Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Neurol Sci ; 45(5): 2027-2033, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38060035

RESUMO

BACKGROUND: Mid- to late-stage Parkinson's disease (PD) is often linked with worsened and significant impairment of motor activities, but existing prognostic markers do not adequately capture the risk of loss of balance in PD patients. This study aims to develop a risk prognostic model for mid- to late-stage PD and identify prognostic factors that are indicative of impending loss of balance and falls. METHODS: The study included 307 participants of which 75 were diagnosed with idiopathic PD and 232 were neurological or non-neurological controls. Among the PD group, 46 were early-stage (Hoehn and Yahr [H&Y] = 1,2) with no significant loss of balance while 29 were mid- to late-stage (H&Y = 3,4,5) which is characterized by loss of balance and falls. Multivariable logistic regression (MLR) was used to develop a prognostic model for mid- to late-stage PD. Model discrimination was assessed by ROC curves. The model was internally validated through bootstrapping and calibration plots. RESULTS: The relevant factors identified and included in the final MLR model were shortness of breath, age, swollen joints, heme oxygenase-1 (HO-1) protein, and total salivary protein. The model had an AUC of 0.82 (95% CI = 0.71-0.92) and was well calibrated (calibration slope = 0.77, intercept = 0.03). The likelihood of shortness of breath (OR = 7.91, 95% CI = 1.63-45.12) was significantly higher among mid- to late-stage PD than early-stage. Age and total salivary protein were also significantly higher among mid- to late-stage PD. CONCLUSION: The MLR prognostic model for mid- to late-stage PD may assist physicians in identifying patients at high risk for loss of balance and falls.


Assuntos
Doença de Parkinson , Humanos , Doença de Parkinson/complicações , Doença de Parkinson/diagnóstico , Prognóstico , Equilíbrio Postural/fisiologia , Dispneia , Proteínas e Peptídeos Salivares
2.
J Neurochem ; 157(6): 2195-2209, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32880973

RESUMO

Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide, and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. HO-1 does not contain an N-terminal signal peptide and the mechanism responsible for its secretion remains unknown. Extracellular vesicles (EVs) are membrane-bound inclusions that transport microRNAs, messenger RNAs, lipids, and proteins among diverse cellular and extracellular compartments. The objective of the current study was to determine whether EVs in human biofluids contain HO-1, and whether the latter may be transported in EVs from brain to periphery. Total, L1 cell adhesion molecule protein (L1CAM)-enriched (neuron-derived), and glutamate aspartate transporter 1 (GLAST)-enriched (astrocyte-derived) EVs were purified from five different human biofluids (saliva [n = 40], plasma [n = 14], serum [n = 10], urine [n = 10], and cerebrospinal fluid [n = 11]) using polymer precipitation and immuno-affinity-based capture methods. L1CAM-enriched, GLAST-enriched, and L1CAM/GLAST-depleted (LGD) EV, along with EV-depleted (EVD), fractions were validated by nanoparticle tracking analysis, enzyme-linked immunosorbent assay (ELISA), and western blot. HO-1 was assayed in all fractions using ELISA and western blot. The majority of HO-1 protein was localized to LGD, L1CAM-enriched, and GLAST-enriched EVs of all human biofluids surveyed after adjusting for age and sex, with little HO-1 protein detected in EVD fractions. HO-1 protein in human biofluids is predominantly localized to EV compartments. A substantial proportion of EV HO-1 in peripheral human biofluids is derived from the central nervous system and may contribute to the systemic manifestations of various neurological conditions.


Assuntos
Líquidos Corporais/enzimologia , Vesículas Extracelulares/enzimologia , Heme Oxigenase-1/metabolismo , Adulto , Idoso , Biomarcadores/metabolismo , Líquidos Corporais/química , Vesículas Extracelulares/química , Feminino , Heme Oxigenase-1/análise , Humanos , Masculino , Pessoa de Meia-Idade
3.
Mov Disord ; 35(3): 468-477, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31800144

RESUMO

BACKGROUND: Parkinson's disease (PD) is the most common movement disorder among adults, affecting 2% of the world population older than 65 years of age. No diagnostic biomarker for routine use in clinical settings currently exists. Dysregulation of microRNAs (miRNAs) has been implicated in various neurodegenerative conditions, including PD. Distinct miRNAs have been demonstrated to be involved in the regulation of α-synuclein, a key player in PD pathogenesis; miR-153 and miR-223 are downregulated in the brain and serum of parkinsonian GFAP.HMOX1 transgenic mice where they directly regulate α-synuclein. OBJECTIVE: To ascertain whether salivary miR-153 and miR-223 are similarly downmodulated in, and may serve as diagnostic biomarkers of, idiopathic PD. METHODS: Using reverse transcriptase quantitative polymerase chain reaction, miR-153 and miR-223 levels were evaluated in the saliva of 77 non-neurological controls and 83 PD patients. Levels of heme oxygenase-1 and α-synuclein were measured using enzyme-linked immunosorbent assay. Analyses were adjusted by age, sex, medication exposure, disease duration, and relevant comorbidities. RESULTS: Log-transformed expression levels of miR-153 and miR-223 were significantly decreased in the saliva of human PD patients in comparison with nonneurological controls. The miRNA expression levels did not change as a function of disease progression (Hoehn and Yahr staging). The area under the receiver operating characteristic curve separating controls from PD patients was 79% (95% confidence interval, 61%-96%) for miR-153 and 77% (95% confidence interval, 59%-95%) for miR-223. The ratios of miRNAs to oligomeric α-synuclein, total α-synuclein, or heme oxygenase-1 protein did not improve accuracy of the test. CONCLUSION: Salivary miR-153 and miR-223 levels may serve as useful, noninvasive, and relatively inexpensive diagnostic biomarkers of idiopathic PD. © 2019 International Parkinson and Movement Disorder Society.


Assuntos
MicroRNAs , Doença de Parkinson , Adulto , Biomarcadores , Humanos , MicroRNAs/genética , Doença de Parkinson/diagnóstico , Doença de Parkinson/genética , Curva ROC , alfa-Sinucleína
4.
Glia ; 67(9): 1730-1744, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31180611

RESUMO

α-Synuclein is a key player in the pathogenesis of Parkinson disease (PD). Expression of human heme oxygenase-1 (HO-1) in astrocytes of GFAP.HMOX1 transgenic (TG) mice between 8.5 and 19 months of age results in a parkinsonian phenotype characterized by neural oxidative stress, nigrostriatal hypodopaminergia associated with locomotor incoordination, and overproduction of α-synuclein. We identified two microRNAs (miR-), miR-153 and miR-223, that negatively regulate α-synuclein in the basal ganglia of male and female GFAP.HMOX1 mice. Serum concentrations of both miRNAs progressively declined in the wild-type (WT) and GFAP.HMOX1 mice between 11 and 19 months of age. Moreover, at each time point surveyed, circulating levels of miR-153 were significantly lower in the TG animals compared to WT controls, while α-synuclein protein concentrations were elevated in erythrocytes of the GFAP.HMOX1 mice at 19 months of age relative to WT values. Primary WT neurons co-cultured with GFAP.HMOX1 astrocytes exhibited enhanced protein oxidation, mitophagy and apoptosis, aberrant expression of genes regulating the dopaminergic phenotype, and an imbalance in gene expression profiles governing mitochondrial fission and fusion. Many, but not all, of these neuronal abnormalities were abrogated by small interfering RNA (siRNA) knockdown of α-synuclein, implicating α-synuclein as a potent, albeit partial, mediator of HO-1's neurodystrophic effects in these parkinsonian mice. Overexpression of HO-1 in stressed astroglia has previously been documented in the substantia nigra of idiopathic PD and may promote α-synuclein production and toxicity by downmodulating miR-153 and/or miR-223 both within the CNS and in peripheral tissues.


Assuntos
Heme Oxigenase-1/metabolismo , Neuroglia/metabolismo , Transtornos Parkinsonianos/metabolismo , alfa-Sinucleína/metabolismo , Animais , Apoptose/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Técnicas de Cocultura , Eritrócitos/metabolismo , Feminino , Expressão Gênica , Heme Oxigenase-1/genética , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , MicroRNAs/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/fisiologia
5.
Mov Disord ; 33(4): 583-591, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29488275

RESUMO

BACKGROUND AND HYPOTHESIS: To date, there are no chemical analytes, including biochemical indices of oxidative stress, metabolites of α-synuclein protein, and differential protein expression patterns on proteomic profiling, for use in clinics as a diagnostic biomarker of idiopathic PD. OBJECTIVES: Heme oxygenase-1 has been implicated in the pathogenesis of PD. The objective of this study is to ascertain whether salivary heme oxygenase-1 may serve as a biomarker for early idiopathic PD. METHODS: Fifty-eight PD patients and 59 non-neurological disease controls were recruited. Levels of heme oxygenase-1 expression were assayed using enzyme-linked immunosorbent assay and western blot analysis of whole, unstimulated saliva. Analyses were adjusted by sex, l-dopa exposure, and relevant comorbidities. RESULTS: We documented: (1) the presence of 32-kDa heme oxygenase-1 protein in human saliva; (2) significantly higher mean heme oxygenase-1 protein concentrations in saliva of PD patients relative to control values; (3) no variability in salivary heme oxygenase-1 levels with sex, age, l-dopa equivalence, or comorbidities; and (4) significantly higher mean salivary heme oxygenase-1 concentrations in patients with H & Y stage 1 PD (early) than control subjects and stage 2 and stage 3 PD patients. The area under the receiver operating characteristic curve that separated controls from PD H & Y stage 1 was 76% (95% confidence interval: 63-90). CONCLUSIONS: Salivary heme oxygenase-1 concentrations may provide a useful, noninvasive, and relatively inexpensive biomarker of early idiopathic PD. © 2018 International Parkinson and Movement Disorder Society.


Assuntos
Biomarcadores/metabolismo , Heme Oxigenase-1/metabolismo , Doença de Parkinson/enzimologia , Saliva/enzimologia , Idoso , Antiparkinsonianos/uso terapêutico , Feminino , Humanos , Levodopa/uso terapêutico , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/tratamento farmacológico , Curva ROC , Estudos Retrospectivos , Saliva/efeitos dos fármacos , Fatores Sexuais
6.
Neurobiol Dis ; 102: 140-146, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28323022

RESUMO

Pharmacotherapies that increase CNS expression of heme oxygenase-1 (HO-1) and other antioxidant proteins have improved outcome in experimental models of spontaneous intracerebral hemorrhage (ICH). In order to more specifically investigate the relationship between HO-1 and ICH outcome, mice expressing human HO-1 driven by the glial fibrillary acidic protein (GFAP) promoter (GFAP·HMOX1 mice) were tested in a model of in situ parenchymal hemorrhage. Injection of collagenase into the striata of wild-type (WT) mice resulted in a 26.3% mortality rate, with deaths equally distributed between males and females. Mortality was reduced to 4.48% in GFAP·HMOX1 mice. Cell viability in the injected striata of surviving WT mice was reduced by about half at one week and was significantly increased in transgenics; this benefit persisted over a 22day observation period. Cell counts guided by design-based stereology indicated loss of ~40% of neurons in WT hemorrhagic striata at one week, which was decreased by half in transgenics; no significant differences in microglia or astrocyte numbers were observed. Blood-brain barrier disruption and short-term neurological deficits were also mitigated in GFAP·HMOX1 mice, but long-term outcome did not differ from that of WT survivors. These results suggest that astrocyte HO-1 overexpression provides robust neuroprotection after acute intracerebral hemorrhage. Further investigation of drug or genetic therapies that selectively increase astrocyte HO-1 is warranted.


Assuntos
Astrócitos/enzimologia , Hemorragia Cerebral/enzimologia , Heme Oxigenase-1/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade Capilar/fisiologia , Sobrevivência Celular/fisiologia , Hemorragia Cerebral/mortalidade , Hemorragia Cerebral/patologia , Hemorragia Cerebral/psicologia , Colagenases , Corpo Estriado/enzimologia , Corpo Estriado/patologia , Modelos Animais de Doenças , Feminino , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Heme Oxigenase-1/genética , Humanos , Masculino , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Neuroproteção/fisiologia
7.
Hum Brain Mapp ; 38(1): 326-338, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27614005

RESUMO

The peri-infarct cortex (PIC) is the site of long-term physiologic changes after ischemic stroke. Traditional methods for delineating the peri-infarct gray matter (GM) have used a volumetric Euclidean distance metric to define its extent around the infarct. This metric has limitations in the case of cortical stroke, i.e., those where ischemia leads to infarction in the cortical GM, because the vascularization of the cerebral cortex follows the complex, folded topology of the cortical surface. Instead, we used a geodesic distance metric along the cortical surface to subdivide the PIC into equidistant rings emanating from the infarct border and compared this new approach to a Euclidean distance metric definition. This was done in 11 patients with [F-18]-Flumazenil ([18-F]-FMZ) positron emission tomography (PET) scans at 2 weeks post-stroke and at 6 month follow-up. FMZ is a PET radiotracer with specific binding to the alpha subunits of the type A γ-aminobutyric acid (GABAA) receptor. Additionally, we used partial-volume correction (PVC) of the PET images to compensate for potential cortical thinning and long-term neuronal loss in follow-up images. The difference in non-displaceable binding potential (BPND ) between the stroke unaffected and affected hemispheres was 35% larger in the geodesic versus the Euclidean peri-infarct models in initial PET images and 48% larger in follow-up PET images. The inter-hemispheric BPND difference was approximately 17-20% larger after PVC when compared to uncorrected PET images. PET studies of peri-infarct GM in cortical strokes should use a geodesic model and include PVC as a preprocessing step. Hum Brain Mapp 38:326-338, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Córtex Cerebral/diagnóstico por imagem , Doenças do Sistema Nervoso/patologia , Neurônios/patologia , Tomografia por Emissão de Pósitrons , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Infarto Encefálico/diagnóstico por imagem , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Doenças do Sistema Nervoso/etiologia , Estudos Retrospectivos , Acidente Vascular Cerebral/complicações
8.
Neuroendocrinology ; 103(5): 417-31, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26335277

RESUMO

The effects of gonadal steroids on neurological well-being and disease constitute a rich and rapidly expanding area of basic and clinical neuroscience. Gonadal hormones exert potent effects on monoaminergic, cholinergic and peptidergic pathways as well as neurosteroidogenesis which, in turn, impact normal brain organization and function. A spectrum of human neurological conditions are influenced by hormonal fluctuations associated with the menstrual cycle, pregnancy, the menopause and use of oral contraceptives. An appreciation of these relationships may facilitate the development of specific hormonal and anti-hormonal therapies for neurological disorders as disparate as catamenial epilepsy and acute intermittent porphyria.


Assuntos
Hormônios Gonadais/metabolismo , Hormônios Gonadais/uso terapêutico , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/terapia , Animais , Feminino , Humanos , Masculino , Doenças do Sistema Nervoso/epidemiologia , Gravidez
9.
Glia ; 63(7): 1270-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25820186

RESUMO

BACKGROUND: Over-expression of the heme-degrading enzyme, heme oxygenase-1 (HO-1) promotes iron deposition, mitochondrial damage, and autophagy in astrocytes and enhances the vulnerability of nearby neuronal constituents to oxidative injury. These neuropathological features and aberrant brain microRNA (miRNA) expression patterns have been implicated in the etiopathogeneses of various neurodevelopmental and aging-related neurodegenerative disorders. OBJECTIVE: To correlate glial HO-1 overexpression with altered miRNA patterns, which have been linked to the aforementioned "core" neuropathological features. METHODS: miRNA microchip assays were performed on HMOX1- and sham-transfected primary rat astroglia and affected miRNAs were further validated by qPCR. The roles of the heme degradation products, carbon monoxide (CO), iron (Fe) and bilirubin on miRNA expression were assessed and salient mRNA targets of the impacted miRNAs were ascertained. RESULTS: In HMOX1-transfected astrocytes, rno-miR-140*, rno-miR-17, and rno-miR-16 were significantly up-regulated, and rno-miR-297, rno-miR-206, rno-miR-187, rno-miR-181a, rno-miR-138 and rno-miR-29c were down-regulated, compared to sham-transfected controls. CO and Fe were implicated in the HMOX1 effects, whereas bilirubin was inert or counteracted the HMOX1-related changes. mRNA levels of Ngfr, Vglut1, Mapk3, Tnf-α, and Sirt1, known targets of the down-regulated miRNAs and abnormal in various human brain disorders, were significantly increased in the HMOX-1-transfected astrocytes. CONCLUSIONS: In chronic CNS disorders, altered expression of salient miRNAs and their mRNA targets may contribute to the neural damage accruing from the over-expression of glial HO-1.


Assuntos
Astrócitos/metabolismo , Heme Oxigenase-1/metabolismo , MicroRNAs/metabolismo , Animais , Bilirrubina/metabolismo , Encéfalo/metabolismo , Encefalopatias/metabolismo , Monóxido de Carbono/metabolismo , Heme Oxigenase-1/genética , Humanos , Ferro/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Transfecção
10.
Stroke ; 46(4): 1093-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25690543

RESUMO

BACKGROUND AND PURPOSE: Heme oxygenase-1 (HO-1) catalyzes the rate-limiting reaction of heme breakdown and may have both antioxidant and pro-oxidant effects. In previous studies, HO-1 overexpression protected astrocytes from heme-mediated injury in vitro. In the present study, we tested the hypothesis that selective astrocyte overexpression of HO-1 improves outcome after intracerebral hemorrhage. METHODS: Male and female transgenic mice overexpressing human HO-1 driven by the GFAP promoter (GFAP.HMOX1) and wild-type controls received striatal injections of autologous blood (25 µL). Blood-brain barrier disruption was assessed by Evans blue assay and striatal cell viability by methylthiazolyldiphenyl-tetrazolium bromide assay. Neurological deficits were quantified by digital analysis of spontaneous cage activity, adhesive removal, and elevated body swing tests. RESULTS: Mortality rate for wild-type mice was 34.8% and was similar for males and females; all GFAP.HMOX1 mice survived. Striatal Evans blue leakage at 24 hours was 23.4±3.2 ng in surviving wild-type mice, compared with 10.9±1.8 ng in transgenics. Perihematomal cell viability was reduced to 61±4% of contralateral at 3 days in wild-type mice, versus 80±4% in transgenics. Focal neurological deficits were significantly reduced and spontaneous cage activity was increased in GFAP.HMOX1 mice. CONCLUSIONS: Selective HO-1 overexpression in astrocytes reduces mortality, blood-brain barrier disruption, perihematomal cell injury, and neurological deficits in an autologous blood injection intracerebral hemorrhage model. Genetic or pharmacological therapies that acutely increase astrocyte HO-1 may be beneficial after intracerebral hemorrhage.


Assuntos
Astrócitos/enzimologia , Hemorragia Cerebral/enzimologia , Heme Oxigenase-1/metabolismo , Animais , Feminino , Proteína Glial Fibrilar Ácida , Heme Oxigenase-1/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso
11.
Int J Mol Sci ; 16(3): 5400-19, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25761244

RESUMO

Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. The Hmox1 promoter contains numerous consensus sequences that render the gene exquisitely sensitive to induction by diverse pro-oxidant and inflammatory stimuli. In "stressed" astroglia, HO-1 hyperactivity promotes mitochondrial iron sequestration and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergetic failure documented in Alzheimer disease, Parkinson disease and certain neurodevelopmental conditions. Glial HO-1 expression may also impact neuroplasticity and cell survival by modulating brain sterol metabolism and the proteasomal degradation of neurotoxic proteins. The glial HO-1 response may represent a pivotal transducer of noxious environmental and endogenous stressors into patterns of neural damage and repair characteristic of many human degenerative and developmental CNS disorders.


Assuntos
Doença de Alzheimer/metabolismo , Heme Oxigenase-1/metabolismo , Doença de Parkinson/metabolismo , Transdução de Sinais , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Animais , Heme Oxigenase-1/genética , Humanos , Ferro/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/terapia
12.
J Neurochem ; 131(6): 778-90, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25111043

RESUMO

Heme oxygenase-1 (HO-1) encoded by the HMOX1 gene is a 32-kDa stress protein that catabolizes heme to biliverdin, free iron, and carbon monoxide (CO). Glial HO-1 is over-expressed in the CNS of subjects with Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). The HMOX1 gene is exquisitely sensitive to oxidative stress and is induced in brain and other tissues in various models of disease and trauma. Induction of the glial HMOX1 gene may lead to pathological brain iron deposition, intracellular oxidative damage, and bioenergetic failure in AD and other human CNS disorders such as PD and MS. Therefore, targeted suppression of glial HO-1 hyperactivity may prove to be a rational and effective therapeutic intervention in AD and related neurodegenerative disorders. In this study, we report the effects of QC-47, QC-56, and OB-28, novel azole-based competitive and reversible inhibitors of HO-1, on oxidative damage to whole-cell and mitochondrial compartments in rat astrocytes transfected with the HMOX1 gene. We also report the effect of OB-28 on the behavior and neuropathology of APP(swe)/PS1(∆E9) mice. OB-28 was found to reduce oxidative damage to whole-cell and mitochondrial compartments in rat astrocytes transfected with the HMOX1 gene. Moreover, OB-28 was found to significantly counter behavioral deficits and neuropathological alterations in APP(swe)/PS1(∆E9) mice. Attenuation of AD-associated behavioral deficits and neuropathological changes suggests that HO-1 may be a promising target for neuroprotective intervention in AD and other neurodegenerative diseases. We propose that the targeted suppression of glial heme oxygenase-1 (HO-1) hyperactivity may prove to be a rational and effective therapeutic intervention in Alzheimer's disease (AD) and related neurodegenerative disorders. We report attenuation by a selective HO-1 inhibitor of oxidative damage to whole-cell and mitochondrial compartments in astrocytes in vitro and amelioration of behavioral anomalies in a transgenic mouse model of AD.


Assuntos
Doença de Alzheimer/metabolismo , Astrócitos/efeitos dos fármacos , Azóis/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Mitocôndrias/efeitos dos fármacos , Envelhecimento/metabolismo , Doença de Alzheimer/genética , Animais , Modelos Animais de Doenças , Heme Oxigenase-1/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neuroglia/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
13.
Anat Rec (Hoboken) ; 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38606671

RESUMO

Subtle craniofacial dysmorphology has been reported in schizophrenia patients. This dysmorphology includes midline facial elongation, frontonasal anomalies and a sexually dimorphic deviation from normal directional asymmetry of the face, with male patients showing reduced and female patients showing enhanced facial asymmetry relative to healthy control subjects. GFAP.HMOX10-12m transgenic mice (Mus musculus) that overexpress heme oxygenase-1 in astrocytes recapitulate many schizophrenia-relevant neurochemical, neuropathological and behavioral features. As morphogenesis of the brain, skull and face are highly interrelated, we hypothesized that GFAP.HMOX10-12m mice may exhibit craniofacial anomalies similar to those reported in persons with schizophrenia. We examined craniofacial anatomy in male GFAP.HMOX10-12m mice and wild-type control mice at the early adulthood age of 6-8 months. We used computer vision techniques for the extraction and analysis of mouse head shape parameters from systematically acquired 2D digital images, and confirmed our results with landmark-based geometric morphometrics. We performed skull bone morphometry using digital calipers to take linear distance measurements between known landmarks. Relative to controls, adult male GFAP.HMOX10-12m mice manifested craniofacial dysmorphology including elongation of the nasal bones, alteration of head shape anisotropy and reduction of directional asymmetry in facial shape features. These findings demonstrate that GFAP.HMOX10-12m mice exhibit craniofacial anomalies resembling those described in schizophrenia patients, implicating heme oxygenase-1 in their development. As a preclinical mouse model, GFAP.HMOX10-12m mice provide a novel opportunity for the study of the etiopathogenesis of craniofacial and other anomalies in schizophrenia and related disorders.

14.
J Neurosci ; 32(32): 10841-53, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22875919

RESUMO

Delineation of key molecules that act epigenetically to transduce diverse stressors into established patterns of disease would facilitate the advent of preventive and disease-modifying therapeutics for a host of neurological disorders. Herein, we demonstrate that selective overexpression of the stress protein heme oxygenase-1 (HO-1) in astrocytes of novel GFAP.HMOX1 transgenic mice results in subcortical oxidative stress and mitochondrial damage/autophagy; diminished neuronal reelin content (males); induction of Nurr1 and Pitx3 with attendant suppression of their targeting miRNAs, 145 and 133b; increased tyrosine hydroxylase and α-synuclein expression with downregulation of the targeting miR-7b of the latter; augmented dopamine and serotonin levels in basal ganglia; reduced D1 receptor binding in nucleus accumbens; axodendritic pathology and altered hippocampal cytoarchitectonics; impaired neurovascular coupling; attenuated prepulse inhibition (males); and hyperkinetic behavior. The GFAP.HMOX1 neurophenotype bears resemblances to human schizophrenia and other neurodevelopmental conditions and implicates glial HO-1 as a prime transducer of inimical (endogenous and environmental) influences on the development of monoaminergic circuitry. Containment of the glial HO-1 response to noxious stimuli at strategic points of the life cycle may afford novel opportunities for the effective management of human neurodevelopmental and neurodegenerative conditions.


Assuntos
Astrócitos/metabolismo , Encéfalo/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Heme Oxigenase-1/metabolismo , Esquizofrenia/genética , Esquizofrenia/patologia , Estimulação Acústica , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/ultraestrutura , Benzamidas/farmacocinética , Benzazepinas/farmacocinética , Monoaminas Biogênicas/metabolismo , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Dopaminérgicos/farmacocinética , Embrião de Mamíferos , Ensaio de Imunoadsorção Enzimática , Transtornos Neurológicos da Marcha/etiologia , Transtornos Neurológicos da Marcha/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Heme Oxigenase-1/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Inibição Psicológica , Fluxometria por Laser-Doppler , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , MicroRNAs/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Proteína Reelina , Esquizofrenia/fisiopatologia , Filtro Sensorial/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trítio/farmacocinética , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
Biochim Biophys Acta ; 1822(3): 350-60, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21782937

RESUMO

Iron participates in a wide array of cellular functions and is essential for normal neural development and physiology. However, if inappropriately managed, the transition metal is capable of generating neurotoxic reactive oxygen species. A number of hereditary conditions perturb body iron homeostasis and some, collectively referred to as neurodegeneration with brain iron accumulation (NBIA), promote pathological deposition of the metal predominantly or exclusively within the central nervous system (CNS). In this article, we discuss seven NBIA disorders with emphasis on the clinical syndromes and neuroimaging. The latter primarily entails magnetic resonance scanning using iron-sensitive sequences. The conditions considered are Friedreich ataxia (FA), pantothenate kinase 2-associated neurodegeneration (PKAN), PLA2G6-associated neurodegeneration (PLAN), FA2H-associated neurodegeneration (FAHN), Kufor-Rakeb disease (KRD), aceruloplasminemia, and neuroferritinopathy. An approach to differential diagnosis and the status of iron chelation therapy for several of these entities are presented. This article is part of a Special Issue entitled: Imaging Brain Aging and Neurodegenerative disease.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Ferro/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Humanos , Doenças Neurodegenerativas/diagnóstico , Neuroimagem/métodos , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia
16.
J Neurochem ; 123(2): 325-36, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22881289

RESUMO

The mechanisms responsible for pathological iron deposition in the aging and degenerating mammalian CNS remain poorly understood. The stress protein, HO-1 mediates the degradation of cellular heme to biliverdin/bilirubin, free iron, and CO and is up-regulated in the brains of persons with Alzheimer's disease and Parkinson's disease. HO-1 induction in primary astroglial cultures promotes deposition of non-transferrin iron, mitochondrial damage and macroautophagy, and predisposes cocultured neuronal elements to oxidative injury. To gain a better appreciation of the role of glial HO-1 in vivo, we probed for aberrant brain iron deposition using Perls' method and dynamic secondary ion mass spectrometry in novel, conditional GFAP.HMOX1 transgenic mice that selectively over-express human HO-1 in the astrocytic compartment. At 48 weeks, the GFAP.HMOX1 mice exhibited increased deposits of glial iron in hippocampus and other subcortical regions without overt changes in iron-regulatory and iron-binding proteins relative to age-matched wild-type animals. Dynamic secondary ion mass spectrometry revealed abundant FeO⁻ signals in the transgenic, but not wild-type, mouse brain that colocalized to degenerate mitochondria and osmiophilic cytoplasmic inclusions (macroautophagy) documented by TEM. Sustained up-regulation of HO-1 in astrocytes promotes pathological brain iron deposition and oxidative mitochondrial damage characteristic of Alzheimer's disease-affected neural tissues. Curtailment of glial HO-1 hyperactivity may limit iron-mediated cytotoxicity in aging and degenerating neural tissues.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Regulação Enzimológica da Expressão Gênica , Heme Oxigenase-1/biossíntese , Ferro/metabolismo , Regulação para Cima/fisiologia , Animais , Astrócitos/patologia , Encéfalo/patologia , Heme Oxigenase-1/genética , Humanos , Sobrecarga de Ferro/metabolismo , Camundongos , Camundongos Transgênicos , Regulação para Cima/genética
17.
J Oral Pathol Med ; 41(3): 235-41, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22077396

RESUMO

BACKGROUND: Oxidative stress has been implicated in the pathogenesis of diabetes mellitus (DM). Levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), 8-epi-prostaglandin-F(2α) (8-epi-PGF2α), and total protein carbonyls were measured to assess whether DM is associated with altered salivary redox homeostasis. METHODS: A total of 215 patients with diabetes and 481 healthy controls were recruited from the Department of Endocrinology at the Jewish General Hospital in Montreal. Levels of oxidative biomarkers were assayed using enzyme-linked immunosorbent assay (ELISA) in whole unstimulated saliva. Associations of the redox data with exposure to insulin, metformin and dietary control were assessed by logistic regression analyses. RESULTS: We observed (i) significantly higher mean levels of 8-OHdG and protein carbonyls in whole unstimulated saliva of patients with diabetes compared to controls, (ii) higher mean levels of protein carbonyls in type 1 diabetes as well as higher mean levels of 8-OHdG and protein carbonyls in type 2 diabetes compared to controls, (iii) elevated levels of protein carbonyls in diet-controlled patients and in patients with diabetes on insulin and metformin, (iv) elevated levels of 8-OHdG in patients on metformin, and (v) significant associations between subjects with DM and salivary 8-OHdG and protein carbonyls. CONCLUSION: DM is associated with increased oxidative modification of salivary DNA and proteins. Salivary redox homeostasis is perturbed in DM and may inform on the presence of the disease and efficacy of therapeutic interventions.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Estresse Oxidativo/fisiologia , Saliva/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Fatores Etários , Biomarcadores/análise , Dano ao DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análise , Desoxiguanosina/metabolismo , Diabetes Mellitus Tipo 1/dietoterapia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dieta para Diabéticos , Dinoprosta/análogos & derivados , Dinoprosta/análise , Dinoprosta/metabolismo , Homeostase/fisiologia , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Peroxidação de Lipídeos , Metformina/uso terapêutico , Oxirredução , Carbonilação Proteica , Proteínas e Peptídeos Salivares/análise , Proteínas e Peptídeos Salivares/metabolismo , Fatores Sexuais
18.
Neuroimage ; 56(3): 951-60, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21073964

RESUMO

MRI-based measurements of surface cortical thickness (SCT) have become a sensitive tool to quantify changes in cortical morphology. When comparing SCT to histological cortical thickness maps, a good correspondence can be found for many but not all human brain areas. Discrepancies especially arise in the sensory motor cortex, where histological cortical thickness is high, but SCT is very low. The aim of this study was to determine whether the relationship between cortical thickness and neuronal density is the same for different cytoarchitectonic areas throughout homo- and heterotypical isocortex. We assessed this relationship using high-resolution [(18)F]-labelled flumazenil (FMZ) PET and SCT-mapping. FMZ binds to the benzodiazepine GABA(A) receptor complex which is localized on axo-dendritic synapses, with a cortical distribution closely following the local density of neurons. SCT and voxelwise FMZ binding potential (BP(ND)) were assessed in ten healthy subjects. After partial volume correction, two subsets with a differential relationship between SCT and BP(ND) were identified: a fronto-parietal homotypical subset where neuronal density is relatively constant and mainly independent of SCT, and a subset comprising heterotypical and mainly temporal and occipital homotypical regions where neuronal density is negatively correlated with SCT. This is the first in-vivo study demonstrating a differential relationship between SCT, neuronal density and cytoarchitectonics in humans. These findings are of direct relevance for the correct interpretation of SCT-based morphometry studies, in that there is no simple relationship between apparent cortical thickness and neuronal density, here attributed to FMZ binding, holding for all cortical regions.


Assuntos
Córtex Cerebral/diagnóstico por imagem , Flumazenil , Moduladores GABAérgicos , Neurônios/diagnóstico por imagem , Compostos Radiofarmacêuticos , Idoso , Algoritmos , Mapeamento Encefálico , Córtex Cerebral/citologia , Interpretação Estatística de Dados , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Individualidade , Marcação por Isótopo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Receptores de GABA/metabolismo , Análise de Regressão
19.
J Neural Transm (Vienna) ; 118(3): 381-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20563825

RESUMO

Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. In AD and mild cognitive impairment (MCI), immunoreactive HO-1 protein is over-expressed in astrocytes and neurons of the hippocampus and cerebral cortex and co-localizes to neurofibrillary tangles, senile plaques and corpora amylacea. Astroglial induction of the Hmox1 gene by ß-amyloid, pro-inflammatory cytokines and hydrogen peroxide promotes mitochondrial sequestration of non-transferrin iron and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergy failure amply documented in AD-affected neural tissues. Glial HO-1 expression may also impact cell survival and neuroplasticity in AD by modulating brain sterol/oxysterol metabolism and the degradation of tau by the proteasome. Suppression of glial HO-1 activity by pharmacological or other means may confer neuroprotection in AD by curtailing iron-mediated neurotoxicity.


Assuntos
Doença de Alzheimer/metabolismo , Transtornos Cognitivos/metabolismo , Heme Oxigenase-1/metabolismo , Mitocôndrias/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/patologia , História do Século XX , Humanos , Mitocôndrias/patologia , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/fisiologia
20.
Can J Neurol Sci ; 38(4): 580-92, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21672698

RESUMO

Alzheimer disease (AD) is a dementing, neurodegenerative disorder that affects approximately 500,000 Canadians and its prevalence is expected to double over the next 30 years. Although several medications may temporarily augment cognitive abilities in AD, there presently exists no proven method to avoid the inevitable clinical deterioration in this devastating condition. The delineation of risk factors for the development of AD offers hope for the advent of effective prevention or interventions that might retard the onset of symptoms. In this article, we provide a comprehensive review of midlife risk factors implicated in the etiopathogenesis of sporadic AD. Although some risk factors are heritable and largely beyond our control, others are determined by lifestyle or environment and are potentially modifiable. In a companion paper, we introduce the concept of an Alzheimer Risk Assessment Clinic for ascertainment and mitigation of these and other putative dementia risk factors in middle-aged adults.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/etiologia , Doença de Alzheimer/complicações , Doença de Alzheimer/epidemiologia , Canadá/epidemiologia , Humanos , Medição de Risco , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA