Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Infect Immun ; 86(11)2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30181350

RESUMO

Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.


Assuntos
Microambiente Celular , Interações Hospedeiro-Patógeno , Mucosa Intestinal/fisiologia , Técnicas de Cultura de Órgãos/métodos , Organoides , Engenharia Tecidual/métodos , História do Século XX , História do Século XXI , Humanos , Modelos Biológicos , Técnicas de Cultura de Órgãos/história , Engenharia Tecidual/história
2.
Biochem Biophys Res Commun ; 472(3): 545-50, 2016 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-26970308

RESUMO

The inflammasome is a caspase-1-activating complex that is implicated in a growing number of acute and chronic pathologies. Interest has increased in identifying small molecular inhibitors of inflammasome signaling because of its role in clinically relevant diseases. It was recently reported that the protein tyrosine kinase, Syk, regulates pathogen-induced inflammasome signaling by phosphorylating a molecular switch on the adapter protein ASC. However, several aspects of the role of Syk in inflammasome signaling and the effects of its inhibition remain unclear. The aim of the present study is to explore in detail the effects of the oxindole Syk inhibitor OXSI-2 on various aspects of nigericin-induced inflammasome signaling. Our results indicate that OXSI-2 inhibits inflammasome assembly, caspase-1 activation, IL-1ß processing and release, mitochondrial ROS generation, and pyroptotic cell death. Using a novel live cell potassium sensor we show that Syk inhibition with OXSI-2 has no effect on potassium efflux kinetics and that blockade of potassium efflux with extracellular potassium alters Syk phosphorylation. The effects of OXSI-2 identified in this study provide context for the role of Syk in inflammasome signaling and demonstrate its importance in oxidative signaling upstream of inflammasome activation and downstream of ion flux.


Assuntos
Indóis/administração & dosagem , Inflamassomos/metabolismo , Potássio/metabolismo , Piroptose/fisiologia , Transdução de Sinais/fisiologia , Sulfonamidas/administração & dosagem , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Interações Medicamentosas , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Macrófagos/fisiologia , Taxa de Depuração Metabólica/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nigericina/administração & dosagem , Oxindóis , Proteínas Tirosina Quinases , Piroptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinase Syk
3.
Neurobiol Dis ; 54: 24-31, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23473743

RESUMO

Increased brain infiltration of polymorphonuclear neutrophils (PMNs) occurs early after stroke and is important in eliciting brain inflammatory response during stroke recovery. In order to understand the molecular mechanism of PMN entry, we investigated the expression and requirement for Slit1, a chemorepulsive guidance cue, and its cognate receptor, Robo1, in a long-term recovery mouse model of cerebral ischemia. The expression levels of Robo1 were significantly decreased bilaterally at 24h following reperfusion. Robo1 expression levels remained suppressed in the ipsilateral cortex until 28d post MCAO-reperfusion, while the levels of Robo1 in the contralateral cortex recovered to the level of sham-operated mouse by 7d reperfusion. Circulating PMNs express high levels of Slit1, but not Robo1. Influx of PMNs into the ischemic core area occurred early (24h) after cerebral ischemia, when endothelial Robo1 expression was significantly reduced in the ischemic brain, indicating that Robo1 may form a repulsive barrier to PMN entry into the brain parenchyma. Indeed, blocking Slit1 on PMNs in a transwell migration assay in combination with an antibody blocking of Robo1 on human umbilical vein endothelial cells (HUVEC) significantly increased PMN transmigration during oxygen glucose deprivation, an in vitro model of ischemia. Collectively, in the normal brain, the presence of Slit1 on PMNs, and Robo1 on cerebral endothelial cells, generated a repulsive force to prevent the infiltration of PMNs into the brain. During stroke recovery, a transient reduction in Robo1 expression on the cerebral endothelial cells allowed the uncontrolled infiltration of Slit1-expressing PMNs into the brain causing inflammatory reactions.


Assuntos
Encéfalo/metabolismo , Quimiotaxia de Leucócito/fisiologia , Células Endoteliais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neutrófilos/metabolismo , Receptores Imunológicos/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Western Blotting , Encéfalo/imunologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Acidente Vascular Cerebral/imunologia , Transfecção , Proteínas Roundabout
4.
mSphere ; 7(4): e0021022, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35913142

RESUMO

The discovery that biomechanical forces regulate microbial virulence was established with the finding that physiological low fluid shear (LFS) forces altered gene expression, stress responses, and virulence of the enteric pathogen Salmonella enterica serovar Typhimurium during the log phase. These log phase LFS-induced phenotypes were independent of the master stress response regulator, RpoS (σS). Given the central importance of RpoS in regulating stationary-phase stress responses of S. Typhimurium cultured under conventional shake flask and static conditions, we examined its role in stationary-phase cultures grown under physiological LFS. We constructed an isogenic rpoS mutant derivative of wild-type S. Typhimurium and compared the ability of these strains to survive in vitro pathogenesis-related stresses that mimic those encountered in the infected host and environment. We also compared the ability of these strains to colonize (adhere, invade, and survive within) human intestinal epithelial cell cultures. Unexpectedly, LFS-induced resistance of stationary-phase S. Typhimurium cultures to acid and bile salts stresses did not rely on RpoS. Likewise, RpoS was dispensable for stationary-phase LFS cultures to adhere to and survive within intestinal epithelial cells. In contrast, the resistance of these cultures to challenges of oxidative and thermal stresses, and their invasion into intestinal epithelial cells was influenced by RpoS. These findings expand our mechanistic understanding of how physiological fluid shear forces modulate stationary-phase S. Typhimurium physiology in unexpected ways and provide clues into microbial mechanobiology and nuances of Salmonella responses to microenvironmental niches in the infected host. IMPORTANCE Bacterial pathogens respond dynamically to a variety of stresses in the infected host, including physical forces of fluid flow (fluid shear) across their surfaces. While pathogens experience wide fluctuations in fluid shear during infection, little is known about how these forces regulate microbial pathogenesis. This is especially important for stationary-phase bacterial growth, which is a critical period to understand microbial resistance, survival, and infection potential, and is regulated in many bacteria by the general stationary-phase stress response protein RpoS. Here, we showed that, unlike conventional culture conditions, several stationary-phase Salmonella pathogenic stress responses were not impacted by RpoS when bacteria were cultured under fluid shear conditions relevant to those encountered in the intestine of the infected host. These findings offer new insight into how physiological fluid shear forces encountered by Salmonella during infection might impact pathogenic responses in unexpected ways that are relevant to their disease-causing ability.


Assuntos
Salmonella typhimurium , Fator sigma , Ácidos/metabolismo , Proteínas de Bactérias/metabolismo , Humanos , Salmonella typhimurium/metabolismo , Fator sigma/genética , Fator sigma/metabolismo , Virulência/genética
5.
Front Cell Infect Microbiol ; 12: 705647, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35711662

RESUMO

Physical forces associated with spaceflight and spaceflight analogue culture regulate a wide range of physiological responses by both bacterial and mammalian cells that can impact infection. However, our mechanistic understanding of how these environments regulate host-pathogen interactions in humans is poorly understood. Using a spaceflight analogue low fluid shear culture system, we investigated the effect of Low Shear Modeled Microgravity (LSMMG) culture on the colonization of Salmonella Typhimurium in a 3-D biomimetic model of human colonic epithelium containing macrophages. RNA-seq profiling of stationary phase wild type and Δhfq mutant bacteria alone indicated that LSMMG culture induced global changes in gene expression in both strains and that the RNA binding protein Hfq played a significant role in regulating the transcriptional response of the pathogen to LSMMG culture. However, a core set of genes important for adhesion, invasion, and motility were commonly induced in both strains. LSMMG culture enhanced the colonization (adherence, invasion and intracellular survival) of Salmonella in this advanced model of intestinal epithelium using a mechanism that was independent of Hfq. Although S. Typhimurium Δhfq mutants are normally defective for invasion when grown as conventional shaking cultures, LSMMG conditions unexpectedly enabled high levels of colonization by an isogenic Δhfq mutant. In response to infection with either the wild type or mutant, host cells upregulated transcripts involved in inflammation, tissue remodeling, and wound healing during intracellular survival. Interestingly, infection by the Δhfq mutant led to fewer transcriptional differences between LSMMG- and control-infected host cells relative to infection with the wild type strain. This is the first study to investigate the effect of LSMMG culture on the interaction between S. Typhimurium and a 3-D model of human intestinal tissue. These findings advance our understanding of how physical forces can impact the early stages of human enteric salmonellosis.


Assuntos
Biomimética , Voo Espacial , Animais , Técnicas de Cocultura , Interações Hospedeiro-Patógeno , Humanos , Mamíferos , Salmonella typhimurium/genética
6.
Neurobiol Dis ; 33(3): 405-14, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19118631

RESUMO

In recent years, GDNF has emerged as a protective and restorative agent in several models of neurodegeneration; however, the exact molecular mechanisms responsible for these effects are not yet fully understood. Here we examined the effects of astrocytes secreting GDNF on neurons subjected to 6OHDA toxicity using in vitro neuron-astroglia co-cultures. Astrocytes were transduced with lentiviral vectors carrying the GDNF gene under the control of either human glial fibrillary acidic protein or cytomegalovirus promoters. The overexpression of GDNF, regardless of the promoter employed, had no obvious adverse effects on astroglia and the engineered cells stably produced and secreted GDNF for extended periods of time (> or =3 weeks). These astrocytes very effectively protected neurons against 6OHDA, in both mouse and human co-culture systems. The neuroprotective effects were mediated not only by GDNF, but also by the antioxidant GSH since its depletion reduced the level of GDNF protection. Furthermore, neurons and astrocytes expressed different components of GDNF signaling complex, suggesting that they might utilize separate pathways to mediate autocrine and paracrine effects of GDNF.


Assuntos
Astrócitos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Glutationa/metabolismo , Neurônios/fisiologia , Oxidopamina/toxicidade , Análise de Variância , Animais , Antioxidantes/metabolismo , Western Blotting , Butionina Sulfoximina/administração & dosagem , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Expressão Gênica , Glutationa/antagonistas & inibidores , Humanos , Imuno-Histoquímica , Camundongos , Neurônios/efeitos dos fármacos , Reação em Cadeia da Polimerase , Transdução de Sinais/fisiologia , Transfecção
7.
Sci Adv ; 3(12): e1602580, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29226240

RESUMO

Quantitative three-dimensional (3D) computed tomography (CT) imaging of living single cells enables orientation-independent morphometric analysis of the intricacies of cellular physiology. Since its invention, x-ray CT has become indispensable in the clinic for diagnostic and prognostic purposes due to its quantitative absorption-based imaging in true 3D that allows objects of interest to be viewed and measured from any orientation. However, x-ray CT has not been useful at the level of single cells because there is insufficient contrast to form an image. Recently, optical CT has been developed successfully for fixed cells, but this technology called Cell-CT is incompatible with live-cell imaging due to the use of stains, such as hematoxylin, that are not compatible with cell viability. We present a novel development of optical CT for quantitative, multispectral functional 4D (three spatial + one spectral dimension) imaging of living single cells. The method applied to immune system cells offers truly isotropic 3D spatial resolution and enables time-resolved imaging studies of cells suspended in aqueous medium. Using live-cell optical CT, we found a heterogeneous response to mitochondrial fission inhibition in mouse macrophages and differential basal remodeling of small (0.1 to 1 fl) and large (1 to 20 fl) nuclear and mitochondrial structures on a 20- to 30-s time scale in human myelogenous leukemia cells. Because of its robust 3D measurement capabilities, live-cell optical CT represents a powerful new tool in the biomedical research field.


Assuntos
Tomografia Óptica/instrumentação , Tomografia Óptica/métodos , Núcleo Celular/metabolismo , Desenho de Equipamento , Tomografia Computadorizada Quadridimensional/instrumentação , Tomografia Computadorizada Quadridimensional/métodos , Humanos , Células K562/patologia , Mitocôndrias/metabolismo , Reprodutibilidade dos Testes , Análise de Célula Única
8.
Sci Rep ; 5: 7890, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25601765

RESUMO

Semaphorin 3A (Sema3A) increased significantly in mouse brain following cerebral ischemia. However, the role of Sema3A in stroke brain remains unknown. Our aim was to determine wether Sema3A functions as a vascular permeability factor and contributes to ischemic brain damage. Recombinant Sema3A injected intradermally to mouse skin, or stereotactically into the cerebral cortex, caused dose- and time-dependent increases in vascular permeability, with a degree comparable to that caused by injection of a known vascular permeability factor vascular endothelial growth factor receptors (VEGF). Application of Sema3A to cultured endothelial cells caused disorganization of F-actin stress fibre bundles and increased endothelial monolayer permeability, confirming Sema3A as a permeability factor. Sema3A-mediated F-actin changes in endothelial cells were through binding to the neuropilin2/VEGFR1 receptor complex, which in turn directly activates Mical2, a F-actin modulator. Down-regulation of Mical2, using specific siRNA, alleviated Sema3A-induced F-actin disorganization, cellular morphology changes and endothelial permeability. Importantly, ablation of Sema3A expression, cerebrovascular permeability and brain damage were significantly reduced in response to transient middle cerebral artery occlusion (tMCAO) and in a mouse model of cerebral ischemia/haemorrhagic transformation. Together, these studies demonstrated that Sema3A is a key mediator of cerebrovascular permeability and contributes to brain damage caused by cerebral ischemia.


Assuntos
Lesões Encefálicas/genética , Permeabilidade Capilar/genética , Semaforina-3A/genética , Acidente Vascular Cerebral/genética , Actinas/genética , Actinas/metabolismo , Animais , Apoptose/genética , Lesões Encefálicas/fisiopatologia , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatologia , Permeabilidade Capilar/fisiologia , Córtex Cerebral/lesões , Córtex Cerebral/patologia , Proteínas do Citoesqueleto/biossíntese , Modelos Animais de Doenças , Humanos , Camundongos , Proteínas dos Microfilamentos , Neuropilina-2/metabolismo , Semaforina-3A/administração & dosagem , Acidente Vascular Cerebral/fisiopatologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Neurobiol Aging ; 35(10): 2329-46, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24775711

RESUMO

Although the support for the use of antioxidants, such as coenzyme Q(10) (CoQ(10)), to treat Parkinson's disease (PD) comes from the extensive scientific evidence, the results of conducted thus far clinical trials are inconclusive. It is assumed that the efficacy of CoQ(10) is hindered by insolubility, poor bioavailability, and lack of brain penetration. We have developed a nanomicellar formulation of CoQ(10) (Ubisol-Q(10)) with improved properties, including the brain penetration, and tested its effectiveness in mouse MPTP (1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine) model with the objectives to assess its potential use as an adjuvant therapy for PD. We used a subchronic MPTP model (5-daily MPTP injections), characterized by 50% loss of dopamine neurons over a period of 28 days. Ubisol-Q(10) was delivered in drinking water. Prophylactic application of Ubisol-Q(10), started 2 weeks before the MPTP exposure, significantly offset the neurotoxicity (approximately 50% neurons died in MPTP group vs. 17% in MPTP+ Ubisol-Q(10) group by day 28). Therapeutic application of Ubisol-Q(10), given after the last MPTP injection, was equally effective. At the time of intervention on day 5 nearly 25% of dopamine neurons were already lost, but the treatment saved the remaining 25% of cells, which otherwise would have died by day 28. This was confirmed by cell counts, analyses of striatal dopamine levels, and improved animals' motor skill on a beam walk test. Similar levels of neuroprotection were obtained with 3 different Ubisol-Q(10) concentrations tested, that is, 30 mg, 6 mg, or 3 mg CoQ(10)/kg body weight/day, showing clearly that high doses of CoQ(10) were not required to deliver these effects. Furthermore, the Ubisol-Q(10) treatments brought about a robust astrocytic activation in the brain parenchyma, indicating that astroglia played an active role in this neuroprotection. Thus, we have shown for the first time that Ubisol-Q(10) was capable of halting the neurodegeneration already in progress; however, to maintain it a continuous supplementation of Ubisol-Q(10) was required. The pathologic processes initiated by MPTP resumed if supplementation was withdrawn. We suggest that in addition to brain delivery of powerful antioxidants, Ubisol-Q(10) might have also supported subcellular oxidoreductase systems allowing them to maintain a favorable cellular redox status, especially in astroglia, facilitating their role in neuroprotection. Based on this data further clinical testing of this formulation in PD patients might be justifiable.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Antioxidantes/uso terapêutico , Micelas , Nanopartículas , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Ubiquinona/análogos & derivados , Animais , Antioxidantes/administração & dosagem , Astrócitos/metabolismo , Química Farmacêutica , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Masculino , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/administração & dosagem , Oxirredução/efeitos dos fármacos , Doença de Parkinson/patologia , Ubiquinona/administração & dosagem , Ubiquinona/uso terapêutico
10.
Biosci Trends ; 6(4): 183-91, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23006965

RESUMO

Membrane rafts, rich in sphingolipids and cholesterol, play an important role in neuronal membrane domain-specific signaling events, maintaining synapses and dendritic spines. The purpose of this study is to examine the neuronal response to membrane raft disruption. Membrane rafts of 8 DIV primary neuronal cultures were isolated based on the resistance to Triton X-100 and ability to float in sucrose gradients. Membrane rafts from primary cortical neurons were also imaged using the membrane raft marker, cholera toxin subunit-B (CTxB), and were co-immunolabelled with the dendritic microtubule associated protein marker, MAP-2, the dendritic and axonal microtubule protein, ß-III-Tubulin, and the axonal microtubule protein, Tau. Exposure of cortical neurons to either the cholesterol depleting compound, methyl-beta-cyclodextrin (MBC), or to the glycosphingolipid metabolism inhibiting agent D-threo-1-phenyl-2-decanoylamino-3- morpholino-1-propanol (D-PDMP), resulted in neuritic retraction prior to the appearance of neuronal death. Further investigation into the effects of MBC revealed a pronounced perturbation of microtubule protein association with membrane rafts during neuritic retraction. Interestingly, stabilizing microtubules with Paclitaxel did not prevent MBC induced neuritic retraction, suggesting that neuritic retraction occurred independently of microtubule disassembly and that microtubule association with membrane rafts is critical for maintaining neuritic stability. Overall, the data indicated that membrane rafts play an important role in neurite stability and neuronal viability.


Assuntos
Córtex Cerebral/patologia , Microdomínios da Membrana/metabolismo , Neuritos/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Morfolinas/farmacologia , Neuritos/efeitos dos fármacos , Imagem com Lapso de Tempo , beta-Ciclodextrinas/farmacologia
11.
PLoS One ; 6(6): e20808, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21687673

RESUMO

Gangliosides, a member of the glycosphingolipid family, are heterogeneously expressed in biological membranes and are particularly enriched within the central nervous system. Gangliosides consist of mono- or poly-sialylated oligosaccharide chains of variable lengths attached to a ceramide unit and are found to be intimately involved in brain disease development. The purpose of this study is to examine the spatial profile of ganglioside species using matrix-assisted laser desorption/ionization (MALDI) imaging (IMS) following middle cerebral artery occlusion (MCAO) reperfusion injury in the mouse. IMS is a powerful method to not only discriminate gangliosides by their oligosaccharide components, but also by their carbon length within their sphingosine base. Mice were subjected to a 30 min unilateral MCAO followed by long-term survival (up to 28 days of reperfusion). Brain sections were sprayed with the matrix 5-Chloro-2-mercaptobenzothiazole, scanned and analyzed for a series of ganglioside molecules using an Applied Biosystems 4800 MALDI TOF/TOF. Traditional histological and immunofluorescence techniques were performed to assess brain tissue damage and verification of the expression of gangliosides of interest. Results revealed a unique anatomical profile of GM1, GD1 and GT1b (d18:1, d20:1 as well as other members of the glycosphingolipid family). There was marked variability in the ratio of expression between ipsilateral and contralateral cortices for the various detected ganglioside species following MCAO-reperfusion injury. Most interestingly, MCAO resulted in the transient induction of both GM2 and GM3 signals within the ipsilateral hemisphere; at the border of the infarcted tissue. Taken together, the data suggest that brain region specific expression of gangliosides, particularly with respect to hydrocarbon length, may play a role in neuronal responses to injury.


Assuntos
Encéfalo/metabolismo , Gangliosídeos/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Ataque Isquêmico Transitório/metabolismo , Imagem Molecular , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Cérebro/metabolismo , Cérebro/patologia , Cérebro/fisiopatologia , Gangliosídeos/química , Regulação da Expressão Gênica , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/patologia , Especificidade de Órgãos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Esfingosina/química , Fatores de Tempo
12.
Brain Res ; 1344: 209-16, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20493826

RESUMO

Membrane rafts, rich in sphingolipids and cholesterol, are membrane microdomains important in neuronal domain-specific signaling events such as during axonal outgrowth and neuronal death. The present study seeks to determine the spatiotemporal association of several axonal guidance signaling molecules with membrane rafts. These molecules are Neuropilin-1 (NRP-1), Fer Kinase, and collapsin response mediator proteins (CRMPs), which are known to have important functions in axonal outgrowth and neuronal death caused by cerebral ischemia. Mice were subjected to sham or a 1h unilateral middle cerebral artery occlusion (MCAO) followed by a time course of reperfusion up to 24h. Brain cortices were separated and membrane rafts were extracted based on their insolubility in Triton X-100 and separation by sucrose gradient fractionation. We demonstrate the early and transient induction of NRP-1 and CRMP-2 in membrane rafts in both ipsilateral and contralateral hemispheres, in contrast to an early, but sustained elevation of Fer kinase and other CRMPs (1, 3, 4, 5) in response to unilateral MCAO. The fact that NRP1/Fer kinase/CRMP-2 co-localize in membrane rafts early during ischemic injury suggests that the membrane rafts may form a scaffold to support and initiate NRP1/Fer/CRMP-2-mediated signal transduction in neuronal damage response during ischemia-reperfusion. Further understanding of the time-specific and membrane domain-specific protein-protein interaction may lead to the identification of therapeutic targets for stroke.


Assuntos
Membrana Celular/metabolismo , Lateralidade Funcional/fisiologia , Regulação da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/patologia , Neuropilina-1/metabolismo , Fosfotransferases/metabolismo , Semaforina-3A/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Fosfotransferases/classificação , Reperfusão/métodos , Fatores de Tempo
13.
Biochem Cell Biol ; 86(5): 361-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18923537

RESUMO

Every year thousands of people suffer from brain injuries and stroke, and develop motor, sensory, and cognitive problems as a result of neuronal loss in the brain. Unfortunately, the damaged brain has a limited ability to enact repair and current modes of treatment are not sufficient to offset the damage. An extensive list of growth factors, neurotrophic factors, cytokines, and drugs has been explored as potential therapies. However, only a limited number of them may actually have the potential to effectively offset the brain injury or stroke-related problems. One of the treatments considered for future brain repair is bone morphogenetic protein 7 (BMP7), a factor currently used in patients to treat non-neurological diseases. The clinical application of BMP7 is based on its neuroprotective role in stroke animal models. This paper reviews the current approaches considered for brain repair and discusses the novel convergent strategies by which BMP7 potentially can induce neuroregeneration.


Assuntos
Proteína Morfogenética Óssea 7/uso terapêutico , Encéfalo , Regeneração Nervosa/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Animais , Proteína Morfogenética Óssea 7/metabolismo , Encéfalo/patologia , Encéfalo/fisiologia , Diferenciação Celular , Sobrevivência Celular , Transplante de Células , Humanos , Neurogênese/fisiologia , Fármacos Neuroprotetores/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Engenharia Tecidual
14.
J Biol Chem ; 280(26): 25060-70, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15878861

RESUMO

Mitochondrial fusion in higher eukaryotes requires at least two essential GTPases, Mitofusin 1 and Mitofusin 2 (Mfn2). We have created an activated mutant of Mfn2, which shows increased rates of nucleotide exchange and decreased rates of hydrolysis relative to wild type Mfn2. Mitochondrial fusion is stimulated dramatically within heterokaryons expressing this mutant, demonstrating that hydrolysis is not requisite for the fusion event, and supporting a role for Mfn2 as a signaling GTPase. Although steady-state mitochondrial fusion required the conserved intermembrane space tryptophan residue, this requirement was overcome within the context of the hydrolysis-deficient mutant. Furthermore, the punctate localization of Mfn2 is lost in the dominant active mutants, indicating that these sites are functionally controlled by changes in the nucleotide state of Mfn2. Upon staurosporine-stimulated cell death, activated Bax is recruited to the Mfn2-containing puncta; however, Bax activation and cytochrome c release are inhibited in the presence of the dominant active mutants of Mfn2. The dominant active form of Mfn2 also protected the mitochondria against free radical-induced permeability transition. In contrast to staurosporine-induced outer membrane permeability transition, pore opening induced through the introduction of free radicals was dependent upon the conserved intermembrane space residue. This is the first evidence that Mfn2 is a signaling GTPase regulating mitochondrial fusion and that the nucleotide-dependent activation of Mfn2 concomitantly protects the organelle from permeability transition. The data provide new insights into the critical relationship between mitochondrial membrane dynamics and programmed cell death.


Assuntos
Proteínas de Membrana/química , Mitocôndrias/metabolismo , Proteínas Mitocondriais/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Apoptose , Sítios de Ligação , Células COS , Citocromos c/metabolismo , DNA Complementar/metabolismo , Corantes Fluorescentes/farmacologia , Radicais Livres , GTP Fosfo-Hidrolases/metabolismo , Genes Dominantes , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Hidrólise , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Microscopia de Fluorescência , Proteínas Mitocondriais/metabolismo , Mutação , Ligação Proteica , Estaurosporina/farmacologia , Fatores de Tempo , Transfecção , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA