Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
NPJ Parkinsons Dis ; 10(1): 90, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664405

RESUMO

Gingipains are protease virulence factors produced by Porphyromonas gingivalis, a Gram-negative bacterium best known for its role in chronic periodontitis. Gingipains were recently identified in the middle temporal gyrus of postmortem Alzheimer's disease (AD) brains, where gingipain load correlated with AD diagnosis and tau and ubiquitin pathology. Since AD and Parkinson's disease (PD) share some overlapping pathologic features, including nigral pathology and Lewy bodies, the current study explored whether gingipains are present in the substantia nigra pars compacta of PD brains. In immunohistochemical techniques and multi-channel fluorescence studies, gingipain antigens were abundant in dopaminergic neurons in the substantia nigra of both PD and neurologically normal control brains. 3-dimensional reconstructions of Lewy body containing neurons revealed that gingipains associated with the periphery of alpha-synuclein aggregates but were occasionally observed inside aggregates. In vitro proteomic analysis demonstrated that recombinant alpha-synuclein is cleaved by lysine-gingipain, generating multiple alpha-synuclein fragments including the non-amyloid component fragments. Immunogold electron microscopy with co-labeling of gingipains and alpha-synuclein confirmed the occasional colocalization of gingipains with phosphorylated (pSER129) alpha-synuclein. In dopaminergic neurons, gingipains localized to the perinuclear cytoplasm, neuromelanin, mitochondria, and nucleus. These data suggest that gingipains localize in dopaminergic neurons in the substantia nigra and interact with alpha-synuclein.

2.
J Alzheimers Dis ; 75(4): 1361-1376, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32390638

RESUMO

BACKGROUND: Porphyromonas gingivalis (P. gingivalis) and its gingipain virulence factors have been identified as pathogenic effectors in Alzheimer's disease (AD). In a recent study we demonstrated the presence of gingipains in over 90% of postmortem AD brains, with gingipains localizing to the cytoplasm of neurons. However, infection of neurons by P. gingivalis has not been previously reported. OBJECTIVE: To demonstrate intraneuronal P. gingivalis and gingipain expression in vitro after infecting neurons derived from human inducible pluripotent stem cells (iPSC) with P. gingivalis for 24, 48, and 72 h. METHODS: Infection was characterized by transmission electron microscopy, confocal microscopy, and bacterial colony forming unit assays. Gingipain expression was monitored by immunofluorescence and RT-qPCR, and protease activity monitored with activity-based probes. Neurodegenerative endpoints were assessed by immunofluorescence, western blot, and ELISA. RESULTS: Neurons survived the initial infection and showed time dependent, infection induced cell death. P. gingivalis was found free in the cytoplasm or in lysosomes. Infected neurons displayed an accumulation of autophagic vacuoles and multivesicular bodies. Tau protein was strongly degraded, and phosphorylation increased at T231. Over time, the density of presynaptic boutons was decreased. CONCLUSION: P. gingivalis can invade and persist in mature neurons. Infected neurons display signs of AD-like neuropathology including the accumulation of autophagic vacuoles and multivesicular bodies, cytoskeleton disruption, an increase in phospho-tau/tau ratio, and synapse loss. Infection of iPSC-derived mature neurons by P. gingivalis provides a novel model system to study the cellular mechanisms leading to AD and to investigate the potential of new therapeutic approaches.


Assuntos
Doença de Alzheimer/microbiologia , Doença de Alzheimer/patologia , Infecções por Bacteroidaceae/complicações , Cisteína Endopeptidases Gingipaínas/metabolismo , Neurônios/microbiologia , Neurônios/patologia , Doença de Alzheimer/enzimologia , Animais , Células Cultivadas , Camundongos , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/microbiologia , Células-Tronco Neurais/patologia , Neurônios/enzimologia , Porphyromonas gingivalis
3.
Pharmacol Res Perspect ; 8(1): e00562, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31999052

RESUMO

COR388, a small-molecule lysine-gingipain inhibitor, is currently being investigated in a Phase 2/3 clinical trial for Alzheimer's disease (AD) with exploratory endpoints in periodontal disease. Gingipains are produced by two species of bacteria, Porphyromonas gingivalis and Porphyromonas gulae, typically associated with periodontal disease and systemic infections in humans and dogs, respectively. P. gulae infection in dogs is associated with periodontal disease, which provides a physiologically relevant model to investigate the pharmacology of COR388. In the current study, aged dogs with a natural oral infection of P. gulae and periodontal disease were treated with COR388 by oral administration for up to 90 days to assess lysine-gingipain target engagement and reduction of bacterial load and downstream pathology. In a 28-day dose-response study, COR388 inhibited the lysine-gingipain target and reduced P. gulae load in saliva, buccal cells, and gingival crevicular fluid. The lowest effective dose was continued for 90 days and was efficacious in continuous reduction of bacterial load and downstream periodontal disease pathology. In a separate histology study, dog brain tissue showed evidence of P. gulae DNA and neuronal lysine-gingipain, demonstrating that P. gulae infection is systemic and spreads beyond its oral reservoir, similar to recent observations of P. gingivalis in humans. Together, the pharmacokinetics and pharmacodynamics of COR388 lysine-gingipain inhibition, along with reduction of bacterial load and periodontal disease in naturally occurring P. gulae infection in the dog, support the use of COR388 in targeting lysine-gingipain and eliminating P. gingivalis infection in humans.


Assuntos
Infecções por Bacteroidaceae/tratamento farmacológico , Doenças do Cão/microbiologia , Cisteína Endopeptidases Gingipaínas/antagonistas & inibidores , Compostos Orgânicos/administração & dosagem , Doenças Periodontais/tratamento farmacológico , Porphyromonas/enzimologia , Bibliotecas de Moléculas Pequenas/administração & dosagem , Administração Oral , Envelhecimento/sangue , Animais , Carga Bacteriana , Proteínas de Bactérias/antagonistas & inibidores , Infecções por Bacteroidaceae/veterinária , Encéfalo/efeitos dos fármacos , Encéfalo/microbiologia , Doenças do Cão/tratamento farmacológico , Cães , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Líquido do Sulco Gengival/efeitos dos fármacos , Líquido do Sulco Gengival/microbiologia , Compostos Orgânicos/química , Compostos Orgânicos/farmacologia , Doenças Periodontais/veterinária , Porphyromonas/efeitos dos fármacos , Porphyromonas/patogenicidade , Saliva/efeitos dos fármacos , Saliva/microbiologia , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
4.
Am J Pathol ; 172(6): 1520-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18467698

RESUMO

The hippocampus in Alzheimer's disease is burdened with amyloid plaques and is one of the few locations where neurogenesis continues throughout adult life. To evaluate the impact of amyloid-beta deposition on neural stem cells, hippocampal neurogenesis was assessed using bromodeoxyuridine incorporation and doublecortin staining in two amyloid precursor protein (APP) transgenic mouse models. In 5-month-old APP23 mice prior to amyloid deposition, neurogenesis showed no robust difference relative to wild-type control mice, but 25-month-old amyloid-depositing APP23 mice showed significant increases in neurogenesis compared to controls. In contrast, 8-month-old amyloid-depositing APPPS1 mice revealed decreases in neurogenesis compared to controls. To study whether alterations in neurogenesis are the result of amyloid-induced changes at the level of neural stem cells, APPPS1 mice were crossed with mice expressing green fluorescence protein (GFP) under a central nervous system-specific nestin promoter. Eight-month-old nestin-GFP x APPPS1 mice exhibited decreases in quiescent nestin-positive astrocyte-like stem cells, while transient amplifying progenitor cells did not change in number. Strikingly, both astrocyte-like and transient-amplifying progenitor cells revealed an aberrant morphologic reaction toward congophilic amyloid-deposits. A similar reaction toward the amyloid was no longer observed in doublecortin-positive immature neurons. Results provide evidence for a disruption of neural stem cell biology in an amyloidogenic environment and support findings that neurogenesis is differently affected among various transgenic mouse models of Alzheimer's disease.


Assuntos
Amiloidose/patologia , Hipocampo/patologia , Neurônios/patologia , Placa Amiloide/patologia , Células-Tronco/patologia , Envelhecimento/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/fisiologia , Células-Tronco/fisiologia
5.
Sci Adv ; 5(1): eaau3333, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30746447

RESUMO

Porphyromonas gingivalis, the keystone pathogen in chronic periodontitis, was identified in the brain of Alzheimer's disease patients. Toxic proteases from the bacterium called gingipains were also identified in the brain of Alzheimer's patients, and levels correlated with tau and ubiquitin pathology. Oral P. gingivalis infection in mice resulted in brain colonization and increased production of Aß1-42, a component of amyloid plaques. Further, gingipains were neurotoxic in vivo and in vitro, exerting detrimental effects on tau, a protein needed for normal neuronal function. To block this neurotoxicity, we designed and synthesized small-molecule inhibitors targeting gingipains. Gingipain inhibition reduced the bacterial load of an established P. gingivalis brain infection, blocked Aß1-42 production, reduced neuroinflammation, and rescued neurons in the hippocampus. These data suggest that gingipain inhibitors could be valuable for treating P. gingivalis brain colonization and neurodegeneration in Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/microbiologia , Infecções por Bacteroidaceae/tratamento farmacológico , Encéfalo/microbiologia , Encéfalo/patologia , Fármacos Neuroprotetores/uso terapêutico , Porphyromonas gingivalis/enzimologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Infecções por Bacteroidaceae/microbiologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Cisteína Endopeptidases Gingipaínas/antagonistas & inibidores , Cisteína Endopeptidases Gingipaínas/metabolismo , Cisteína Endopeptidases Gingipaínas/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/metabolismo , Projetos Piloto , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/genética , Estudos Prospectivos , Saliva/microbiologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas tau/metabolismo
6.
J Neurosci ; 25(48): 11125-32, 2005 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16319312

RESUMO

The significance of the peripheral immune system in Alzheimer's disease pathogenesis remains controversial. To study the CNS invasion of hematopoietic cells in the course of cerebral amyloidosis, we used a green fluorescence protein (GFP)-bone marrow chimeric amyloid precursor protein transgenic mouse model (APP23 mice). No difference in the number of GFP-positive invading cells was observed between young APP23 mice and nontransgenic control mice. In contrast, in aged, amyloid-depositing APP23 mice, a significant increase in the number of invading ameboid-like GFP-positive cells was found compared with age-matched nontransgenic control mice. Interestingly, independent of the time after transplantation, only a subpopulation of amyloid deposits was surrounded by invading cells. This suggests that not all amyloid plaques are a target for invading cells or, alternatively, all amyloid plaques attract invading cells but only for a limited time, possibly at an early stage of plaque evolution. Immunological and ultrastructural phenotyping revealed that macrophages and T-cells accounted for a significant portion of these ameboid-like invading cells. Macrophages did not show evidence of amyloid phagocytosis at the electron microscopic level, and no obvious signs for T-cell-mediated inflammation or neurodegeneration were observed. The observation that hematopoietic cells invade the brain in response to cerebral amyloidosis may hold an unrecognized therapeutic potential.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Células da Medula Óssea , Transplante de Medula Óssea , Encéfalo/metabolismo , Movimento Celular , Hematopoese , Amiloide , Neuropatias Amiloides/metabolismo , Neuropatias Amiloides/patologia , Neuropatias Amiloides/fisiopatologia , Animais , Células da Medula Óssea/patologia , Encefalopatias/metabolismo , Encefalopatias/patologia , Encefalopatias/fisiopatologia , Proteínas de Fluorescência Verde , Macrófagos , Masculino , Camundongos , Camundongos Transgênicos , Fagocitose , Linfócitos T/patologia , Quimeras de Transplante
7.
J Neurosci ; 22(2): 515-22, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11784797

RESUMO

APP23 transgenic mice express mutant human amyloid precursor protein and develop amyloid plaques predominantly in neocortex and hippocampus progressively with age, similar to Alzheimer's disease. We have previously reported neuron loss in the hippocampal CA1 region of 14- to 18-month-old APP23 mice. In contrast, no neuron loss was found in neocortex. In the present study we have reinvestigated neocortical neuron numbers in adult and aged APP23 mice. Surprisingly, results revealed that 8-month-old APP23 mice have 13 and 14% more neocortical neurons compared with 8-month-old wild-type and 27-month-old APP23 mice, respectively. In 27-month-old APP23 mice we found an inverse correlation between amyloid load and neuron number. These results suggest that APP23 mice have more neurons until they develop amyloid plaques but then lose neurons in the process of cerebral amyloidogenesis. Supporting this notion, we found more neurons with a necrotic-apoptotic phenotype in the neocortex of 24-month-old APP23 mice compared with age-matched wild-type mice. Stimulated by recent reports that demonstrated neurogenesis after targeted neuron death in the mouse neocortex, we have also examined neurogenesis in APP23 mice. Strikingly, we found a fourfold to sixfold increase in newly produced cells in 24-month-old APP23 mice compared with both age-matched wild-type mice and young APP23 transgenic mice. However, subsequent cellular phenotyping revealed that none of the newly generated cells in neocortex had a neuronal phenotype. The majority were microglial and to a lesser extent astroglial cells. We conclude that cerebral amyloidosis in APP23 mice causes a modest neuron loss in neocortex and induces marked gliogenesis.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Amiloidose/metabolismo , Neocórtex/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloidose/patologia , Animais , Bromodesoxiuridina , Contagem de Células , Morte Celular , Divisão Celular , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/patologia , Neuroglia/patologia , Neurônios/patologia , Fenótipo
8.
Neurobiol Aging ; 25(3): 333-40, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15123339

RESUMO

Age-related changes in neurogenesis and its modulation by caloric restriction (CR) were studied in C57BL/6 mice. To this end, bromodeoxyuridine (BrdU) labeling was used to assess neuronal and glial precursor proliferation and survival in the granular cell layer (GCL) and the hilus of the dentate gyrus of 2-, 12-, 18-, and 24-month-old mice. For both regions, we found an age-dependent decrease in proliferation but not in survival of newborn cells. Interestingly, the reduction in proliferation occurred between 2 and 18 months of age with no additional decline between 18- and 24-month-old mice. Phenotyping of the newborn cells revealed a decrease in the neuron fraction in the GCL between 2 and 12 months of age but not thereafter. The majority of BrdU cells in the hilus colocalized with astrocytic but none with neuronal markers. CR from 3 to 11 months of age had no effect on neurogenesis in the GCL, but had a survival-promoting effect on newly generated glial cells in the hilus of the dentate gyrus. In conclusion, C57BL/6 mice reveal a substantial reduction in neurogenesis in the dentate gyrus until late adulthood with no further decline with aging. Long-term CR does not counteract this age-related decline in neurogenesis but promotes survival of hilar glial cells.


Assuntos
Envelhecimento/fisiologia , Restrição Calórica , Giro Denteado/crescimento & desenvolvimento , Privação de Alimentos/fisiologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Biomarcadores , Bromodesoxiuridina , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Sobrevivência Celular/fisiologia , Giro Denteado/citologia , Giro Denteado/fisiologia , Regulação para Baixo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
9.
PLoS One ; 8(4): e61789, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23613937

RESUMO

Research with experimental stroke models has identified a wide range of therapeutic proteins that can prevent the brain damage caused by this form of acute neurological injury. Despite this, we do not yet have safe and effective ways to deliver therapeutic proteins to the injured brain, and this remains a major obstacle for clinical translation. Current targeted strategies typically involve invasive neurosurgery, whereas systemic approaches produce the undesirable outcome of non-specific protein delivery to the entire brain, rather than solely to the injury site. As a potential way to address this, we developed a protein delivery system modeled after the endogenous immune cell response to brain injury. Using ex-vivo-engineered dendritic cells (DCs), we find that these cells can transiently home to brain injury in a rat model of stroke with both temporal and spatial selectivity. We present a standardized method to derive injury-responsive DCs from bone marrow and show that injury targeting is dependent on culture conditions that maintain an immature DC phenotype. Further, we find evidence that when loaded with therapeutic cargo, cultured DCs can suppress initial neuron death caused by an ischemic injury. These results demonstrate a non-invasive method to target ischemic brain injury and may ultimately provide a way to selectively deliver therapeutic compounds to the injured brain.


Assuntos
Encéfalo/metabolismo , Células Dendríticas , Proteínas/administração & dosagem , Proteínas/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Lesões Encefálicas/tratamento farmacológico , Modelos Animais de Doenças , Ratos
10.
Syst Biol Reprod Med ; 56(4): 292-302, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20662593

RESUMO

We evaluated the impact of uteroplacental insufficiency (UPI), and subsequent intrauterine growth restriction (IUGR), on serum testosterone and hippocampal expression of Cyp19a1 variants and aromatase in rats. Additionally, we determined UPI induced histone modification of the promoter regions of Cyp19a1 variants using chromatin immunoprecipitation. Cyp19a1 is the gene encoding the protein aromatase, that catalyzes the biosynthesis of estrogens from androgens and is necessary for masculinization of the brain. IUGR was induced via bilateral uterine artery. UPI increased serum testosterone in day of life 0 (D(0)) and day of life 21 (D(21)) IUGR males to 224% and 299% of control values, respectively. While there was no significant impact of UPI on testosterone in D(0) females, testosterone in D(21) IUGR females was 187% of controls. Cyp19a1 variant 1.f and variant II are expressed in the rat hippocampus at D(0) and D(21). UPI significantly reduced expression of Cyp19a1 variant 1.f in D(0) males, with no impact in females. Similarly at D(0), UPI reduced expression of aromatase, the protein encoded by Cyp19a1, in males. Dimethylation of H3K4 was increased in the promoter region of variant 1.f (P1.f) and trimethylation of H3K4 was decreased in the promoter region of variant II (PII). At D(21), dimethylation of H3K4 is significantly reduced in PII of IUGR males. We conclude that UPI increases serum testosterone and reduces Cyp19a1 variant 1.f expression in the hippocampus of D(0) IUGR males. Additionally, UPI alters the chromatin structure of CYP19a1 at both D(0) and D(21).


Assuntos
Aromatase/genética , Cromatina/química , Retardo do Crescimento Fetal/enzimologia , Animais , Aromatase/química , Sequência de Bases , Imunoprecipitação da Cromatina , Primers do DNA , Feminino , Masculino , Regiões Promotoras Genéticas , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA