Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Neurosci ; 125: 103855, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37084991

RESUMO

Traumatic brain injury is a leading cause of morbidity and mortality in adults and children in developed nations. Following the primary injury, microglia, the resident innate immune cells of the CNS, initiate several inflammatory signaling cascades and pathophysiological responses that may persist chronically; chronic neuroinflammation following TBI has been closely linked to the development of neurodegeneration and neurological dysfunction. Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that have been shown to regulate several key mechanisms in the inflammatory response to TBI. Increasing evidence has shown that the modulation of the PI3K/AKT signaling pathway has the potential to influence the cellular response to inflammatory stimuli. However, directly targeting PI3K signaling poses several challenges due to its regulatory role in several cell survival pathways. We have previously identified that the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), the major negative regulator of PI3K/AKT signaling, is dysregulated following exposure to repetitive mild traumatic brain injury (r-mTBI). Moreover, this dysregulated PI3K/AKT signaling was correlated with chronic microglial-mediated neuroinflammation. Therefore, we interrogated microglial-specific PTEN as a therapeutic target in TBI by generating a microglial-specific, Tamoxifen inducible conditional PTEN knockout model using a CX3CR1 Cre recombinase mouse line PTENfl/fl/CX3CR1+/CreERT2 (mcg-PTENcKO), and exposed them to our 20-hit r-mTBI paradigm. Animals were treated with tamoxifen at 76 days post-last injury, and the effects of microglia PTEN deletion on immune-inflammatory responses were assessed at 90-days post last injury. We observed that the deletion of microglial PTEN ameliorated the proinflammatory response to repetitive brain trauma, not only reducing chronic microglial activation and proinflammatory cytokine production but also rescuing TBI-induced reactive astrogliosis, demonstrating that these effects extended beyond microglia alone. Additionally, we observed that the pharmacological inhibition of PTEN with BpV(HOpic) ameliorated the LPS-induced activation of microglial NFκB signaling in vitro. Together, these data provide support for the role of PTEN as a regulator of chronic neuroinflammation following repetitive mild TBI.


Assuntos
Lesões Encefálicas Traumáticas , Microglia , Animais , Camundongos , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neuroinflamatórias , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
Neurobiol Dis ; 150: 105237, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33383188

RESUMO

A pathological characteristic of repetitive traumatic brain injury (TBI) is the deposition of hyperphosphorylated and aggregated tau species in the brain and increased levels of extracellular monomeric tau are believed to play a role in the pathogenesis of neurodegenerative tauopathies. The pathways by which extracellular tau is eliminated from the brain, however, remains elusive. The purpose of this study was to examine tau uptake by cerebrovascular cells and the effect of TBI on these processes. We found monomeric tau interacts with brain vascular mural cells (pericytes and smooth muscle cells) to a greater extent than other cerebrovascular cells, indicating mural cells may contribute to the elimination of extracellular tau, as previously described for other solutes such as beta-amyloid. Consistent with other neurodegenerative disorders, we observed a progressive decline in cerebrovascular mural cell markers up to 12 months post-injury in a mouse model of repetitive mild TBI (r-mTBI) and human TBI brain specimens, when compared to control. These changes appear to reflect mural cell degeneration and not cellular loss as no difference in the mural cell population was observed between r-mTBI and r-sham animals as determined through flow cytometry. Moreover, freshly isolated r-mTBI cerebrovessels showed reduced tau uptake at 6 and 12 months post-injury compared to r-sham animals, which may be the result of diminished cerebrovascular endocytosis, as caveolin-1 levels were significantly decreased in mouse r-mTBI and human TBI cerebrovessels compared to their respective controls. Further emphasizing the interaction between mural cells and tau, similar reductions in mural cell markers, tau uptake, and caveolin-1 were observed in cerebrovessels from transgenic mural cell-depleted animals. In conclusion, our studies indicate repeated injuries to the brain causes chronic mural cell degeneration, reducing the caveolar-mediated uptake of tau by these cells. Alterations in tau uptake by vascular mural cells may contribute to tau deposition in the brain following head trauma and could represent a novel therapeutic target for TBI or other neurodegenerative disorders.


Assuntos
Doença de Alzheimer/metabolismo , Concussão Encefálica/metabolismo , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Microglia/metabolismo , Miócitos de Músculo Liso/metabolismo , Pericitos/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/irrigação sanguínea , Caveolina 1/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Presenilina-1/genética , Recidiva
3.
BMC Neurosci ; 22(1): 39, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34034683

RESUMO

BACKGROUND: Matrix metallopeptidase 9 (MMP9) has been implicated in a variety of neurological disorders, including Alzheimer's disease (AD), where MMP9 levels are elevated in the brain and cerebrovasculature. Previously our group demonstrated apolipoprotein E4 (apoE4) was less efficient in regulating MMP9 activity in the brain than other apoE isoforms, and that MMP9 inhibition facilitated beta-amyloid (Aß) elimination across the blood-brain barrier (BBB) METHODS: In the current studies, we evaluated the impact of MMP9 modulation on Aß disposition and neurobehavior in AD using two approaches, (1) pharmacological inhibition of MMP9 with SB-3CT in apoE4 x AD (E4FAD) mice, and (2) gene deletion of MMP9 in AD mice (MMP9KO/5xFAD) RESULTS: Treatment with the MMP9 inhibitor SB-3CT in E4FAD mice led to reduced anxiety compared to placebo using the elevated plus maze. Deletion of the MMP9 gene in 5xFAD mice also reduced anxiety using the open field test, in addition to improving sociability and social recognition memory, particularly in male mice, as assessed through the three-chamber task, indicating certain behavioral alterations in AD may be mediated by MMP9. However, neither pharmacological inhibition of MMP9 or gene deletion of MMP9 affected spatial learning or memory in the AD animals, as determined through the radial arm water maze. Moreover, the effect of MMP9 modulation on AD neurobehavior was not due to changes in Aß disposition, as both brain and plasma Aß levels were unchanged in the SB-3CT-treated E4FAD animals and MMP9KO/AD mice compared to their respective controls. CONCLUSIONS: In total, while MMP9 inhibition did improve specific neurobehavioral deficits associated with AD, such as anxiety and social recognition memory, modulation of MMP9 did not alter spatial learning and memory or Aß tissue levels in AD animals. While targeting MMP9 may represent a therapeutic strategy to mitigate aspects of neurobehavioral decline in AD, further work is necessary to understand the nature of the relationship between MMP9 activity and neurological dysfunction.


Assuntos
Doença de Alzheimer/metabolismo , Ansiedade/metabolismo , Metaloproteinase 9 da Matriz/deficiência , Interação Social , Aprendizagem Espacial/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/genética , Animais , Ansiedade/tratamento farmacológico , Ansiedade/genética , Ansiedade/psicologia , Encéfalo/metabolismo , Feminino , Compostos Heterocíclicos com 1 Anel/farmacologia , Compostos Heterocíclicos com 1 Anel/uso terapêutico , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Presenilina-1/genética , Interação Social/efeitos dos fármacos , Aprendizagem Espacial/efeitos dos fármacos , Sulfonas/farmacologia , Sulfonas/uso terapêutico
4.
Brain Inj ; 30(12): 1414-1427, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27834539

RESUMO

PRIMARY OBJECTIVE: To investigate the status of the cerebrovasculature following repetitive mild traumatic brain injury (r-mTBI). RESEARCH DESIGN: TBI is a risk factor for development of various neurodegenerative disorders. A common feature of neurodegenerative disease is cerebrovascular dysfunction which includes alterations in cerebral blood flow (CBF). TBI can result in transient reductions in CBF, with severe injuries often accompanied by varying degrees of vascular pathology post-mortem. However, at this stage, few studies have investigated the cerebrovasculature at chronic time points following repetitive mild brain trauma. METHODS AND PROCEDURES: r-mTBI was delivered to wild-type mice (12 months old) twice per week for 3 months and tested for spatial memory deficits (Barnes Maze task) at 1 and 6 months post-injury. At 7 months post-injury CBF was assessed via Laser Doppler Imaging and, following euthanasia, the brain was probed for markers of cerebrovascular dysfunction and inflammation. MAIN OUTCOMES AND RESULTS: Memory impairment was identified at 1 month post-injury and persisted as late as 6 months post-injury. Furthermore, significant immunopathological insult, reductions in global CBF and down-regulation of cerebrovascular-associated markers were observed. CONCLUSIONS: These results demonstrate impaired cognitive behaviour alongside chronic cerebrovascular dysfunction in a mouse model of repetitive mild brain trauma.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Circulação Cerebrovascular/fisiologia , Transtornos Cerebrovasculares/etiologia , Regulação para Baixo/fisiologia , Actinas/metabolismo , Animais , Barreira Hematoencefálica/fisiopatologia , Proteínas de Ligação ao Cálcio/metabolismo , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Laminina/metabolismo , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100/sangue
5.
J Biol Chem ; 289(49): 33927-44, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25331948

RESUMO

We have previously shown that the L-type calcium channel (LCC) antagonist nilvadipine reduces brain amyloid-ß (Aß) accumulation by affecting both Aß production and Aß clearance across the blood-brain barrier (BBB). Nilvadipine consists of a mixture of two enantiomers, (+)-nilvadipine and (-)-nilvadipine, in equal proportion. (+)-Nilvadipine is the active enantiomer responsible for the inhibition of LCC, whereas (-)-nilvadipine is considered inactive. Both nilvadipine enantiomers inhibit Aß production and improve the clearance of Aß across the BBB showing that these effects are not related to LCC inhibition. In addition, treatment of P301S mutant human Tau transgenic mice (transgenic Tau P301S) with (-)-nilvadipine reduces Tau hyperphosphorylation at several Alzheimer disease (AD) pertinent epitopes. A search for the mechanism of action of (-)-nilvadipine revealed that this compound inhibits the spleen tyrosine kinase (Syk). We further validated Syk as a target-regulating Aß by showing that pharmacological inhibition of Syk or down-regulation of Syk expression reduces Aß production and increases the clearance of Aß across the BBB mimicking (-)-nilvadipine effects. Moreover, treatment of transgenic mice overexpressing Aß and transgenic Tau P301S mice with a selective Syk inhibitor respectively decreased brain Aß accumulation and Tau hyperphosphorylation at multiple AD relevant epitopes. We show that Syk inhibition induces an increased phosphorylation of the inhibitory Ser-9 residue of glycogen synthase kinase-3ß, a primary Tau kinase involved in Tau phosphorylation, by activating protein kinase A, providing a mechanism explaining the reduction of Tau phosphorylation at GSK3ß-dependent epitopes following Syk inhibition. Altogether our data highlight Syk as a promising target for preventing both Aß accumulation and Tau hyperphosphorylation in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Células CHO , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular Tumoral , Cricetulus , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , NF-kappa B/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Nifedipino/análogos & derivados , Nifedipino/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Transdução de Sinais , Quinase Syk , Proteínas tau/genética
6.
Ann Neurol ; 75(2): 241-54, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24243523

RESUMO

OBJECTIVE: Traumatic brain injury (TBI) is a recognized risk factor for later development of neurodegenerative disease. However, the mechanisms contributing to neurodegeneration following TBI remain obscure. METHODS: In this study, we have utilized a novel mild TBI (mTBI) model to examine the chronic neurobehavioral and neuropathological outcomes following single and repetitive mTBI at time points from 6 to 18 months following injury. RESULTS: Our results reveal that at 6, 12, and 18 months after injury, animals exposed to a single mTBI have learning impairments when compared to their sham controls without exhibiting spatial memory retention deficits. In contrast, animals exposed to repetitive injury displayed persistent cognitive deficits, slower rate of learning, and progressive behavioral impairment over time. These deficits arise in parallel with a number of neuropathological abnormalities, including progressive neuroinflammation and continuing white matter degradation up to 12 months following repetitive injury. Neither single nor repetitive mTBI was associated with elevated brain levels of amyloid beta or abnormal tau phosphorylation at 6 or 12 months after injury. INTERPRETATION: Importantly, these data provide evidence that, although a single mTBI produces a clinical syndrome and pathology that remain static in the period following injury, repetitive injuries produce behavioral and pathological changes that continue to evolve many months after the initial injuries. As such, this model recapitulates many aspects described in human studies of TBI, providing a suitable platform on which to investigate the evolving pathologies following mild TBI and potential strategies for therapeutic intervention.


Assuntos
Ansiedade/etiologia , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Transtornos Cognitivos/etiologia , Transtornos dos Movimentos/etiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Corpo Caloso/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Fibras Nervosas Mielinizadas/patologia , Fragmentos de Peptídeos/metabolismo , Retenção Psicológica/fisiologia , Teste de Desempenho do Rota-Rod , Fatores de Tempo , Proteínas tau/metabolismo
7.
Mol Cell Neurosci ; 56: 255-62, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23831388

RESUMO

Emerging evidence suggests beta-amyloid (Aß) deposition in the Alzheimer's disease (AD) brain is the result of impaired clearance, due in part to diminished Aß transport across the blood-brain barrier (BBB). Recently, modulation of the cannabinoid system was shown to reduce Aß brain levels and improve cognitive behavior in AD animal models. The purpose of the current studies was to investigate the role of the cannabinoid system in the clearance of Aß across the BBB. Using in vitro and in vivo models of BBB clearance, Aß transit across the BBB was examined in the presence of cannabinoid receptor agonists and inhibitors. In addition, expression levels of the Aß transport protein, lipoprotein receptor-related protein1 (LRP1), were determined in the brain and plasma of mice following cannabinoid treatment. Cannabinoid receptor agonism or inhibition of endocannabinoid-degrading enzymes significantly enhanced Aß clearance across the BBB (2-fold). Moreover, cannabinoid receptor inhibition negated the stimulatory influence of cannabinoid treatment on Aß BBB clearance. Additionally, LRP1 levels in the brain and plasma were elevated following cannabinoid treatment (1.5-fold), providing rationale for the observed increase in Aß transit from the brain to the periphery. The current studies demonstrate, for the first time, a role for the cannabinoid system in the transit of Aß across the BBB. These findings provide insight into the mechanism by which cannabinoid treatment reduces Aß burden in the AD brain and offer additional evidence on the utility of this pathway as a treatment for AD.


