Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
EClinicalMedicine ; 60: 102033, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37396803

ABSTRACT

Background: To date, there is no homogeneous evidence of whether earlier age at menopause is associated with incident dementia. In addition, the underlying mechanism and driven mediators are largely unknown. We aimed to fill these knowledge gaps. Methods: This community-based cohort study included 154,549 postmenopausal women without dementia at enrolment (between 2006 and 2010) from the UK Biobank who were followed up until June 2021. We followed up until June 2021. Age at menopause was entered as a categorical variable (<40, 40-49, and ≥50 years) with ≥50 years taken as a reference. The primary outcome was all-cause dementia in a time-to-event analysis and the secondary outcomes included Alzheimer's disease, vascular dementia, and other types of dementia. In addition, we investigated the association between magnetic resonance (MR) brain structure indices with earlier menopause, and explored the potential underlying driven mediators on the relationship between earlier menopause and dementia. Findings: 2266 (1.47%) dementia cases were observed over a median follow-up period of 12.3 years. After adjusting for confounders, women with earlier menopause showed a higher risk of all-cause dementia compared with those ≥50 years (adjusted-HRs [95% CIs]: 1.21 [1.09-1.34] and 1.71 [1.38-2.11] in the 40-49 years and <40 years groups, respectively; P for trend <0.001). No significant interactions between earlier menopause and polygenic risk score, cardiometabolic factors, type of menopause, or hormone-replacement therapy strata were found. Earlier menopause was negatively associated with brain MR global and regional grey matter indices, and positively associated with white matter hyperintensity. The relationship between earlier menopause and dementia was partially mediated by menopause-related comorbidities including sleep disturbance, mental health disorder, frailty, chronic pain, and metabolic syndrome, with the proportion (95% CI) of mediation effect being 3.35% (2.18-5.40), 1.38% (1.05-3.20), 5.23% (3.12-7.83), 3.64% (2.88-5.62) and 3.01% (2.29-4.40), respectively. Multiple mediator analysis showed a combined effect being 13.21% (11.11-18.20). Interpretation: Earlier age at menopause was associated with risk of incident dementia and deteriorating brain health. Further studies are warranted to clarify the underlying mechanisms by which earlier age at menopause is linked to an increased risk of dementia, and to determine public health strategies to attenuate this association. Funding: National Natural Science Foundation of China, the Science and Technology Program of Guangzhou, the Key Area Research and Development Program of Guangdong Province, the China Postdoctoral Science Foundation, and the Guangdong Basic and Applied Basic Research Foundation.

2.
Nutrients ; 15(8)2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37111191

ABSTRACT

Malnutrition is related to worsened prognosis, but the association between nutritional risk status and overall survival in radiation-induced brain necrosis (RN) has never been studied. We included consecutive patients who had received radiotherapy for head and neck cancer (HNC) and subsequently developed RN from 8 January 2005 through to 19 January 2020. The primary outcome was overall survival. We utilized three commonly-used nutritional assessments: the Geriatric Nutritional Risk Index (GNRI), Prognostic Nutritional Index (PNI), and the COntrolling NUTritional Status (CONUT) measure, to quantify the baseline nutritional risk. A total of 398 eligible patients were included. During a median follow-up of 2.3 years, 42 (10.6%) patients died of any cause. Malnutrition at admission was associated with an increased risk of future death, as assessed by the GNRI (per 1-point decreased, HR 1.05, 95%CI 1.02-1.09, p = 0.001), the PNI (per 1-point decreased, HR 1.07, 95%CI 1.03-1.12, p = 0.002), and the CONUT (per 1-point increased, HR 1.22, 95%CI 1.08-1.37, p = 0.001). There were no nonlinear correlations between all three indices and post-RN survival. Among HNC survivors with RN, the assessment of nutritional risk by composite indices upon admission could help identify patients who might be at high risk of future death and deliver better nutritional management.


Subject(s)
Head and Neck Neoplasms , Malnutrition , Humans , Aged , Nutrition Assessment , Prognosis , Risk Factors , Retrospective Studies , Nutritional Status , Malnutrition/etiology , Malnutrition/complications , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/complications , Brain , Necrosis/complications
3.
Neurosci Lett ; 802: 137170, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36898650

ABSTRACT

BACKGROUND AND PURPOSE: Previous studies have explored the clinical consequences of cortical microinfarction, mainly age-related cognitive decline. However, functional impairment of deep cortical microinfarction remains poorly understood. Based on anatomical knowledge and previous research, we infer that damage to the deep cortex may lead to cognitive deficits and communication impairment between the superficial cortex and thalamus. This study aimed to develop a new model of deep cortical microinfarction based on femtosecond laser ablation of a perforating artery. METHODS: Twenty-eight mice were anesthetized with isoflurane, and a cranial window was thinned using a microdrill. Intensively focused femtosecond laser pulses were used to produce perforating arteriolar occlusions and ischemic brain damage was examined using histological analysis. RESULTS: Occlusion of different perforating arteries induced different types of cortical microinfarctions. Blocking the perforating artery, which enters the cerebral cortex vertically and has no branches within 300 µm below, can result in deep cortical microinfarction. Moreover, this model showed neuronal loss and microglial activation in the lesions as well as dysplasia of nerve fibers and ß-amyloid deposition in the corresponding superficial cortex. CONCLUSIONS: We present here a new model of deep cortical microinfarction in mice, in which specific perforating arteries are selectively occluded by a femtosecond laser, and we preliminarily observe several long-term effects related to cognition. This animal model is helpful in investigating the pathophysiology of deep cerebral microinfarction. However, further clinical and experimental studies are required to explore deep cortical microinfarctions in greater molecular and physiological detail.


Subject(s)
Cerebral Cortex , Lasers , Mice , Animals , Cerebral Cortex/pathology , Skull , Arterioles
4.
Sci Transl Med ; 15(684): eabm6543, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36812346

ABSTRACT

Radiation-induced brain injury (RIBI) is a debilitating sequela after radiotherapy to treat head and neck cancer, and 20 to 30% of patients with RIBI fail to respond to or have contraindications to the first-line treatments of bevacizumab and corticosteroids. Here, we reported a Simon's minmax two-stage, single-arm, phase 2 clinical trial (NCT03208413) to assess the efficacy of thalidomide in patients with RIBI who were unresponsive to or had contraindications to bevacizumab and corticosteroid therapies. The trial met its primary endpoint, with 27 of 58 patients enrolled showing ≥25% reduction in the volume of cerebral edema on fluid-attenuated inversion recovery-magnetic resonance imaging (FLAIR-MRI) after treatment (overall response rate, 46.6%; 95% CI, 33.3 to 60.1%). Twenty-five (43.1%) patients demonstrated a clinical improvement based on the Late Effects Normal Tissues-Subjective, Objective, Management, Analytic (LENT/SOMA) scale, and 36 (62.1%) experienced cognitive improvement based on the Montreal Cognitive Assessment (MoCA) scores. In a mouse model of RIBI, thalidomide restored the blood-brain barrier and cerebral perfusion, which were attributed to the functional rescue of pericytes secondary to elevation of platelet-derived growth factor receptor ß (PDGFRß) expression by thalidomide. Our data thus demonstrate the therapeutic potential of thalidomide for the treatment of radiation-induced cerebral vasculature impairment.


Subject(s)
Brain Injuries , Radiation Injuries , Animals , Mice , Thalidomide , Blood-Brain Barrier/pathology , Bevacizumab/therapeutic use , Brain/pathology , Radiation Injuries/pathology , Brain Injuries/drug therapy , Brain Injuries/pathology
5.
Neuron ; 111(5): 696-710.e9, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36603584

ABSTRACT

The crosstalk between the nervous and immune systems has gained increasing attention for its emerging role in neurological diseases. Radiation-induced brain injury (RIBI) remains the most common medical complication of cranial radiotherapy, and its pathological mechanisms have yet to be elucidated. Here, using single-cell RNA and T cell receptor sequencing, we found infiltration and clonal expansion of CD8+ T lymphocytes in the lesioned brain tissues of RIBI patients. Furthermore, by strategies of genetic or pharmacologic interruption, we identified a chemotactic action of microglia-derived CCL2/CCL8 chemokines in mediating the infiltration of CCR2+/CCR5+ CD8+ T cells and tissue damage in RIBI mice. Such a chemotactic axis also participated in the progression of cerebral infarction in the mouse model of ischemic injury. Our findings therefore highlight the critical role of microglia in mediating the dysregulation of adaptive immune responses and reveal a potential therapeutic strategy for non-infectious brain diseases.


Subject(s)
Brain Injuries , Microglia , Animals , Mice , Microglia/physiology , CD8-Positive T-Lymphocytes/metabolism , Brain Injuries/pathology , Brain/metabolism , Chemokine CCL2/metabolism , Mice, Inbred C57BL
6.
BMC Med ; 20(1): 484, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36522755

ABSTRACT

BACKGROUND: Some observational studies had found that shift work would increase risks of metabolic disorders, cancers, and cardiovascular diseases, but there was no homogeneous evidence of such an association between shift work and incident dementia. This study aimed to investigate whether shift work would increase the risk of dementia in a general population. METHODS: One hundred seventy thousand seven hundred twenty-two employed participants without cognitive impairment or dementia at baseline recruited between 2006 and 2010 were selected from the UK Biobank cohort study. Follow-up occurred through June 2021. Shift work status at baseline was self-reported by participants and they were categorized as non-shift workers or shift workers. Among shift workers, participants were further categorized as night shift workers or shift but non-night shift workers. The primary outcome was all-cause dementia in a time-to-event analysis, and the secondary outcomes were subtypes of dementia, including Alzheimer's disease, vascular dementia, and other types of dementia. RESULTS: In total, 716 dementia cases were observed among 170,722 participants over a median follow-up period of 12.4 years. Shift workers had an increased risk of all-cause dementia as compared with non-shift workers after multivariable adjustment (hazard ratio [HR], 1.30, 95% confidence interval [CI], 1.08-1.58); however, among shift workers, night shift work was not associated with the risk of dementia (HR, 1.04, 95% CI, 0.73-1.47). We found no significant interaction between shift work and genetic predisposition to dementia on the primary outcome (P for interaction = 0.77). CONCLUSIONS: Shift work at baseline was associated with an increased risk of all-cause dementia. Among shift workers, there was no significant association between night shift work and the risk of dementia. The increased incidence of dementia in shift workers did not differ between participants in different genetic risk strata for dementia.


Subject(s)
Alzheimer Disease , Shift Work Schedule , Humans , Cohort Studies , Shift Work Schedule/adverse effects , Risk Factors , Alzheimer Disease/epidemiology , Incidence
7.
Stroke ; 53(12): 3751-3762, 2022 12.
Article in English | MEDLINE | ID: mdl-36305312

ABSTRACT

BACKGROUND: Emerging evidence highlighted vascular injury in aggravating radiation-induced brain injury (RIBI), a common complication of radiotherapy. This study aimed to delineate the pathological feature of cerebral small vessel and investigate the functional roles of Notch signaling in RIBI. METHODS: Brain tissue and functional MRI from RIBI patients were collected and analyzed for radiation-induced vasculopathy. A RIBI mouse model was induced by a single dose of 30-Gy cranial irradiation. Vascular morphology, pulsatility, and reactivity to pharmacological interventions, such as nimodipine and 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid, were monitored by 2-photon imaging in mice at 6 weeks postirradiation. Western blot, real-time quantitative PCR, immunofluorescence staining, and behavioral tests were performed. The effect of N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester, a Notch inhibitor, was used to investigate the vascular pathogenesis of RIBI mouse model. RESULTS: Morphologically, radiation resulted in vascular malformation featured by focal contractile rings together with general stenosis. Functionally, radiation also led to hypoperfusion, attenuated vascular pulsatility, and decreased dilation to nimodipine and 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid. Mechanically, Notch activation and increased expression of α-SMA protein were found in both surgical specimens of RIBI patients and the irradiated mice. Importantly, Notch inhibition by N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester significantly alleviated cerebral hypoperfusion, vasculopathy, and cognitive deficits in the RIBI mouse model. CONCLUSIONS: Radiation-induced cerebral vasculopathy showed bead-like shape and increased contractile state. Inhibition of Notch signaling by N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester effectively attenuated vasculopathy and relieved cognitive impairment, suggesting Notch signaling as a therapeutic target for the treatment of RIBI.


Subject(s)
Brain Injuries , Cerebrovascular Disorders , Radiation Injuries , Animals , Mice , Nimodipine , Myocytes, Smooth Muscle/pathology , Signal Transduction , Cerebrovascular Disorders/complications , Brain Injuries/pathology , Esters/metabolism , Esters/pharmacology , Receptors, Notch/metabolism
8.
J Neuroinflammation ; 19(1): 231, 2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36131309

ABSTRACT

BACKGROUND: Radiation-induced brain injury (RIBI) is the most serious complication of radiotherapy in patients with head and neck tumors, which seriously affects the quality of life. Currently, there is no effective treatment for patients with RIBI, and identifying new treatment that targets the pathological mechanisms of RIBI is urgently needed. METHODS: Immunofluorescence staining, western blotting, quantitative real-time polymerase chain reaction (Q-PCR), co-culture of primary neurons and microglia, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay, enzyme-linked immunosorbent assay (ELISA), and CRISPR-Cas9-mediated gene editing techniques were employed to investigate the protective effects and underlying mechanisms of pregabalin that ameliorate microglial activation and neuronal injury in the RIBI mouse model. RESULTS: Our findings showed that pregabalin effectively repressed microglial activation, thereby reducing neuronal damage in the RIBI mouse model. Pregabalin mitigated inflammatory responses by directly inhibiting cytoplasmic translocation of high-mobility group box 1 (HMGB1), a pivotal protein released by irradiated neurons which induced subsequent activation of microglia and inflammatory cytokine expression. Knocking out neuronal HMGB1 or microglial TLR2/TLR4/RAGE by CRISPR/Cas9 technique significantly inhibited radiation-induced NF-κB activation and pro-inflammatory transition of microglia. CONCLUSIONS: Our findings indicate the protective mechanism of pregabalin in mitigating microglial activation and neuronal injury in RIBI. It also provides a therapeutic strategy by targeting HMGB1-TLR2/TLR4/RAGE signaling pathway in the microglia for the treatment of RIBI.


Subject(s)
Brain Injuries , HMGB1 Protein , Animals , Brain Injuries/metabolism , Cytokines/metabolism , DNA Nucleotidylexotransferase/metabolism , DNA Nucleotidylexotransferase/pharmacology , HMGB1 Protein/metabolism , Mice , Microglia/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Pregabalin/metabolism , Pregabalin/pharmacology , Pregabalin/therapeutic use , Quality of Life , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
9.
FASEB J ; 34(10): 13361-13375, 2020 10.
Article in English | MEDLINE | ID: mdl-32851721

ABSTRACT

Radiotherapy is one of the most effective treatments for head and neck tumors. However, delayed radiation-induced brain necrosis (RN) remains a serious issue due to the lack of satisfying prevention and effective treatment. The pathological role of radiation in the delayed onset of brain necrosis is still largely unknown, and the traditional animal model of whole brain irradiation, although being widely used, does not produce reliable and localized brain necrosis mimicking clinical features of RN. In this study, we demonstrated a successful RN mouse model using optimized gamma knife irradiation in male C57BL/6 mice. On the premise that brain necrosis started to appear at 6 weeks postirradiation in our RN model, as confirmed by both MRI and histopathological examinations, we systematically examined different time points before the onset of RN for the histopathological changes and biochemical indicators. Our initial results demonstrated that in the ipsilateral hemisphere of the irradiated brains, a significant decrease in neuronal numbers that occurred at 4 weeks and a sustained increase in TNF-α, iNOS, and other inflammatory cytokines beginning at 1-week postirradiation. Changes of cell morphology and cell numbers of both microglia and astrocytes occurred as early as 1-week postirradiation, and intervention by bevacizumab administration resulted in reduced microglia activation and reduction of radiation-induced lesion volume, indicating that chronic glial activation may result in subsequent elevation of inflammatory factors, which led to the delayed onset of neuronal loss and brain necrosis. Since C57BL/6 is the most widely used strain of genetic engineered mouse model, our data provide an invaluable platform for the mechanistic study of RN pathogenesis, identification of potential imaging and biological biomarkers, and the development of therapeutic treatment for the disease.


Subject(s)
Astrocytes , Bevacizumab , Brain , Gamma Rays/adverse effects , Microglia , Radiation Injuries, Experimental , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Bevacizumab/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/pathology , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Necrosis , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology
10.
Addict Biol ; 24(4): 577-589, 2019 07.
Article in English | MEDLINE | ID: mdl-29569345

ABSTRACT

Alcohol addiction is a chronic neuropsychiatric disorder that represents one of the most serious global public health problems. Yet, currently there still lacks an effective pharmacotherapy. Omega-3 polyunsaturated fatty acids (N-3 PUFAs) have exhibited beneficial effects in a variety of neurological disorders, particularly in reversing behavioral deficits and neurotoxicity induced by prenatal alcohol exposure and binge drinking. In the present study, we investigated if fish oil, which is rich in N-3 PUFAs, had beneficial effects on preventing relapse and alleviating withdrawal symptoms after chronic alcohol exposure. Our results demonstrated that fish oil significantly reduced the chronic alcohol exposure-induced aberrant dendritic morphologic changes of the medium-sized spiny neurons in the core and the shell of nucleus accumbens. This inhibited the expression of AMPAR2-lacking AMPARs and their accumulation on the post synaptic membranes of medium-sized spiny neurons and eventually alleviated withdrawal symptoms and alcohol dependence. Our study therefore suggests that N-3 PUFAs are promising for treating withdrawal symptoms and alcohol dependence.


Subject(s)
Alcoholism/pathology , Central Nervous System Depressants/pharmacology , Dendrites/drug effects , Ethanol/pharmacology , Fish Oils/pharmacology , Nucleus Accumbens/drug effects , Synapses/drug effects , Alcohol Withdrawal Seizures , Animals , Dendrites/pathology , Locomotion/drug effects , Mice , Nucleus Accumbens/cytology , Nucleus Accumbens/pathology , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Recurrence , Synapses/pathology
11.
Mol Neurobiol ; 55(2): 1703-1713, 2018 02.
Article in English | MEDLINE | ID: mdl-28211008

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive neuronal degeneration and pathological accumulation of amyloid plaques in the brain. It has been proposed that the prion-like spreading of amyloid beta (Aß) protein could contribute to the progression of the disease. Olfactory bulb (OB) is one of the earliest brain regions affected in AD and olfaction is easily impaired prior to cognitive symptoms. However, it remains unclear whether Aß accumulation in the OB would spread along olfactory projections to other connected brain regions and trigger further neurodegeneration. In the present study, we experimentally injected recombinant human Aß1-42 (monomers and oligomers, respectively) into the mouse OB and tracked the spreading of Aß to connected brain regions over 3 days. The results showed that both Aß monomers and oligomers were rapidly and readily transferred from the injection site to interconnected brain regions in a neural connection manner and triggered neuronal apoptosis in the affected brain regions. Oligomeric Aß1-42 spread more efficiently and induced more neuronal apoptosis in the affected brain regions compared to monomeric Aß1-42. Therefore, the study provides evidence that Aß peptides can transfer via neural connections and the pattern of Aß peptide spreading provides understanding to manage AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Brain/drug effects , Nerve Degeneration/pathology , Olfactory Bulb/drug effects , Alzheimer Disease/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/metabolism , Neural Pathways/drug effects , Neural Pathways/metabolism , Neural Pathways/pathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology
12.
Mol Med Rep ; 16(2): 1739-1746, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627708

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by selective degeneration of upper and lower motor neurons. The disease progression is associated with the astrocytic environment. Aquaporin-4 (AQP4) water channels are the most abundant AQPs expressed in astrocytes, exerting important influences on central nervous system homeostasis. The present study aimed to characterize the alterations in AQP4 expression and loca-lization in superoxide dismutase 1 (SOD1) G93A transgenic mice. SOD1G93A mice were sacrificed during the presymptomatic, disease onset and end stages and immunostaining was performed on spinal cord sections to investigate neuronal loss, glial activation and AQP4 expression in the spinal cord. It was observed that global AQP4 expression increased in the spinal cord of SOD1G93A mice as the disease progressed. However, AQP4 polarization decreased as the disease progressed, and AQP4 polarized localization at the endfeet of astrocytes was decreased in the spinal ventral horn of SOD1G93A mice at the disease onset and end stages. Meanwhile, motor neuron dege-neration and decreased glutamate transporter 1 expression in astrocytes in SOD1G93A mice were observed as the disease progressed. The results of the present study demonstrated that AQP4 depolarization is a widespread pathological condition and may contribute to motor neuron degeneration in ALS.


Subject(s)
Aquaporin 4/biosynthesis , Nerve Degeneration/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Anterior Horn Cells/metabolism , Anterior Horn Cells/pathology , Aquaporin 4/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Disease Progression , Glial Fibrillary Acidic Protein/metabolism , Homeostasis , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration/pathology , Spinal Cord/metabolism
13.
Mol Neurobiol ; 54(9): 7327-7334, 2017 11.
Article in English | MEDLINE | ID: mdl-27815837

ABSTRACT

Depression is associated with somatic immune changes, and neuroinflammation is now recognized as hallmark for depressive disorders. N-3 (or omega-3) polyunsaturated fatty acids (PUFAs) are well known to suppress neuroinflammation, reduce oxidative stress, and protect neuron from injury. We pretreated animals with fish oil and induced acute depression-like behaviors with systemic lipopolysaccharide (LPS) injection. The levels of cytokines and stress hormones were determined from plasma and different brain areas. The results showed that fish oil treatment prevent LPS-induce depressive behavior by suppression of neuroinflammation. LPS induced acute neuroinflammation in different brain regions, which were prevented in fish oil fed mice. However, neither LPS administration nor fish oil treatment has strong effect on stress hormone secretion in the hypothalamus and adrenal. Fish oil might provide a useful therapy against inflammation-associated depression.


Subject(s)
Depression/chemically induced , Depression/prevention & control , Fish Oils/administration & dosage , Lipopolysaccharides/toxicity , Oxidative Stress/drug effects , Animals , Depression/pathology , Inflammation/chemically induced , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology
14.
BMC Complement Altern Med ; 15: 137, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25925762

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting 2% of the population aged over 65 years old. Mitochondrial defects and oxidative stress actively participate in degeneration of dopaminergic (DA) neurons in PD. Paeonolum, a main component isolated from Moutan cortex, has potent antioxidant ability. Here, we have examined the effects of paeonolum against MPP(+)-induced neurotoxicity in zebrafish and PC12 cells. METHODS: The overall viability and neurodegeneration of DA neurons was assessed in ETvmat2:green fluorescent protein (GFP) transgenic zebrafish, in which most monoaminergic neurons are labeled by GFP. Damage to PC12 cells was measured using a cell viability assay and assessment of nuclear morphology. Intracellular reactive oxygen species (ROS) and the level of total GSH were assessed. The mitochondrial cell death pathway including mitochondrial membrane potential, cytochrome C release and caspase-3 activity were also examined in PC12 cells. RESULTS: Paeonolum protected against MPP(+)-induced DA neurodegeneration and locomotor dysfunction in zebrafish in a concentration-dependent manner. Similar neuroprotection was replicated in the PC12 cellular model of MPP(+) toxicity. Paeonolum attenuated MPP(+)-induced intracellular ROS accumulation and restored the level of total GSH in PC12 cells. Furthermore, paeonolum significantly inhibited the mitochondrial cell death pathway induced by MPP(+). CONCLUSIONS: Collectively, the present study demonstrates that paeonolum protects zebrafish and PC12 cells against MPP(+)-induced neurotoxicity.


Subject(s)
Antioxidants/therapeutic use , Dopaminergic Neurons/drug effects , Neuroprotective Agents/therapeutic use , Paeonia/chemistry , Parkinson Disease/prevention & control , Phytotherapy , Plant Extracts/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Caspase 3/metabolism , Cell Death/drug effects , Cytochromes c/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Humans , MPTP Poisoning/metabolism , MPTP Poisoning/prevention & control , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/adverse effects , Oxidative Stress/drug effects , PC12 Cells , Parkinson Disease/metabolism , Plant Extracts/pharmacology , Rats , Reactive Oxygen Species/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...