Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Metab Brain Dis ; 30(2): 411-26, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24771110

RESUMEN

Traumatic brain injury (TBI) is accompanied with enhanced matrix metalloproteinase-9 (MMP-9) activity and elevated levels of plasma fibrinogen (Fg), which is a known inflammatory agent. Activation of MMP-9 and increase in blood content of Fg (i.e. hyperfibrinogenemia, HFg) both contribute to cerebrovascular disorders leading to blood brain barrier disruption. It is well-known that activation of MMP-9 contributes to vascular permeability. It has been shown that at an elevated level (i.e. HFg) Fg disrupts blood brain barrier. However, mechanisms of their actions during TBI are not known. Mild TBI was induced in wild type (WT, C57BL/6 J) and MMP-9 gene knockout (Mmp9(-/-)) homozygous, mice. Pial venular permeability to fluorescein isothiocyanate-conjugated bovine serum albumin in pericontusional area was observed 14 days after injury. Mice memory was tested with a novel object recognition test. Increased expression of Fg endothelial receptor intercellular adhesion protein-1 and formation of caveolae were associated with enhanced activity of MMP-9 causing an increase in pial venular permeability. As a result, an enhanced deposition of Fg and cellular prion protein (PrP(C)) were found in pericontusional area. These changes were attenuated in Mmp9(-/-) mice and were associated with lesser loss of short-term memory in these mice than in WT mice. Our data suggest that mild TBI-induced increased cerebrovascular permeability enhances deposition of Fg-PrP(C) and loss of memory, which is ameliorated in the absence of MMP-9 activity. Thus, targeting MMP-9 activity and blood level of Fg can be a possible therapeutic remedy to diminish vasculo-neuronal damage after TBI.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/metabolismo , Circulación Cerebrovascular/genética , Fibrinógeno/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Animales , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/psicología , Capilares/patología , Corteza Cerebral/lesiones , Venas Cerebrales/metabolismo , Contusiones/genética , Contusiones/metabolismo , Contusiones/psicología , Molécula 1 de Adhesión Intercelular/biosíntesis , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/psicología , Memoria a Corto Plazo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad , Proteínas PrPC/metabolismo
2.
Cell Biochem Biophys ; 69(2): 283-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24307281

RESUMEN

The role of the inflammatory agent fibrinogen (Fg) in increased pial venular permeability has been shown previously. It was suggested that an activation of matrix metalloproteinase-9 (MMP-9) is involved in Fg-induced enhanced transcytosis through endothelial cells (ECs). However, direct link between Fg, caveolae formation, and MMP-9 activity has never been shown. We hypothesized that at an elevated level, Fg enhances formation of functional caveolae through activation of MMP-9. Male wild-type (WT, C57BL/6J) or MMP-9 gene knockout (MMP9-/-) mice were infused with Fg (4 mg/ml, final blood concentration) or equal volume of phosphate buffered saline (PBS). After 2 h, mice were sacrificed and brains were collected for immunohistochemical analyses. Mouse brain ECs were treated with 4 mg/ml of Fg or PBS in the presence or absence of MMP-9 activity inhibitor, tissue inhibitor of metalloproteinases-4 (TIMP-4, 12 ng/ml). Formation of functional caveolae was assessed by confocal microscopy. Fg-induced increased formation of caveolae, which was defined by an increased co-localization of caveolin-1 (Cav-1) and plasmalemmal vesicle-associated protein-1 and was associated with an increased phosphorylation of Cav-1, was ameliorated in the presence of TIMP-4. These results suggest that at high levels, Fg enhances formation of functional caveolae that may involve Cav-1 signaling and MMP-9 activation.


Asunto(s)
Caveolas/metabolismo , Fibrinógeno/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/patología , Proteínas Portadoras/metabolismo , Caveolina 1/metabolismo , Línea Celular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fibrinógeno/farmacología , Inmunohistoquímica , Masculino , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Inhibidores Tisulares de Metaloproteinasas/farmacología , Inhibidor Tisular de Metaloproteinasa-4
3.
Curr Neurovasc Res ; 9(4): 274-81, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22873727

RESUMEN

Trauma introduces damaging stressors that compromise protein, lipid, and nucleic acid integrity. Aggregates of unfolded and misfolded proteins in the endoplasmic reticulum (ER) triggers the ER stress response (ERSR)/unfolded protein response (UPR) leading to activation of three signaling pathways mediated by PERK, ATF6, and IRE1. Initially, the ERSR/UPR is pro-homeostatic as it globally slows translation while increasing translation of chaperone proteins and inducing ER-associated degradation. If the cellular stress is not controlled, apoptosis is subsequently induced through several mechanisms, of which the most well-described is CHOP. Following spinal cord injury (SCI), mice deficient in CHOP signaling show increased spared white matter and enhanced locomotor recovery by 6 weeks. At 24 hours after SCI, ATF4 and CHOP are upregulated in under perfused microvessels. We observed vascular protection 3 days post-SCI and a significant decrease in macrophage infiltration by the end of the first week. These results suggest that modulating ER-stress signaling in endothelial cells and macrophages may protect against vascular injury and attenuate inflammation post-SCI.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Eliminación de Gen , Microvasos/metabolismo , Fármacos Neuroprotectores/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Factor de Transcripción CHOP/deficiencia , Animales , Estrés del Retículo Endoplásmico/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/patología , Microvasos/fisiopatología , Actividad Motora/genética , Corteza Motora/fisiología , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Vértebras Torácicas , Factor de Transcripción CHOP/genética
4.
Pain Res Treat ; 2012: 414697, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22229088

RESUMEN

Following inflammation, primary sensory neurons in the dorsal root ganglion (DRG) alter the production of several proteins. Most DRG neurons are glutamatergic, using glutaminase as the enzyme for glutamate production, but little is known about glutaminase following inflammation. In the present study, adjuvant-induced arthritis (AIA) was produced in rats with complete Freund's adjuvant into the hindpaw. At 7 days of AIA, DRG were examined with glutaminase immunohistochemistry, Western blot immunoreactivity, and enzyme activity. Image analysis revealed that glutaminase was elevated most in small-sized neurons (21%) (P < 0.05). Western blot analysis revealed a 19% increase (P < 0.05) in total glutaminase and 21% in mitochondrial glutaminase (P < 0.05). Glutaminase enzyme activity was elevated 29% (P < 0.001) from 2.20 to 2.83 moles/kg/hr. Elevated glutaminase in primary sensory neurons could lead to increased glutamate production in spinal primary afferent terminals contributing to central sensitization or in the peripheral process contributing to peripheral sensitization.

5.
J Cereb Blood Flow Metab ; 32(1): 150-63, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21989482

RESUMEN

Elevated blood level of Fibrinogen (Fg) is commonly associated with vascular dysfunction. We tested the hypothesis that at pathologically high levels, Fg increases cerebrovascular permeability by activating matrix metalloproteinases (MMPs). Fibrinogen (4 mg/mL blood concentration) or equal volume of phosphate-buffered saline (PBS) was infused into male wild-type (WT; C57BL/6J) or MMP-9 gene knockout (MMP9-/-) mice. Pial venular leakage of fluorescein isothiocyanate-bovine serum albumin to Fg or PBS alone and to topically applied histamine (10(-5) mol/L) were assessed. Intravital fluorescence microscopy and image analysis were used to assess cerebrovascular protein leakage. Pial venular macromolecular leakage increased more after Fg infusion than after infusion of PBS in both (WT and MMP9-/-) mice but was more pronounced in WT compared with MMP9-/- mice. Expression of vascular endothelial cadherin (VE-cadherin) was less and plasmalemmal vesicle-associated protein-1 (PV-1) was greater in Fg-infused than in PBS-infused both mice groups. However, in MMP9-/- mice, VE-cadherin expression was greater and PV-1 expression was less than in WT mice. These data indicate that at higher levels, Fg compromises microvascular integrity through activation of MMP-9 and downregulation of VE-cadherin and upregulation of PV-1. Our results suggest that elevated blood level of Fg could have a significant role in cerebrovascular dysfunction and remodeling.


Asunto(s)
Permeabilidad Capilar/fisiología , Venas Cerebrales/metabolismo , Fibrinógeno/farmacología , Metaloproteinasa 9 de la Matriz/fisiología , Animales , Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Permeabilidad Capilar/efectos de los fármacos , Proteínas Portadoras/biosíntesis , Venas Cerebrales/efectos de los fármacos , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Fibrinógeno/fisiología , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Infusiones Intraarteriales , Masculino , Metaloproteinasa 9 de la Matriz/genética , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Albúmina Sérica Bovina/farmacocinética , Regulación hacia Arriba , Vénulas/efectos de los fármacos , Vénulas/metabolismo
6.
Neurobiol Dis ; 42(1): 21-34, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21168495

RESUMEN

Recent data have implicated thrombospondin-1 (TSP-1) signaling in the acute neuropathological events that occur in microvascular endothelial cells (ECs) following spinal cord injury (SCI) (Benton et al., 2008b). We hypothesized that deletion of TSP-1 or its receptor CD47 would reduce these pathological events following SCI. CD47 is expressed in a variety of tissues, including vascular ECs and neutrophils. CD47 binds to TSP-1 and inhibits angiogenesis. CD47 also binds to the signal regulatory protein (SIRP)α and facilitates neutrophil diapedesis across ECs to sites of injury. After contusive SCI, TSP-1(-/-) mice did not show functional improvement compared to wildtype (WT) mice. CD47(-/-) mice, however, exhibited functional locomotor improvements and greater white matter sparing. Whereas targeted deletion of either CD47 or TSP-1 improved acute epicenter vascularity in contused mice, only CD47 deletion reduced neutrophil diapedesis and increased microvascular perfusion. An ex vivo model of the CNS microvasculature revealed that CD47(-/-)-derived microvessels (MVs) prominently exhibit adherent WT or CD47(-/-) neutrophils on the endothelial lumen, whereas WT-derived MVs do not. This implicates a defect in diapedesis mediated by the loss of CD47 expression on ECs. In vitro transmigration assays confirmed the role of SIRPα in neutrophil diapedesis through EC monolayers. We conclude that CD47 deletion modestly, but significantly, improves functional recovery from SCI via an increase in vascular patency and a reduction of SIRPα-mediated neutrophil diapedesis, rather than the abrogation of TSP-1-mediated anti-angiogenic signaling.


Asunto(s)
Antígeno CD47/genética , Terapia Genética/métodos , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/terapia , Médula Espinal/metabolismo , Animales , Antígeno CD47/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Actividad Motora/genética , Recuperación de la Función/genética , Médula Espinal/patología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología
7.
Transl Stroke Res ; 2(4): 556-74, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24323683

RESUMEN

Acute traumatic spinal cord injury (SCI) is characterized by a progressive secondary degeneration which exacerbates the loss of penumbral tissue and neurological function. Here, we first provide an overview of the known pathophysiological mechanisms involving injured microvasculature and molecular regulators that contribute to the loss and dysfunction of existing and new blood vessels. We also highlight the differences between traumatic and ischemic injuries which may yield clues as to the more devastating nature of traumatic injuries, possibly involving toxicity associated with hemorrhage. We also discuss known species differences with implications for choosing models, their relevance and utility to translate new treatments towards the clinic. Throughout this review, we highlight the potential opportunities and proof-of-concept experimental studies for targeting therapies to endothelial cell-specific responses. Lastly, we comment on the need for vascular mechanisms to be included in drug development and non-invasive diagnostics such as serum and cerebrospinal fluid biomarkers and imaging of spinal cord pathology.

8.
Microcirculation ; 17(7): 557-67, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21040121

RESUMEN

UNLABELLED: We have demonstrated that MFs isolated from adipose retain angiogenic potential in vitro and form a mature, perfused network when implanted. However, adipose-derived microvessels are rich in provascularizing cells that could uniquely drive neovascularization in adipose-derived MFs implants. OBJECTIVE: Investigate the ability of MFs from a different vascular bed to recapitulate adipose-derived microvessel angiogenesis and network formation and analyze adipose-derived vessel plasticity by assessing whether vessel function could be modulated by astrocyte-like cells. METHODS: MFs were isolated by limited collagenase digestion from rodent brain or adipose and assembled into 3D collagen gels in the presence or absence of GRPs. The resulting neovasculatures that formed following implantation were assessed by measuring 3D vascularity and vessel permeability to small and large molecular tracers. RESULTS: Similar to adipose-derived MFs, brain-derived MFs can sprout and form a perfused neovascular network when implanted. Furthermore, when co-implanted in the constructs, GRPs caused adipose-derived vessels to express the brain endothelial marker glucose transporter-1 and to significantly reduce microvessel permeability. CONCLUSION: Neovascularization involving isolated microvessel elements is independent of the tissue origin and degree of vessel specialization. In addition, adipose-derived vessels have the ability to respond to environmental signals and change vessel characteristics.


Asunto(s)
Microvasos/crecimiento & desarrollo , Microvasos/trasplante , Neovascularización Fisiológica , Adipocitos/citología , Adipocitos/trasplante , Animales , Astrocitos/citología , Permeabilidad Capilar , Separación Celular , Corteza Cerebral/irrigación sanguínea , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Epidídimo/irrigación sanguínea , Epidídimo/citología , Técnicas In Vitro , Masculino , Microvasos/citología , Microvasos/fisiología , Neuroglía/citología , Neuroglía/trasplante , Ratas
9.
Curr Neurovasc Res ; 7(3): 238-50, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20590523

RESUMEN

The endothelial-specific expression of plasmalemmal vesicle associated protein-1 (PV-1) is typical of fenestrated endothelium observed in pulmonary capillaries and some endocrine organs. In the central nervous system (CNS) it is expressed during development but disappears concomitant with maturation of the blood-CNS barrier [1]. Consistent with observations made in models of stroke, Alzheimer's disease, and tumorigenesis, we show PV-1 expression in the spinal cord specifically upregulated by pathologically-activated endothelial cells (ECs) in response to traumatic spinal cord injury (SCI). Adult female C57Bl/6 mice received a moderate T9/10 contusive SCI. PV-1 assessed by qRT-PCR and immunohistochemistry 3 hours to 14 days post-injury showed expression as early as 1 day post-SCI, with levels decreasing by 14 days. This expression was associated with microvessels in the injury epicenter and penumbral zone, with the time course and distribution correlated with progressing peripheral inflammatory cell infiltration. PV-1-immunoreactive ECs were angiogenic as demonstrated by intravascular binding of Griffonia simplicifolia isolectin B4 (IB4). ECs expressing high levels of PV-1 were anatomically and physiologically abnormal with altered/absent immunostaining for occludin and zonula occludens-1 (ZO-1), and decreased expression of glial fibrillary acidic protein (GFAP) and aquaporin-4 (AQP4). Glucose transporter type I (Glut-1) expression decreased in affected, PV-1 positive microvessels with little colocalization of PV-1 and Glut-1 apparent by 7 days post-SCI. These data suggest that upregulation of microvascular expression of PV-1 post-SCI may promote major components of secondary injury including extravasation of cellular and acellular mediators of inflammation and may accelerate loss of neuropil and decline in the functional and anatomical integrity of the neurovascular unit (NVU).


Asunto(s)
Astrocitos/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Glucosa/metabolismo , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Acuaporina 4/metabolismo , Astrocitos/patología , Transporte Biológico , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Inflamación/patología , Ratones , Ocludina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Traumatismos de la Médula Espinal/patología , Vértebras Torácicas
10.
Brain ; 133(Pt 4): 1026-42, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20375135

RESUMEN

Blood vessel loss and inflammation cause secondary degeneration following spinal cord injury. Angiopoietin-1 through the Tie2 receptor, and other ligands through alphavbeta3 integrin, promote endothelial cell survival during developmental or tumour angiogenesis. Here, daily intravenous injections with an alphavbeta3-binding peptide named C16 or an angiopoietin-1 mimetic following a spinal cord contusion at thoracic level 9 in mice rescued epicentre blood vessels, white matter and locomotor function, and reduced detrimental inflammation. Preserved vascularity and reduced inflammation correlated with improved outcomes. C16 and angiopoietin-1 reduced leukocyte transmigration in vitro. Growth factor receptors and integrins facilitate each others' function. Therefore, angiopoietin-1 and C16 were combined and the effects were additive, resulting in almost complete functional recovery. The treatment had lasting effects when started 4 h following injury and terminated after one week. These results identify alphavbeta3 integrin and the endothelial-selective angiopoietin-1 as vascular and inflammatory regulators that can be targeted in a clinically relevant manner for neuroprotection after central nervous system trauma.


Asunto(s)
Angiopoyetina 1/administración & dosificación , Integrina alfaVbeta3/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Traumatismos de la Médula Espinal/prevención & control , Médula Espinal/irrigación sanguínea , Médula Espinal/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Quimioterapia Combinada , Femenino , Humanos , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Vértebras Torácicas , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA