and function foods. These nutraceuticals may have potential therapeutic efficacies in fighting the threat of the SARS-CoV-2/COVID-19 pandemic.


Assuntos
Tratamento Farmacológico da COVID-19 , Suplementos Nutricionais , Melatonina/uso terapêutico , Vitaminas/uso terapêutico , Zinco/uso terapêutico , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Ácido Ascórbico/farmacologia , Ácido Ascórbico/uso terapêutico , Suplementos Nutricionais/análise , Alimento Funcional/análise , Humanos , Melatonina/farmacologia , SARS-CoV-2/efeitos dos fármacos , Vitamina D/farmacologia , Vitamina D/uso terapêutico , Vitamina E/farmacologia , Vitamina E/uso terapêutico , Vitaminas/farmacologia , Zinco/farmacologia
8.
Int J Mol Sci ; 21(22)2020 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-33202644

RESUMO

Lysophosphatidic acid receptor 1 (LPA1) contributes to brain injury following transient focal cerebral ischemia. However, the mechanism remains unclear. Here, we investigated whether nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation might be an underlying mechanism involved in the pathogenesis of brain injury associated with LPA1 following ischemic challenge with transient middle cerebral artery occlusion (tMCAO). Suppressing LPA1 activity by its antagonist attenuated NLRP3 upregulation in the penumbra and ischemic core regions, particularly in ionized calcium-binding adapter molecule 1 (Iba1)-expressing cells like macrophages of mouse after tMCAO challenge. It also suppressed NLRP3 inflammasome activation, such as caspase-1 activation, interleukin 1ß (IL-1ß) maturation, and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) speck formation, in a post-ischemic brain. The role of LPA1 in NLRP3 inflammasome activation was confirmed in vitro using lipopolysaccharide-primed bone marrow-derived macrophages, followed by LPA exposure. Suppressing LPA1 activity by either pharmacological antagonism or genetic knockdown attenuated NLRP3 upregulation, caspase-1 activation, IL-1ß maturation, and IL-1ß secretion in these cells. Furthermore, nuclear factor-κB (NF-κB), extracellular signal-regulated kinase 1/2 (ERK1/2), and p38 were found to be LPA1-dependent effector pathways in these cells. Collectively, results of the current study first demonstrate that LPA1 could contribute to ischemic brain injury by activating NLRP3 inflammasome with underlying effector mechanisms.


Assuntos
Lesões Encefálicas/metabolismo , Ataque Isquêmico Transitório/metabolismo , Lisofosfolipídeos/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Caspase 1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Interleucina-1beta/metabolismo , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR
9.
Int J Mol Sci ; 21(3)2020 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-31991572

RESUMO

Tumor necrosis factor-alpha (TNF-α) is a well-known pro-inflammatory cytokine responsible for the modulation of the immune system. TNF-α plays a critical role in almost every type of inflammatory disorder, including central nervous system (CNS) diseases. Although TNF-α is a well-studied component of inflammatory responses, its functioning in diverse cell types is still unclear. TNF-α functions through its two main receptors: tumor necrosis factor receptor 1 and 2 (TNFR1, TNFR2), also known as p55 and p75, respectively. Normally, the functions of soluble TNF-α-induced TNFR1 activation are reported to be pro-inflammatory and apoptotic. While TNF-α mediated TNFR2 activation has a dual role. Several synthetic drugs used as inhibitors of TNF-α for diverse inflammatory diseases possess serious adverse effects, which make patients and researchers turn their focus toward natural medicines, phytochemicals in particular. Phytochemicals targeting TNF-α can significantly improve disease conditions involving TNF-α with fewer side effects. Here, we reviewed known TNF-α inhibitors, as well as lately studied phytochemicals, with a role in inhibiting TNF-α itself, and TNF-α-mediated signaling in inflammatory diseases focusing mainly on CNS disorders.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Compostos Fitoquímicos/uso terapêutico , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa , Animais , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
10.
J Neuroinflammation ; 16(1): 170, 2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-31429777

RESUMO

BACKGROUND: Lysophosphatidic acid receptor 1 (LPA1) is in the spotlight because its synthetic antagonist has been under clinical trials for lung fibrosis and psoriasis. Targeting LPA1 might also be a therapeutic strategy for cerebral ischemia because LPA1 triggers microglial activation, a core pathogenesis in cerebral ischemia. Here, we addressed this possibility using a mouse model of transient middle cerebral artery occlusion (tMCAO). METHODS: To address the role of LPA1 in the ischemic brain damage, we used AM095, a selective LPA1 antagonist, as a pharmacological tool and lentivirus bearing a specific LPA1 shRNA as a genetic tool. Brain injury after tMCAO challenge was accessed by determining brain infarction and neurological deficit score. Role of LPA1 in tMCAO-induced microglial activation was ascertained by immunohistochemical analysis. Proinflammatory responses in the ischemic brain were determined by qRT-PCR and immunohistochemical analyses, which were validated in vitro using mouse primary microglia. Activation of MAPKs and PI3K/Akt was determined by Western blot analysis. RESULTS: AM095 administration immediately after reperfusion attenuated brain damage such as brain infarction and neurological deficit at 1 day after tMCAO, which was reaffirmed by LPA1 shRNA lentivirus. AM095 administration also attenuated brain infarction and neurological deficit at 3 days after tMCAO. LPA1 antagonism attenuated microglial activation; it reduced numbers and soma size of activated microglia, reversed their morphology into less toxic one, and reduced microglial proliferation. Additionally, LPA1 antagonism reduced mRNA expression levels of proinflammatory cytokines and suppressed NF-κB activation, demonstrating its regulatory role of proinflammatory responses in the ischemic brain. Particularly, these LPA1-driven proinflammatory responses appeared to occur in activated microglia because NF-κB activation occurred mainly in activated microglia in the ischemic brain. Regulatory role of LPA1 in proinflammatory responses of microglia was further supported by in vitro findings using lipopolysaccharide-stimulated cultured microglia, showing that suppressing LPA1 activity reduced mRNA expression levels of proinflammatory cytokines. In the ischemic brain, LPA1 influenced PI3K/Akt and MAPKs; suppressing LPA1 activity decreased MAPK activation and increased Akt phosphorylation. CONCLUSION: This study demonstrates that LPA1 is a new etiological factor for cerebral ischemia, strongly indicating that its modulation can be a potential strategy to reduce ischemic brain damage.


Assuntos
Lesões Encefálicas/metabolismo , Ataque Isquêmico Transitório/metabolismo , Microglia/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Lesões Encefálicas/patologia , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Microglia/patologia
11.
J Neuroinflammation ; 15(1): 284, 2018 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-30305119

RESUMO

BACKGROUND: The pathogenic roles of receptor-mediated sphingosine 1-phosphate (S1P) signaling in cerebral ischemia have been evidenced mainly through the efficacy of FTY720 that binds non-selectively to four of the five S1P receptors (S1P1,3,4,5). Recently, S1P1 and S1P2 were identified as specific receptor subtypes that contribute to brain injury in cerebral ischemia; however, the possible involvement of other S1P receptors remains unknown. S1P3 can be the candidate because of its upregulation in the ischemic brain, which was addressed in this study, along with underlying pathogenic mechanisms. METHODS: We used transient middle cerebral artery occlusion/reperfusion (tMCAO), a mouse model of transient focal cerebral ischemia. To identify S1P3 as a pathogenic factor in cerebral ischemia, we employed a specific S1P3 antagonist, CAY10444. Brain damages were assessed by brain infarction, neurological score, and neurodegeneration. Histological assessment was carried out to determine microglial activation, morphological transformation, and proliferation. M1/M2 polarization and relevant signaling pathways were determined by biochemical and immunohistochemical analysis. RESULTS: Inhibiting S1P3 immediately after reperfusion with CAY10444 significantly reduced tMCAO-induced brain infarction, neurological deficit, and neurodegeneration. When S1P3 activity was inhibited, the number of activated microglia was markedly decreased in both the periischemic and ischemic core regions in the ischemic brain 1 and 3 days following tMCAO. Moreover, inhibiting S1P3 significantly restored the microglial shape from amoeboid to ramified microglia in the ischemic core region 3 days after tMCAO, and it attenuated microglial proliferation in the ischemic brain. In addition to these changes, S1P3 signaling influenced the proinflammatory M1 polarization, but not M2. The S1P3-dependent regulation of M1 polarization was clearly shown in activated microglia, which was affirmed by determining the in vivo activation of microglial NF-κB signaling that is responsible for M1 and in vitro expression levels of proinflammatory cytokines in activated microglia. As downstream effector pathways in an ischemic brain, S1P3 influenced phosphorylation of ERK1/2, p38 MAPK, and Akt. CONCLUSIONS: This study identified S1P3 as a pathogenic mediator in an ischemic brain along with underlying mechanisms, involving its modulation of microglial activation and M1 polarization, further suggesting that S1P3 can be a therapeutic target for cerebral ischemia.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Polaridade Celular/fisiologia , Infarto da Artéria Cerebral Média/complicações , Microglia/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fluoresceínas/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas dos Microfilamentos/metabolismo , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Receptores de Esfingosina-1-Fosfato , Tiazolidinas/uso terapêutico
12.
J Tradit Chin Med ; 34(1): 69-75, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25102694

RESUMO

OBJECTIVE: To investigate the neuroprotective effects of Fructus Chebulae extract using both in vivo and in vitro models of cerebral ischemia. METHODS: As an in vitro model, oxygen glucose deprivation followed by reoxygenation (OGD-R) and hydrogen peroxide (H2O2) induced cellular damage in rat pheochromocytoma (PC12) cells was used to investigate the neuroprotective effects of extract of Fructus Chebulae. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to calculate cell survival. For in vivo, occlusion of left middle cerebral artery on rats was carried out as a focal cerebral ischemic model. RESULTS: Fructus Chebulae extract increases the PC12 cell survival against OGD-R and H2O2 by 68% and 91.4% respectively. Fructus Chebulae also decreases the cerebral infarct volume by 39% and extent of hemisphere swelling from 17% in control group to 10% in Fructus Chebulae treated group. CONCLUSION: Fructus Chebulae, as a traditional medicine, can rescue the neuronal cell death against ischemia related damage. The possible mechanism for the neuroprotection might be the inhibition of oxidative damages occurring after acute phase of cerebral ischemia.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Combretaceae/química , Medicamentos de Ervas Chinesas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Animais , Isquemia Encefálica/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
13.
Front Neurosci ; 18: 1393293, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38770241

RESUMO

While recent advances in diagnostics and therapeutics offer promising new approaches for Alzheimer's disease (AD) diagnosis and treatment, there is still an unmet need for an effective remedy, suggesting new avenues of research are required. Besides many plausible etiologies for AD pathogenesis, mounting evidence supports a possible role for microbial infections. Various microbes have been identified in the postmortem brain tissues of human AD patients. Among bacterial pathogens in AD, Chlamydia pneumoniae (Cp) has been well characterized in human AD brains and is a leading candidate for an infectious involvement. However, no definitive studies have been performed proving or disproving Cp's role as a causative or accelerating agent in AD pathology and cognitive decline. In this review, we discuss recent updates for the role of Cp in human AD brains as well as experimental models of AD. Furthermore, based on the current literature, we have compiled a list of potential mechanistic pathways which may connect Cp with AD pathology.

14.
Prog Retin Eye Res ; 101: 101273, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38759947

RESUMO

The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid ß-protein (Aß) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aß deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.


Assuntos
Doença de Alzheimer , Retina , Doenças Retinianas , Doença de Alzheimer/fisiopatologia , Humanos , Doenças Retinianas/fisiopatologia , Doenças Retinianas/diagnóstico , Retina/fisiopatologia , Animais , Tomografia de Coerência Óptica/métodos , Peptídeos beta-Amiloides/metabolismo , Vasos Retinianos/fisiopatologia , Vasos Retinianos/diagnóstico por imagem
15.
Molecules ; 18(3): 3529-42, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23519197

RESUMO

Terminalia chebula, native to Southeast Asia, is a popular medicinal plant in Ayurveda. It has been previously reported to have strong antioxidant and anti-inflammatory efficacy. In this study, we aimed to investigate if fruit extract from T. chebula might protect neuronal cells against ischemia and related diseases by reduction of oxidative damage and inflammation in rat pheochromocytoma cells (PC12) using in vitro oxygen-glucose deprivation followed by reoxygenation (OGD-R) ischemia and hydrogen peroxide (H2O2) induced cell death. Cell survival was evaluated by a 2-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Free radical scavenging, lipid peroxidation and nitric oxide inhibition were measured by diphenyl-1-picrylhydrazyl (DPPH), thiobarbituric acid (TBA) and Griess reagent, respectively. We found that T. chebula extract: (1) increases the survival of cells subjected to OGD-R by 68%, and H2O2 by 91.4%; (2) scavenges the DPPH free radical by 96% and decreases malondialdehyde (MDA) levels from 237.0 ± 15.2% to 93.7 ± 2.2%; (3) reduces NO production and death rate of microglia cells stimulated by lipopolysaccharide (LPS). These results suggest that T. chebula extract has the potential as a natural herbal medicine, to protect the cells from ischemic damage and the possible mechanism might be the inhibition of oxidative and inflammatory processes.


Assuntos
Morte Celular/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Microglia/imunologia , Extratos Vegetais/farmacologia , Terminalia/química , Animais , Compostos de Bifenilo/química , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/isolamento & purificação , Radicais Livres/química , Glucose/deficiência , Peróxido de Hidrogênio/farmacologia , Peroxidação de Lipídeos , Lipopolissacarídeos/farmacologia , Malondialdeído/metabolismo , Microglia/efeitos dos fármacos , Oxidantes/farmacologia , Estresse Oxidativo , Células PC12 , Picratos/química , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Ratos
16.
Ageing Res Rev ; 84: 101819, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36526257

RESUMO

Acetylcholinesterase inhibitors (ChEI) are the global standard of care for the symptomatic treatment of Alzheimer's disease (AD) and show significant positive effects in neurodegenerative diseases with cognitive and behavioral symptoms. Although experimental and large-scale clinical evidence indicates the potential long-term efficacy of ChEI, primary outcomes are generally heterogeneous across outpatient clinics and regional healthcare systems. Sub-optimal dosing or slow tapering, heterogeneous guidelines about the timing for therapy initiation (prodromal versus dementia stages), healthcare providers' ambivalence to treatment, lack of disease awareness, delayed medical consultation, prescription of ChEI in non-AD cognitive disorders, contribute to the negative outcomes. We present an evidence-based overview of determinants, spanning genetic, molecular, and large-scale networks, involved in the response to ChEI in patients with AD and other neurodegenerative diseases. A comprehensive understanding of cerebral and retinal cholinergic system dysfunctions along with ChEI response predictors in AD is crucial since disease-modifying therapies will frequently be prescribed in combination with ChEI. Therapeutic algorithms tailored to genetic, biological, clinical (endo)phenotypes, and disease stages will help leverage inter-drug synergy and attain optimal combined response outcomes, in line with the precision medicine model.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/diagnóstico , Acetilcolinesterase/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Medicina de Precisão
17.
Front Immunol ; 14: 1155935, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37325640

RESUMO

Introduction: Osteopontin (OPN; also known as SPP1), an immunomodulatory cytokine highly expressed in bone marrow-derived macrophages (BMMΦ), is known to regulate diverse cellular and molecular immune responses. We previously revealed that glatiramer acetate (GA) stimulation of BMMΦ upregulates OPN expression, promoting an anti-inflammatory, pro-healing phenotype, whereas OPN inhibition triggers a pro-inflammatory phenotype. However, the precise role of OPN in macrophage activation state is unknown. Methods: Here, we applied global proteome profiling via mass spectrometry (MS) analysis to gain a mechanistic understanding of OPN suppression versus induction in primary macrophage cultures. We analyzed protein networks and immune-related functional pathways in BMMΦ either with OPN knockout (OPNKO) or GA-mediated OPN induction compared with wild type (WT) macrophages. The most significant differentially expressed proteins (DEPs) were validated using immunocytochemistry, western blot, and immunoprecipitation assays. Results and discussion: We identified 631 DEPs in OPNKO or GA-stimulated macrophages as compared to WT macrophages. The two topmost downregulated DEPs in OPNKO macrophages were ubiquitin C-terminal hydrolase L1 (UCHL1), a crucial component of the ubiquitin-proteasome system (UPS), and the anti-inflammatory Heme oxygenase 1 (HMOX-1), whereas GA stimulation upregulated their expression. We found that UCHL1, previously described as a neuron-specific protein, is expressed by BMMΦ and its regulation in macrophages was OPN-dependent. Moreover, UCHL1 interacted with OPN in a protein complex. The effects of GA activation on inducing UCHL1 and anti-inflammatory macrophage profiles were mediated by OPN. Functional pathway analyses revealed two inversely regulated pathways in OPN-deficient macrophages: activated oxidative stress and lysosome-mitochondria-mediated apoptosis (e.g., ROS, Lamp1-2, ATP-synthase subunits, cathepsins, and cytochrome C and B subunits) and inhibited translation and proteolytic pathways (e.g., 60S and 40S ribosomal subunits and UPS proteins). In agreement with the proteome-bioinformatics data, western blot and immunocytochemical analyses revealed that OPN deficiency perturbs protein homeostasis in macrophages-inhibiting translation and protein turnover and inducing apoptosis-whereas OPN induction by GA restores cellular proteostasis. Taken together, OPN is essential for macrophage homeostatic balance via the regulation of protein synthesis, UCHL1-UPS axis, and mitochondria-mediated apoptotic processes, indicating its potential application in immune-based therapies.


Assuntos
Osteopontina , Complexo de Endopeptidases do Proteassoma , Osteopontina/genética , Osteopontina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteostase , Proteoma/metabolismo , Macrófagos , Mitocôndrias/metabolismo , Apoptose
18.
Molecules ; 17(12): 14765-77, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23519251

RESUMO

This study evaluated the anti-obesity effects of HT048, a combination of C. pinnatifida fruit and C. unshiu peel extracts, in high-fat diet (HFD)-induced obese rats. 4-Week-old male Sprague Dawley (SD) rats were divided into normal and high fat diet (HFD) groups. The HFD groups were further divided into five groups treated with distilled water, orlistat (40 mg/kg, twice daily, p.o) and HT048 (30, 100 and 300 mg/kg, twice daily, p.o.) for 12 weeks. Orlistat, an anti-obesity drug, was used as positive control in the HFD-induced obese rats. We measured the food intake, body weight, epididymal adipose tissue and liver weights, and serum total cholesterol (TC), triglyceride (TG), alanine transaminase (ALT), and aspartate aminotransferase (AST) levels. The body weight and epididymal adipose tissue and liver weights of the HT048 100 and 300 mg/kg treated groups were significantly lower than that of the HFD control group. Also, serum TC, TG, ALT, and AST levels in the HT048 100 and 300 mg/kg treated groups were significantly decreased. Moreover, the orlistat treated group showed significantly reduced body weight and improved serum lipoprotein, compared with the HFD control group. These results show that HT048 supplements improved obesity-related body weight and serum lipoprotein parameters in a HFD-induced obese rat model.


Assuntos
Fármacos Antiobesidade/farmacologia , Dieta Hiperlipídica , Obesidade/tratamento farmacológico , Fitoterapia , Extratos Vegetais/farmacologia , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Crataegus/química , Gorduras na Dieta/administração & dosagem , Sinergismo Farmacológico , Frutas/química , Lactonas/farmacologia , Masculino , Orlistate , Ratos , Ratos Sprague-Dawley , Triglicerídeos/sangue
19.
ACS Chem Neurosci ; 12(14): 2562-2572, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34251185

RESUMO

Despite being a major global health concern, cerebral ischemia/stroke has limited therapeutic options. Tissue plasminogen activator (tPA) is the only available medication to manage acute ischemic stroke, but this medication is associated with adverse effects and has a narrow therapeutic time window. Curcumin, a polyphenol that is abundantly present in the rhizome of the turmeric plant (Curcuma longa), has shown promising neuroprotective effects in animal models of neurodegenerative diseases, including cerebral ischemia. In the central nervous system (CNS), neuroprotective effects of curcumin have been experimentally validated in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and cerebral ischemia. Curcumin can exert pleiotropic effects in the postischemic brain including antioxidant, anti-inflammatory, antiapoptotic, vasculoprotective, and direct neuroprotective efficacies. Importantly, neuroprotective effects of curcumin has been reported in both ischemic and hemorrhagic stroke models. A broad-spectrum neuroprotective efficacy of curcumin suggested that curcumin can be an appealing therapeutic strategy to treat cerebral ischemia. In this review, we aimed to address the pharmacotherapeutic potential of curcumin in cerebral ischemia including its cellular and molecular mechanisms of neuroprotection revealing curcumin as an appealing therapeutic candidate for cerebral ischemia.


Assuntos
Isquemia Encefálica , Curcumina , Fármacos Neuroprotetores , Acidente Vascular Cerebral , Animais , Isquemia Encefálica/tratamento farmacológico , Curcumina/farmacologia , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Ativador de Plasminogênio Tecidual
20.
Neuromolecular Med ; 23(1): 211-223, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32914259

RESUMO

Sphingosine 1-phosphate (S1P) is an important lipid biomolecule that exerts pleiotropic cellular actions as it binds to and activates its five G-protein-coupled receptors, S1P1-5. Through these receptors, S1P can mediate diverse biological activities in both healthy and diseased conditions. S1P is produced by S1P-producing enzymes, sphingosine kinases (SphK1 and SphK2), and is abundantly present in different organs, including the brain. The medically important roles of receptor-mediated S1P signaling are well characterized in multiple sclerosis because FTY720 (Gilenya™, Novartis), a non-selective S1P receptor modulator, is currently used as a treatment for this disease. In cerebral ischemia, its role is also notable because of FTY720's efficacy in both rodent models and human patients with cerebral ischemia. In particular, some of the S1P receptors, including S1P1, S1P2, and S1P3, have been identified as pathogenic players in cerebral ischemia. Other than these receptors, S1P itself and S1P-producing enzymes have been shown to play certain roles in cerebral ischemia. This review aims to compile the current updates and overviews about the roles of S1P signaling, along with a focus on S1P receptors in cerebral ischemia, based on recent studies that used in vivo rodent models of cerebral ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Lisofosfolipídeos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Esfingosina/análogos & derivados , Animais , Dano Encefálico Crônico/etiologia , Dano Encefálico Crônico/metabolismo , Isquemia Encefálica/complicações , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Cloridrato de Fingolimode/uso terapêutico , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Inflamação , AVC Isquêmico/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Ratos , Transdução de Sinais/fisiologia , Esfingosina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA