Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-475015

RESUMO

Accumulating evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes various neurological symptoms in coronavirus disease 2019 (COVID-19) patients. The most dominant immune cells in the brain are microglia. Yet, the relationship between neurological manifestations, neuroinflammation, and host immune response of microglia to SARS-CoV-2 has not been well characterized. Here, we report that SARS-CoV-2 can directly infect human microglia, eliciting M1-like pro-inflammatory responses, followed by cytopathic effects. Specifically, SARS-CoV-2 infected human microglial clone 3 (HMC3), leading to inflammatory activation and cell death. RNA-seq analysis also revealed that ER stress and immune responses were induced in the early and apoptotic processes in the late phase of viral infection. SARS-CoV-2-infected HMC3 showed the M1 phenotype and produced pro-inflammatory cytokines such as interleukin (IL)-1{beta}, IL-6, and tumour necrosis factor (TNF-), but not the anti-inflammatory cytokine IL-10. After this pro-inflammatory activation, SARS-CoV-2 infection promoted both intrinsic and extrinsic death receptor-mediated apoptosis in HMC3. Using K18-hACE2 transgenic mice, murine microglia were also infected by intranasal inoculation of SARS-CoV-2. This infection induced the acute production of pro-inflammatory microglial IL-6 and TNF- and provoked a chronic loss of microglia. Our findings suggest that microglia are potential mediators of SARS-CoV-2-induced neurological problems and, consequently, can be targets of therapeutic strategies against neurological diseases in COVID-19 patients. IMPORTANCERecent studies reported neurological manifestations and complications in COVID-19 patients, which are associated with neuroinflammation. As microglia are the dominant immune cells in brains, it needs to be elucidate the relationship between neuroinflammation and host immune response of microglia to SARS-CoV-2. Here, we suggest that SARS-CoV-2 can directly infect human microglia with cytopathic effect (CPE) using human microglial clone 3 (HMC3). The infected microglia were promoted to pro-inflammatory activation following apoptotic cell death. This pro-inflammatory activation was accompanied by the high production of pro-inflammatory cytokines, and led to neurotoxic-M1 phenotype polarization. In vivo, murine microglia were infected and produced pro-inflammatory cytokines and provoked a chronic loss using K18-hACE2 mice. Thus, our data present that SARS-CoV-2-infected microglia are potential mediators of neurological problems in COVID-19 patients. In addition, HMC3 cells are susceptible to SARS-CoV-2 and exhibit the CPE, which can be further used to investigate cellular and molecular mechanisms of neuroinflammation reported in COVID-19 patients.

2.
Experimental Neurobiology ; : 289-299, 2019.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-739537

RESUMO

Transient receptor potential vanilloid subtype 1 (TRPV1) on astrocytes prevents ongoing degeneration of nigrostriatal dopamine (DA) neurons in MPP⁺-lesioned rats via ciliary neurotrophic factor (CNTF). The present study determined whether such a beneficial effect of astrocytic TRPV1 could be achieved after completion of injury of DA neurons, rather than ongoing injury, which seems more relevant to therapeutics. To test this, the MPP⁺-lesioned rat model utilized here exhibited approximately 70~80% degeneration of nigrostriatal DA neurons that was completed at 2 weeks post medial forebrain bundle injection of MPP⁺. TRPV1 agonist, capsaicin (CAP), was intraperitoneally administered. CNTF receptor alpha neutralizing antibody (CNTFRαNAb) was nigral injected to evaluate the role of CNTF endogenously produced by astrocyte through TRPV1 activation on DA neurons. Delayed treatment of CAP produced a significant reduction in amphetamine-induced rotational asymmetry. Accompanying this behavioral recovery, CAP treatment increased CNTF levels and tyrosine hydroxylase (TH) activity in the substantia nigra pars compacta (SNpc), and levels of DA and its metabolites in the striatum compared to controls. Interestingly, behavioral recovery and increases in biochemical indices were not reflected in trophic changes of the DA system. Instead, behavioral recovery was temporal and dependent on the continuous presence of CAP treatment. The results suggest that delayed treatment of CAP increases nigral TH enzyme activity and striatal levels of DA and its metabolites by CNTF endogenously derived from CAP-activated astrocytes through TRPV1, leading to functional recovery. Consequently, these findings may be useful in the treatment of DA imbalances associated with Parkinson's disease.


Assuntos
Animais , Ratos , Anticorpos Neutralizantes , Astrócitos , Capsaicina , Fator Neurotrófico Ciliar , Dopamina , Neurônios Dopaminérgicos , Feixe Prosencefálico Mediano , Modelos Animais , Neurônios , Doença de Parkinson , Parte Compacta da Substância Negra , Receptor do Fator Neutrófico Ciliar , Tirosina 3-Mono-Oxigenase
3.
Experimental Neurobiology ; : 309-319, 2018.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-716236

RESUMO

The present study investigated the effects of interleukin (IL)-4 on dopamine (DA) neurons in the substantia nigra (SN) in vivo of lipopolysaccharide (LPS)-treated rat. Tyrosine hydroxylase immunohistochemistry showed a significant loss of nigral DA neurons at 3 and 7 day post-LPS. In parallel, IL-4 immunoreactivity was upregulated as early as 1 day, reached a peak at 3 day and remained elevated at 7 day post-LPS. IL-4 immunoreactivity was detected exclusively in microglia. IL-4 neutralizing antibody (NA) significantly increased survival of DA neurons in LPS-treated SN in vivo by inhibiting microglial activation and production of proinflammatory mediator such as IL-1β as assessed by immunihistochemical, RT-PCR and ELISA analysis, respectively. Accompanying neuroprotection are IL-4NA effects on decreased disruption of blood-brain barrier and astrocytes. The present data suggest that endogenously expressed IL-4 from reactive microglia may be involved in the neuropathological processes of degeneration of DA neurons occurring in Parkinson's disease.


Assuntos
Animais , Ratos , Anticorpos Neutralizantes , Astrócitos , Barreira Hematoencefálica , Dopamina , Neurônios Dopaminérgicos , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Interleucina-4 , Interleucinas , Lipopolissacarídeos , Microglia , Neurônios , Neuroproteção , Doença de Parkinson , Substância Negra , Tirosina 3-Mono-Oxigenase
4.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-914286

RESUMO

We recently reported that adeno-associated virus serotype 1 (AAV1) transduction of murine nigral dopaminergic (DA) neurons with constitutively active ras homolog enriched in brain with a mutation of serine to histidine at position 16 [Rheb(S16H)] induced the production of neurotrophic factors, resulting in neuroprotective effects on the nigrostriatal DA system in animal models of Parkinson’s disease (PD). To further investigate whether AAV1-Rheb(S16H) transduction has neuroprotective potential against neurotoxic inflammation, which is known to be a potential event related to PD pathogenesis, we examined the effects of Rheb(S16H) expression in nigral DA neurons under a neurotoxic inflammatory environment induced by the endogenous microglial activator prothrombin kringle-2 (pKr-2). Our observations showed that Rheb(S16H) transduction played a role in the neuroprotection of the nigrostriatal DA system against pKr-2-induced neurotoxic inflammation, even though there were similar levels of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-1-beta (IL-1β), in the AAV1-Rheb(S16H)-treated substantia nigra (SN) compared to the SN treated with pKr-2 alone; the neuroprotective effects may be mediated by the activation of neurotrophic signaling pathways following Rheb(S16H) transduction of nigral DA neurons. We conclude that AAV1-Rheb(S16H) transduction of neuronal populations to activate the production of neurotrophic factors and intracellular neurotrophic signaling pathways may offer promise for protecting adult neurons from extracellular neurotoxic inflammation.

5.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-198932

RESUMO

The effects of capsaicin (CAP), a transient receptor potential vanilloid subtype 1 (TRPV1) agonist, were determined on nigrostriatal dopamine (DA) neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD). The results showed that TRPV1 activation by CAP rescued nigrostriatal DA neurons, enhanced striatal DA functions and improved behavioral recovery in MPTP-treated mice. CAP neuroprotection was associated with reduced expression of proinflammatory cytokines (tumor necrosis factor-α and interleukin-1β) and reactive oxygen species/reactive nitrogen species from activated microglia-derived NADPH oxidase, inducible nitric oxide synthase or reactive astrocyte-derived myeloidperoxidase. These beneficial effects of CAP were reversed by treatment with the TRPV1 antagonists capsazepine and iodo-resiniferatoxin, indicating TRPV1 involvement. This study demonstrates that TRPV1 activation by CAP protects nigrostriatal DA neurons via inhibition of glial activation-mediated oxidative stress and neuroinflammation in the MPTP mouse model of PD. These results suggest that CAP and its analogs may be beneficial therapeutic agents for the treatment of PD and other neurodegenerative disorders that are associated with neuroinflammation and glial activation-derived oxidative damage.


Assuntos
Animais , Camundongos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Capsaicina , Citocinas , Dopamina , Neurônios Dopaminérgicos , NADPH Oxidases , Necrose , Doenças Neurodegenerativas , Neurônios , Neuroproteção , Óxido Nítrico Sintase Tipo II , Nitrogênio , Estresse Oxidativo , Oxigênio , Doença de Parkinson
6.
Experimental Neurobiology ; : 266-277, 2017.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-18846

RESUMO

Silibinin, an active constituent of silymarin extracted from milk thistle, has been previously reported to confer protection to the adult brain against neurodegeneration. However, its effects against epileptic seizures have not been examined yet. In order to investigate the effects of silibinin against epileptic seizures, we used a relevant mouse model in which seizures are manifested as status epilepticus, induced by kainic acid (KA) treatment. Silibinin was injected intraperitoneally, starting 1 day before an intrahippocampal KA injection and continued daily until analysis of each experiment. Our results indicated that silibinin-treatment could reduce seizure susceptibility and frequency of spontaneous recurrent seizures (SRS) induced by KA administration, and attenuate granule cell dispersion (GCD), a morphological alteration characteristic of the dentate gyrus (DG) in temporal lobe epilepsy (TLE). Moreover, its treatment significantly reduced the aberrant levels of apoptotic, autophagic and pro-inflammatory molecules induced by KA administration, resulting in neuroprotection in the hippocampus. Thus, these results suggest that silibinin may be a beneficial natural compound for preventing epileptic events.


Assuntos
Adulto , Animais , Humanos , Camundongos , Encéfalo , Giro Denteado , Epilepsia , Epilepsia do Lobo Temporal , Hipocampo , Ácido Caínico , Silybum marianum , Neuroproteção , Convulsões , Silimarina , Estado Epiléptico
7.
Experimental Neurobiology ; : 147-155, 2016.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-78641

RESUMO

Although accumulating evidence suggests that microglia-mediated neuroinflammation may be crucial for the initiation and progression of Parkinson's disease (PD), and that the control of neuroinflammation may be a useful strategy for preventing the degeneration of nigrostriatal dopaminergic (DA) projections in the adult brain, it is still unclear what kinds of endogenous biomolecules initiate microglial activation, consequently resulting in neurodegeneration. Recently, we reported that the increase in the levels of prothrombin kringle-2 (pKr-2), which is a domain of prothrombin that is generated by active thrombin, can lead to disruption of the nigrostriatal DA projection. This disruption is mediated by neurotoxic inflammatory events via the induction of microglial Toll-like receptor 4 (TLR4) in vivo , thereby resulting in less neurotoxicity in TLR4-deficient mice. Moreover, inhibition of microglial activation following minocycline treatment, which has anti-inflammatory activity, protects DA neurons from pKr-2-induced neurotoxicity in the substantia nigra (SN) in vivo. We also found that the levels of pKr-2 and microglial TLR4 were significantly increased in the SN of PD patients compared to those of age-matched controls. These observations suggest that there may be a correlation between pKr-2 and microglial TLR4 in the initiation and progression of PD, and that inhibition of pKr-2-induced microglial activation may be protective against the degeneration of the nigrostriatal DA system in vivo . To describe the significance of pKr-2 overexpression, which may have a role in the pathogenesis of PD, we have reviewed the mechanisms of pKr-2-induced microglial activation, which results in neurodegeneration in the SN of the adult brain.


Assuntos
Adulto , Animais , Humanos , Camundongos , Encéfalo , Microglia , Minociclina , Neurônios , Doença de Parkinson , Protrombina , Substância Negra , Trombina , Receptor 4 Toll-Like
8.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-147098

RESUMO

The cannabinoid (CB2) receptor type 2 has been proposed to prevent the degeneration of dopamine neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. However, the mechanisms underlying CB2 receptor-mediated neuroprotection in MPTP mice have not been elucidated. The mechanisms underlying CB2 receptor-mediated neuroprotection of dopamine neurons in the substantia nigra (SN) were evaluated in the MPTP mouse model of Parkinson's disease (PD) by immunohistochemical staining (tyrosine hydroxylase, macrophage Ag complex-1, glial fibrillary acidic protein, myeloperoxidase (MPO), and CD3 and CD68), real-time PCR and a fluorescein isothiocyanate-labeled albumin assay. Treatment with the selective CB2 receptor agonist JWH-133 (10 μg kg⁻¹, intraperitoneal (i.p.)) prevented MPTP-induced degeneration of dopamine neurons in the SN and of their fibers in the striatum. This JWH-133-mediated neuroprotection was associated with the suppression of blood-brain barrier (BBB) damage, astroglial MPO expression, infiltration of peripheral immune cells and production of inducible nitric oxide synthase, proinflammatory cytokines and chemokines by activated microglia. The effects of JWH-133 were mimicked by the non-selective cannabinoid receptor WIN55,212 (10 μg kg⁻¹, i.p.). The observed neuroprotection and inhibition of glial-mediated neurotoxic events were reversed upon treatment with the selective CB2 receptor antagonist AM630, confirming the involvement of the CB2 receptor. Our results suggest that targeting the cannabinoid system may be beneficial for the treatment of neurodegenerative diseases, such as PD, that are associated with glial activation, BBB disruption and peripheral immune cell infiltration.


Assuntos
Animais , Camundongos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Barreira Hematoencefálica , Quimiocinas , Citocinas , Dopamina , Neurônios Dopaminérgicos , Fluoresceína , Proteína Glial Fibrilar Ácida , Macrófagos , Microglia , Doenças Neurodegenerativas , Neuroproteção , Óxido Nítrico Sintase Tipo II , Doença de Parkinson , Peroxidase , Reação em Cadeia da Polimerase em Tempo Real , Receptor CB2 de Canabinoide , Receptores de Canabinoides , Substância Negra
9.
Laboratory Animal Research ; : 112-122, 2014.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-112262

RESUMO

UG0712 is a new ginsenoside extract processed from ginseng leaves. A subchronic toxicity study of UG0712 was conducted in male and female SD rats. Rats were treated with UG0712 at doses of 100, 400 and 1,600 mg/kg/day for 13 weeks, and observed followed by 4-week recovery period at a highest dose. No-treatment-related effects were observed regarding the mortality, ophthalmic examination, urinalysis and histopathology. Although the changes in clinical sign, body weight, organ weight, hematology, and serum biochemistry were observed, they were temporal and pharmacological effects. Based on the present experiment conditions, the no observed adverse effect level was considered to be more than 1,600 mg/kg/day in both sexes of rats.


Assuntos
Animais , Feminino , Humanos , Masculino , Ratos , Bioquímica , Peso Corporal , Hematologia , Mortalidade , Nível de Efeito Adverso não Observado , Tamanho do Órgão , Panax , Urinálise
10.
Artigo em Coreano | WPRIM (Pacífico Ocidental) | ID: wpr-89064

RESUMO

BACKGROUND: Postoperative nausea and vomiting (PONV) is one of the most common complications after general anesthesia. Sevoflurane is a newer inhalational anesthetic agent and is commonly used in general anesthesia, especially in pediatric patients and outpatients. However, the incidence of PONV after volatile induction and maintenance of anesthesia (VIMA) with sevoflurane in pediatric patients has not been reported in Korea. In this study, the incidence of PONV after VIMA with sevoflurane in pediatric patients was evaluated. METHODS: A total of 251 pediatric patients, scheduled for inguinal surgery, extremity operation and perianal surgery, were selected for the study. None of the pediatric patients was given premedicants, opioids or reversal agents during VIMA. RESULTS: The incidence of PONV after VIMA with sevoflurane in pediatric patients was 9.2%. There were no significant differences in PONV incidence according to the sex or age. PONV commonly occurred immediately after anesthesia, with the incidence of 4.9% in the operating room and 3.2% in the recovery room, respectively. CONCLUSIONS: The VIMA technique with sevoflurane for brief operations could reduce the incidence of PONV but not to zero incidence. Therefore, anesthesiologists should pay attention to the possibility of PONV, and should consider not only the anesthetic methods and agents but also the use of antiemetics, especially in high risk patients.


Assuntos
Humanos , Analgésicos Opioides , Anestesia , Anestesia Geral , Antieméticos , Extremidades , Incidência , Coreia (Geográfico) , Náusea , Salas Cirúrgicas , Pacientes Ambulatoriais , Náusea e Vômito Pós-Operatórios , Sala de Recuperação , Vômito
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...