Assuntos
Peptídeos beta-Amiloides/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Endotélio Vascular/metabolismo , Fragmentos de Peptídeos/farmacocinética , Animais , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transcitose/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
8.
Exp Neurol ; 374: 114702, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38301863

RESUMO

Repetitive mild traumatic brain injuries (r-mTBI) sustained in the military or contact sports have been associated with the accumulation of extracellular tau in the brain, which may contribute to the pathogenesis of neurodegenerative tauopathies. The expression of the apolipoprotein E4 (apoE4) isoform has been associated with higher levels of tau in the brain, and worse clinical outcomes after r-mTBI, though the influence of apoE genotype on extracellular tau dynamics in the brain is poorly understood. We recently demonstrated that extracellular tau can be eliminated across blood-brain barrier (BBB), which is progressively impaired following r-mTBI. The current studies investigated the influence of repetitive mild TBI (r-mTBI) and apoE genotype on the elimination of extracellular solutes from the brain. Following intracortical injection of biotin-labeled tau into humanized apoE-Tr mice, the levels of exogenous tau residing in the brain of apoE4 mice were elevated compared to other isoforms, indicating reduced tau elimination. Additionally, we found exposure to r-mTBI increased tau residence in apoE2 mice, similar to our observations in E2FAD animals. Each of these findings may be the result of diminished tau efflux via LRP1 at the BBB, as LRP1 inhibition significantly reduced tau uptake in endothelial cells and decreased tau transit across an in vitro model of the BBB (basolateral-to-apical). Notably, we showed that injury and apoE status, (particularly apoE4) resulted in chronic alterations in BBB integrity, pericyte coverage, and AQP4 polarization. These aberrations coincided with an atypical reactive astrocytic gene signature indicative of diminished CSF-ISF exchange. Our work found that CSF movement was reduced in the chronic phase following r-mTBI (>18 months post injury) across all apoE genotypes. In summary, we show that apoE genotype strongly influences cerebrovascular homeostasis, which can lead to age-dependent deficiencies in the elimination of toxic proteins from the brain, like tau, particularly in the aftermath of head trauma.


Assuntos
Apolipoproteína E4 , Concussão Encefálica , Camundongos , Animais , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Camundongos Transgênicos , Células Endoteliais/metabolismo , Encéfalo/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Concussão Encefálica/metabolismo
9.
J Neurooncol ; 111(2): 133-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23184143

RESUMO

The present study evaluates, both functionally and biochemically, brain tumor-induced alterations in brain capillary endothelial cells. Brain tumors were induced in Balb/c mice via intracranial injection of Lewis Lung carcinoma cells into the right hemisphere of the mouse brain using stereotaxic apparatus. Blood-brain barrier (BBB) permeability was assessed at various stages of tumor development, using both radiolabeled tracer permeability and magnetic resonance imaging with gadolinium diethylene-triamine-pentaacetate contrast enhancement (Gad-DTPA). The expression of the drug efflux transporter, P-glycoprotein (P-gp), in the BBB at various stages of tumor development was also evaluated by Western blot and immunohistochemistry. Median mouse survival following tumor cell injection was 17 days. The permeability of the BBB to (3)H-mannitol was similar in both brain hemispheres at 7 and 10 days post-injection. By day 15, there was a twofold increase in (3)H-mannitol permeability in the tumor bearing hemispheres compared to the non-tumor hemispheres. Examination of BBB permeability with Gad-DTPA contrast enhanced MRI indicated cerebral vascular permeability changes were confined to the tumor area. The permeability increase observed at the later stages of tumor development correlated with an increase in cerebral vascular volume suggesting angiogenesis within the tumor bearing hemisphere. Furthermore, the Gad-DPTA enhancement observed within the tumor area was significantly less than Gad-DPTA enhancement within the circumventricular organs not protected by the BBB. Expression of P-gp in both the tumor bearing and non-tumor bearing portions of the brain appeared similar at all time points examined. These studies suggest that although BBB integrity is altered within the tumor site at later stages of development, the BBB is still functional and limiting in terms of solute and drug permeability in and around the tumor.


Assuntos
Barreira Hematoaquosa/fisiopatologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/mortalidade , Permeabilidade Capilar/fisiologia , Carcinoma Pulmonar de Lewis/patologia , Modelos Animais de Doenças , Feminino , Lateralidade Funcional , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Ácido Pentético , Fatores de Tempo , Trítio/metabolismo
10.
Neurodegener Dis ; 11(1): 13-21, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22572854

RESUMO

BACKGROUND: While apolipoprotein E4 (apoE4) is highly correlated with the development of Alzheimer's disease (AD), its role in AD pathology and, in particular, beta-amyloid (Aß) removal from the brain, is not clearly defined. OBJECTIVE: To elucidate the influence of apoE on the clearance of Aß across the blood-brain barrier (BBB). METHODS: Aß(1-42) was intracerebrally administered to transgenic mice expressing human apoE isoforms and examined in the periphery. RESULTS: apoE3 and apoE4 mice had 5 times and 2 times, respectively, more Aß(1-42) appearing in the plasma than wild-type or apoE knockout mice, indicating an enhanced clearance of Aß from the brain to the periphery. In vitro, unbound basolateral apoE3 (i.e., not bound to Aß), and to a lesser extent unbound apoE4, at concentrations ≤10 nM facilitated basolateral-to-apical fluorescein-Aß(1-42) transcytosis across a BBB model, while apoE isoforms bound to Aß significantly disrupted Aß transcytosis. Additionally, following apical exposure to the BBB model, we found that apoE4 bound to Aß is able to penetrate the BBB more readily than apoE3 bound to Aß and does so via the RAGE (receptor for advanced glycation end products) transporter. CONCLUSION: These studies indicate a multifaceted, isoform-dependent role for apoE in the exchange of Aß across the BBB and may partially explain the association of apoE4 and Aß brain accumulation in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Barreira Hematoencefálica/fisiologia , Fragmentos de Peptídeos/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Animais , Apolipoproteína E4/farmacologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/farmacologia , Encéfalo/anatomia & histologia , Células Cultivadas , Células Endoteliais , Fibronectinas/administração & dosagem , Lateralidade Funcional , Proteína HMGB1/farmacologia , Humanos , Técnicas In Vitro , Injeções Intraventriculares , Camundongos , Camundongos Knockout , Microvasos/citologia , Fragmentos de Peptídeos/administração & dosagem , Transcitose/efeitos dos fármacos , Transcitose/genética
11.
Mol Ther Methods Clin Dev ; 29: 303-318, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37359418

RESUMO

Traumatic optic neuropathy (TON) is a condition in which acute injury to the optic nerve from direct or indirect trauma results in vision loss. The most common cause of TON is indirect injury to the optic nerve caused by concussive forces that are transmitted to the optic nerve. TON occurs in up to 5% of closed-head trauma patients and there is currently no known effective treatment. One potential treatment option for TON is ST266, a cell-free biological solution containing the secretome of amnion-derived multipotent progenitor (AMP) cells. We investigated the efficacy of intranasal ST266 in a mouse model of TON induced by blunt head trauma. Injured mice treated with a 10-day regimen of ST266 showed an improvement in spatial memory and learning, a significant preservation of retinal ganglion cells, and a decrease in neuropathological markers in the optic nerve, optic tract, and dorsal lateral geniculate nucleus. ST266 treatment effectively downregulated the NLRP3 inflammasome-mediated neuroinflammation pathway after blunt trauma. Overall, treatment with ST266 was shown to improve functional and pathological outcomes in a mouse model of TON, warranting future exploration of ST266 as a cell-free therapeutic candidate for testing in all optic neuropathies.

12.
J Neurosci ; 30(34): 11251-8, 2010 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-20739545

RESUMO

Alzheimer's disease (AD) is the most common form of dementia among the aging population and is characterized pathologically by the progressive intracerebral accumulation of beta-amyloid (Abeta) peptides and neurofibrillary tangles. The level of proangiogenic growth factors and inflammatory mediators with proangiogenic activity is known to be elevated in AD brains which has led to the supposition that the cerebrovasculature of AD patients is in a proangiogenic state. However, angiogenesis depends on the balance between proangiogenic and antiangiogenic factors and the brains of AD patients also show an accumulation of endostatin and Abeta peptides which have been shown to be antiangiogenic. To determine whether angiogenesis is compromised in the brains of two transgenic mouse models of AD overproducing Abeta peptides (Tg APPsw and Tg PS1/APPsw mice), we assessed the growth and vascularization of orthotopically implanted murine gliomas since they require a high degree of angiogenesis to sustain their growth. Our data reveal that intracranial tumor growth and angiogenesis is significantly reduced in Tg APPsw and Tg PS1/APPsw mice compared with their wild-type littermates. In addition, we show that Abeta inhibits the angiogenesis stimulated by glioma cells when cocultured with human brain microvascular cells on a Matrigel layer. Altogether our data suggest that the brain of transgenic mouse models of AD does not constitute a favorable environment to support neoangiogenesis and may explain why vascular insults synergistically precipitate the cognitive presentation of AD.


Assuntos
Doença de Alzheimer/patologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Glioma/patologia , Neovascularização Patológica/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/prevenção & controle , Animais , Neoplasias Encefálicas/prevenção & controle , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Cricetinae , Modelos Animais de Doenças , Glioma/genética , Glioma/prevenção & controle , Humanos , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias/métodos , Transplante de Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/prevenção & controle
13.
Mol Med ; 17(3-4): 149-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21170472

RESUMO

Several large population-based or clinical trial studies have suggested that certain dihydropyridine (DHP) L-type calcium channel blockers (CCBs) used for the treatment of hypertension may confer protection against the development of Alzheimer disease (AD). However, other studies with drugs of the same class have shown no beneficial clinical effects. To determine whether certain DHPs are able to impact underlying disease processes in AD (specifically the accumulation of the Alzheimer Aß peptide), we investigated the effect of several antihypertensive DHPs and non-DHP CCBs on Aß production. Among the antihypertensive DHPs tested, a few, including nilvadipine, nitrendipine and amlodipine inhibited Aß production in vitro, whereas others had no effect or raised Aß levels. In vivo, nilvadipine and nitrendipine acutely reduced brain Aß levels in a transgenic mouse model of AD (Tg PS1/APPsw) and improved Aß clearance across the blood-brain barrier (BBB), whereas amlodipine and nifedipine were ineffective showing that the Aß-lowering activity of the DHPs is independent of their antihypertensive activity. Chronic oral treatment with nilvadipine decreased Aß burden in the brains of Tg APPsw (Tg2576) and Tg PS1/APPsw mice, and also improved learning abilities and spatial memory. Our data suggest that the clinical benefit conferred by certain antihypertensive DHPs against AD is unrelated to their antihypertensive activity, but rely on their ability to lower brain Aß accumulation by affecting both Aß production and Aß clearance across the BBB.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anti-Hipertensivos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Di-Hidropiridinas/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Anlodipino/farmacologia , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Células CHO , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Nifedipino/análogos & derivados , Nifedipino/farmacologia , Nitrendipino/farmacologia
14.
Microcirculation ; 18(5): 373-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21418385

RESUMO

OBJECTIVE: To investigate the role of RAGE in the epitope-dependent effects of Aß antibodies used as a peripheral sink therapy in AD. METHODS: An in vitro model of the BBB was used to examine the effect of various Aß antibodies or Aß peptide fragments on Aß exchange across the BBB. RESULTS: An N-terminal Aß antibody significantly enhanced the basolateral-to-apical transcytosis of fluorescein-Aß(1-42) across the BBB model (41%), while no effect was apparent with a C-terminal Aß antibody. Interestingly, modulation of RAGE in the presence of a C-terminal Aß antibody resulted in a 65% increase in Aß clearance across the BBB model, suggesting the C-terminal antibody-Aß complex is susceptible to RAGE transport. Additionally, N-terminal peptide fragments of Aß attenuated the brain penetration of full length Aß in the BBB model, indicating the N-terminal region of Aß is required for brain uptake. CONCLUSIONS: Antibodies masking the N-terminal region of Aß increase Aß clearance across the BBB by preventing Aß from interacting with the RAGE transporter, whereas antibodies bound to the C-terminus of Aß are taken up by RAGE and, hence, do not influence the BBB clearance of Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais Murinos/farmacologia , Barreira Hematoencefálica/metabolismo , Epitopos/metabolismo , Modelos Biológicos , Transcitose/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais Murinos/imunologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/imunologia , Barreira Hematoencefálica/imunologia , Células Cultivadas , Epitopos/imunologia , Humanos , Estrutura Terciária de Proteína , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo
15.
Biopharm Drug Dispos ; 32(4): 233-44, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21446053

RESUMO

Venlafaxine and its metabolite desvenlafaxine are serotonin-norepinephrine reuptake inhibitors currently prescribed for the treatment of depression. Previously, it was reported that venlafaxine is an inducer of MDR1, the gene responsible for P-glycoprotein (P-gp). The present study expanded upon these findings by examining the effect of venlafaxine and desvenlafaxine on the expression of both P-gp and the breast cancer resistance protein (BCRP) in human brain endothelial cells (HBMEC), an in vitro model of the blood-brain barrier (BBB). The HBMEC were treated for 1 h with various concentrations (500 nM to 50 µM) of venlafaxine and desvenlafaxine. Western blot analysis revealed treatment with venlafaxine significantly induced the expression of P-gp (2-fold) and BCRP (1.75-fold) in a dose-dependent manner, while treatment with desvenlafaxine had no effect on drug efflux transporter expression. To determine the functional significance of this effect, the permeability of a known drug efflux probe, rhodamine 123, across the BBB model and Caco-2 cells, a model of intestinal absorption, were examined. Treatment with venlafaxine (1-50 µM) for 1 h significantly reduced the apical-to-basolateral permeability of R123 across the BBB model (30%) and Caco-2 cell monolayers (25%), indicative of increased drug efflux transporter expression at the apical membrane. Conversely, desvenlafaxine had no effect on R123 permeability in either cellular model. These studies indicate that venlafaxine, but not desvenlafaxine is an inducer of drug efflux transporter expression, which consequently increases the potential for clinical drug-drug interactions. Therefore, based on these preliminary results, caution should be taken when prescribing venlafaxine with other P-gp substrates.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Cicloexanóis/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Transporte Biológico , Encéfalo/metabolismo , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Cicloexanóis/toxicidade , Succinato de Desvenlafaxina , Relação Dose-Resposta a Droga , Interações Medicamentosas , Células Endoteliais , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/metabolismo , Humanos , Absorção Intestinal/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/efeitos dos fármacos , Rodamina 123/farmacocinética , Rifampina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Cloridrato de Venlafaxina
16.
Fluids Barriers CNS ; 18(1): 48, 2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702292

RESUMO

Repetitive head trauma has been associated with the accumulation of tau species in the brain. Our prior work showed brain vascular mural cells contribute to tau processing in the brain, and that these cells progressively degenerate following repetitive mild traumatic brain injury (r-mTBI). The current studies investigated the role of the cerebrovasculature in the elimination of extracellular tau from the brain, and the influence of r-mTBI on these processes. Following intracranial injection of biotin-labeled tau, the levels of exogenous labeled tau residing in the brain were elevated in a mouse model of r-mTBI at 12 months post-injury compared to r-sham mice, indicating reduced tau elimination from the brain following head trauma. This may be the result of decreased caveolin-1 mediated tau efflux at the blood-brain barrier (BBB), as the caveolin inhibitor, methyl-ß-cyclodextrin, significantly reduced tau uptake in isolated cerebrovessels and significantly decreased the basolateral-to-apical transit of tau across an in vitro model of the BBB. Moreover, we found that the upstream regulator of endothelial caveolin-1, Mfsd2a, was elevated in r-mTBI cerebrovessels compared to r-sham, which coincided with a decreased expression of cerebrovascular caveolin-1 in the chronic phase following r-mTBI (> 3 months post-injury). Lastly, angiopoietin-1, a mural cell-derived protein governing endothelial Mfsd2a expression, was secreted from r-mTBI cerebrovessels to a greater extent than r-sham animals. Altogether, in the chronic phase post-injury, release of angiopoietin-1 from degenerating mural cells downregulates caveolin-1 expression in brain endothelia, resulting in decreased tau elimination across the BBB, which may describe the accumulation of tau species in the brain following head trauma.


Assuntos
Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Proteínas tau/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
17.
Mol Brain ; 14(1): 110, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34238312

RESUMO

Cerebrovascular dysfunction is a hallmark feature of Alzheimer's disease (AD). One of the greatest risk factors for AD is the apolipoprotein E4 (E4) allele. The APOE4 genotype has been shown to negatively impact vascular amyloid clearance, however, its direct influence on the molecular integrity of the cerebrovasculature compared to other APOE variants (APOE2 and APOE3) has been largely unexplored. To address this, we employed a 10-plex tandem isobaric mass tag approach in combination with an ultra-high pressure liquid chromatography MS/MS (Q-Exactive) method, to interrogate unbiased proteomic changes in cerebrovessels from AD and healthy control brains with different APOE genotypes. We first interrogated changes between healthy control cases to identify underlying genotype specific effects in cerebrovessels. EIF2 signaling, regulation of eIF4 and 70S6K signaling and mTOR signaling were the top significantly altered pathways in E4/E4 compared to E3/E3 cases. Oxidative phosphorylation, EIF2 signaling and mitochondrial dysfunction were the top significant pathways in E2E2 vs E3/E3cases. We also identified AD-dependent changes and their interactions with APOE genotype and found the highest number of significant proteins from this interaction was observed in the E3/E4 (192) and E4/E4 (189) cases. As above, EIF2, mTOR signaling and eIF4 and 70S6K signaling were the top three significantly altered pathways in E4 allele carriers (i.e. E3/E4 and E4/E4 genotypes). Of all the cerebrovascular cell-type specific markers identified in our proteomic analyses, endothelial cell, astrocyte, and smooth muscle cell specific protein markers were significantly altered in E3/E4 cases, while endothelial cells and astrocyte specific protein markers were altered in E4/E4 cases. These proteomic changes provide novel insights into the longstanding link between APOE4 and cerebrovascular dysfunction, implicating a role for impaired autophagy, ER stress, and mitochondrial bioenergetics. These APOE4 dependent changes we identified could provide novel cerebrovascular targets for developing disease modifying strategies to mitigate the effects of APOE4 genotype on AD pathogenesis.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/genética , Apolipoproteínas E/genética , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Demência/genética , Predisposição Genética para Doença , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tamanho do Órgão , Proteoma/metabolismo , Proteômica , Frações Subcelulares/metabolismo
18.
Front Aging Neurosci ; 13: 658605, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34079449

RESUMO

Cerebrovascular dysfunction and cerebral amyloid angiopathy (CAA) are hallmark features of Alzheimer's disease (AD). Molecular damage to cerebrovessels in AD may result in alterations in vascular clearance mechanisms leading to amyloid deposition around blood vessels and diminished neurovascular-coupling. The sequelae of molecular events leading to these early pathogenic changes remains elusive. To address this, we conducted a comprehensive in-depth molecular characterization of the proteomic changes in enriched cerebrovessel fractions isolated from the inferior frontal gyrus of autopsy AD cases with low (85.5 ± 2.9 yrs) vs. high (81 ± 4.4 yrs) CAA score, aged-matched control (87.4 ± 1.5 yrs) and young healthy control (47 ± 3.3 yrs) cases. We employed a 10-plex tandem isobaric mass tag approach in combination with our ultra-high pressure liquid chromatography MS/MS (Q-Exactive) method. Enriched cerebrovascular fractions showed very high expression levels of proteins specific to endothelial cells, mural cells (pericytes and smooth muscle cells), and astrocytes. We observed 150 significantly regulated proteins in young vs. aged control cerebrovessels. The top pathways significantly modulated with aging included chemokine, reelin, HIF1α and synaptogenesis signaling pathways. There were 213 proteins significantly regulated in aged-matched control vs. high CAA cerebrovessels. The top three pathways significantly altered from this comparison were oxidative phosphorylation, Sirtuin signaling pathway and TCA cycle II. Comparison between low vs. high CAA cerebrovessels identified 84 significantly regulated proteins. Top three pathways significantly altered between low vs. high CAA cerebrovessels included TCA Cycle II, Oxidative phosphorylation and mitochondrial dysfunction. Notably, high CAA cases included more advanced AD pathology thus cerebrovascular effects may be driven by the severity of amyloid and Tangle pathology. These descriptive proteomic changes provide novel insights to explain the age-related and AD-related cerebrovascular changes contributing to AD pathogenesis. Particularly, disturbances in energy bioenergetics and mitochondrial biology rank among the top AD pathways altered in cerebrovessels. Targeting these failed mechanisms in endothelia and mural cells may provide novel disease modifying targets for developing therapeutic strategies against cerebrovascular deterioration and promoting cerebral perfusion in AD. Our future work will focus on interrogating and validating these novel targets and pathways and their functional significance.

19.
J Cereb Blood Flow Metab ; 41(6): 1362-1378, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33050825

RESUMO

Incidences of repetitive mild TBI (r-mTBI), like those sustained by contact sports athletes and military personnel, are thought to be a risk factor for development of neurodegenerative disorders. Those suffering from chronic TBI-related illness demonstrate deficits in cerebrovascular reactivity (CVR), the ability of the cerebral vasculature to respond to a vasoactive stimulus. CVR is thus an important measure of traumatic cerebral vascular injury (TCVI), and a possible in vivo endophenotype of TBI-related neuropathogenesis. We combined laser speckle imaging of CVR in response to hypercapnic challenge with neurobehavioral assessment of learning and memory, to investigate if decreased cerebrovascular responsiveness underlies impaired cognitive function in our mouse model of chronic r-mTBI. We demonstrate a profile of blunted hypercapnia-evoked CVR in the cortices of r-mTBI mice like that of human TBI, alongside sustained memory and learning impairment, without biochemical or immunohistopathological signs of cerebral vessel laminar or endothelium constituent loss. Transient decreased expression of alpha smooth muscle actin and platelet-derived growth factor receptor ß, indicative of TCVI, is obvious only at the time of the most pronounced CVR deficit. These findings implicate CVR as a valid preclinical measure of TCVI, perhaps useful for developing therapies targeting TCVI after recurrent mild head trauma.


Assuntos
Concussão Encefálica/fisiopatologia , Circulação Cerebrovascular/fisiologia , Hipercapnia/fisiopatologia , Animais , Modelos Animais de Doenças , Hipercapnia/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
J Neurochem ; 112(1): 66-76, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19818105

RESUMO

Beta-amyloid peptides (Abeta) are the major constituents of senile plaques and cerebrovascular deposits in the brains of Alzheimer's disease patients. We have shown previously that soluble forms of Abeta are anti-angiogenic both in vitro and in vivo. However, the mechanism of the anti-angiogenic activity of Abeta peptides is unclear. In this study, we examined the effects of Abeta1-42 on vascular endothelial growth factor receptor 2 (VEGFR-2) signaling, which plays a key role in angiogenesis. Abeta inhibited VEGF-induced migration of endothelial cells, as well as VEGF-induced permeability of an in vitro model of the blood brain barrier. Consistently, exogenous VEGF dose-dependently antagonized the anti-angiogenic activity of Abeta in a capillary network assay. Abeta1-42 also blocked VEGF-induced tyrosine phosphorylation of VEGFR-2 in two types of primary endothelial cells, suggesting an antagonistic action of Abeta toward VEGFR-2 signaling in cells. Moreover, Abeta was able to directly interact with the extracellular domain of VEGFR-2 and to compete with the binding of VEGF to its receptor in a cell-free assay. Co-immunoprecipitation experiments confirmed that Abeta can bind VEGFR-2 both in vitro and in vivo. Altogether, our data suggest that Abeta acts as an antagonist of VEGFR-2 and provide a mechanism explaining the anti-angiogenic activity of Abeta peptides.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/fisiologia , Fragmentos de Peptídeos/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Inibidores da Angiogênese/antagonistas & inibidores , Inibidores da Angiogênese/fisiologia , Animais , Movimento Celular/fisiologia , Células Cultivadas , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA