Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Circulation ; 141(25): 2078-2094, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32183562

RESUMEN

BACKGROUND: Spontaneous deep intracerebral hemorrhage (ICH) is a devastating subtype of stroke without specific treatments. It has been thought that smooth muscle cell (SMC) degeneration at the site of arteriolar wall rupture may be sufficient to cause hemorrhage. However, deep ICHs are rare in some aggressive small vessel diseases that are characterized by significant arteriolar SMC degeneration. Here we hypothesized that a second cellular defect may be required for the occurrence of ICH. METHODS: We studied a genetic model of spontaneous deep ICH using Col4a1+/G498V and Col4a1+/G1064D mouse lines that are mutated for the α1 chain of collagen type IV. We analyzed cerebroretinal microvessels, performed genetic rescue experiments, vascular reactivity analysis, and computational modeling. We examined postmortem brain tissues from patients with sporadic deep ICH. RESULTS: We identified in the normal cerebroretinal vasculature a novel segment between arterioles and capillaries, herein called the transitional segment (TS), which is covered by mural cells distinct from SMCs and pericytes. In Col4a1 mutant mice, this TS was hypermuscularized, with a hyperplasia of mural cells expressing more contractile proteins, whereas the upstream arteriole exhibited a loss of SMCs. TSs mechanistically showed a transient increase in proliferation of mural cells during postnatal maturation. Mutant brain microvessels, unlike mutant arteries, displayed a significant upregulation of SM genes and Notch3 target genes, and genetic reduction of Notch3 in Col4a1+/G498V mice protected against ICH. Retina analysis showed that hypermuscularization of the TS was attenuated, but arteriolar SMC loss was unchanged in Col4a1+/G498V, Notch3+/- mice. Moreover, hypermuscularization of the retinal TS increased its contractility and tone and raised the intravascular pressure in the upstream feeding arteriole. We similarly found hypermuscularization of the TS and focal arteriolar SMC loss in brain tissues from patients with sporadic deep ICH. CONCLUSIONS: Our results suggest that hypermuscularization of the TS, through increased Notch3 activity, is involved in the occurrence of ICH in Col4a1 mutant mice, by raising the intravascular pressure in the upstream feeding arteriole and promoting its rupture at the site of SMC loss. Our human data indicate that these 2 mutually reinforcing vascular defects may represent a general mechanism of deep ICH.


Asunto(s)
Hemorragia Cerebral/etiología , Hemorragia Cerebral/prevención & control , Microvasos/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Biomarcadores , Hemorragia Cerebral/diagnóstico , Hemorragia Cerebral/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Genotipo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Microvasos/fisiopatología , Imagen Molecular , Mutación , Miocitos del Músculo Liso/metabolismo , Receptor Notch3/metabolismo , Retina/metabolismo , Retina/patología , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología
2.
Acta Neuropathol Commun ; 7(1): 187, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31753008

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a genetic paradigm of small vessel disease (SVD) caused by NOTCH3 mutations that stereotypically lead to the vascular accumulation of NOTCH3 around smooth muscle cells and pericytes. White matter (WM) lesions (WMLs) are the earliest and most frequent abnormalities, and can be associated with lacunar infarcts and enlarged perivascular spaces (ePVS). The prevailing view is that blood brain barrier (BBB) leakage, possibly mediated by pericyte deficiency, plays a pivotal role in the formation of WMLs. Herein, we investigated the involvement of BBB leakage and pericyte loss in CADASIL WMLs. Using post-mortem brain tissue from 12 CADASIL patients and 10 age-matched controls, we found that WMLs are heterogeneous, and that BBB leakage reflects the heterogeneity. Specifically, while fibrinogen extravasation was significantly increased in WMLs surrounding ePVS and lacunes, levels of fibrinogen leakage were comparable in WMLs without other pathology ("pure" WMLs) to those seen in the normal appearing WM of patients and controls. In a mouse model of CADASIL, which develops WMLs but no lacunes or ePVS, we detected no extravasation of endogenous fibrinogen, nor of injected small or large tracers in WMLs. Moreover, there was no evidence of pericyte coverage modification in any type of WML in either CADASIL patients or mice. These data together indicate that WMLs in CADASIL encompass distinct classes of WM changes and argue against the prevailing hypothesis that pericyte coverage loss and BBB leakage are the primary drivers of WMLs. Our results also have important implications for the interpretation of studies on the BBB in living patients, which may misinterpret evidence of BBB leakage within WM hyperintensities as suggesting a BBB related mechanism for all WMLs, when in fact this may only apply to a subset of these lesions.


Asunto(s)
Barrera Hematoencefálica/patología , Encéfalo/patología , CADASIL/patología , Sustancia Blanca/patología , Anciano , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , CADASIL/metabolismo , Permeabilidad Capilar/fisiología , Estudios de Cohortes , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Sustancia Blanca/irrigación sanguínea , Sustancia Blanca/metabolismo
3.
Ann Neurol ; 84(2): 246-259, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30014602

RESUMEN

OBJECTIVE: CADASIL (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy), caused by dominant mutations in the NOTCH3 receptor, is the most aggressive small vessel disease of the brain. A key feature of its pathogenesis is accumulation of the extracellular domain of NOTCH3 receptor (Notch3ECD ) in small vessels, with formation of characteristic extracellular deposits termed granular osmiophilic material (GOM). Here, we investigated the therapeutic potential of a mouse monoclonal antibody (5E1) that specifically recognizes Notch3ECD . METHODS: The binding affinity of 5E1 toward purified NOTCH3 was assessed using Octet analysis. The ability of 5E1 to bind Notch3ECD deposits in brain vessels and its effects on disease-related phenotypes were evaluated in the CADASIL mouse model, which overexpresses a mutant rat NOTCH3. Notch3ECD and GOM deposition, white matter lesions, and cerebral blood flow deficits were assessed at treatment initiation (10 weeks) and study completion (30 weeks) using quantitative immunohistochemistry, electron microscopy, and laser-Doppler flowmetry. RESULTS: 5E1 antibody bound recombinant rat NOTCH3 with an average affinity of 317nM. A single peripheral injection of 5E1 robustly decorated Notch3ECD deposits in the brain vasculature. Chronic administration of 5E1 did not attenuate Notch3ECD or GOM deposition and was not associated with perivascular microglial activation. It also failed to halt the development of white matter lesions. Despite this, 5E1 treatment markedly protected against impaired cerebral blood flow responses to neural activity and topical application of vasodilators and normalized myogenic responses of cerebral arteries. INTERPRETATION: This study establishes immunotherapy targeting Notch3ECD as a new avenue for disease-modifying treatment in CADASIL that warrants further development. Ann Neurol 2018;84:246-259.


Asunto(s)
CADASIL/metabolismo , CADASIL/terapia , Circulación Cerebrovascular/fisiología , Matriz Extracelular/metabolismo , Inmunoterapia/métodos , Receptor Notch3/metabolismo , Animales , CADASIL/inmunología , Matriz Extracelular/inmunología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Unión Proteica/fisiología , Ratas , Receptor Notch3/administración & dosificación , Receptor Notch3/inmunología
4.
J Pathol ; 244(4): 408-420, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29266233

RESUMEN

Mutations in the α1 (COL4A1) or α2 (COL4A2) chains of collagen type IV, a major component of the vascular basement membrane, cause intracerebral haemorrhages with variable expressivity and reduced penetrance by mechanisms that remain poorly understood. Here we sought to investigate the cellular mechanisms of COL4A1-related intracerebral haemorrhage and identify a marker for haemorrhage risk stratification. A combination of histological, immunohistochemical, and electron microscopy analyses were used to analyse the brain parenchyma, cerebrovasculature, and retinal vessels of mice expressing the disease-causing COL4A1 p.G498V mutation. Mutant mice developed cerebral microhaemorrhages and macroscopic haemorrhages (macrohaemorrhages), the latter with reduced penetrance, mimicking the human disease. Microhaemorrhages that occurred in early postnatal life were associated with a transient, generalized increase in blood-brain barrier permeability at the level of capillaries. Macrohaemorrhages, which occurred later in life, originated from deep brain arteries with focal loss of smooth muscle cells. Similar smooth muscle cell loss was detected in retinal arteries, and a time-course analysis of arterial lesions showed that smooth muscle cells are recruited normally in arterial wall during development, but undergo progressive apoptosis-mediated degeneration. By assessing in parallel the extent of these retinal arterial lesions and the presence/absence of macrohaemorrhages, we found that the arterial lesion load in the retina is strongly correlated with the burden of macrohaemorrhages. We conclude that microhaemorrhages and macrohaemorrhages are driven by two distinct mechanisms. Moreover, smooth muscle cell degeneration is a critical factor underlying the partial penetrance of COL4A1-related macrohaemorrhages, and retinal imaging is a promising tool for identifying high-risk patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Hemorragia Cerebral/patología , Colágeno Tipo IV/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Accidente Cerebrovascular/patología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Proliferación Celular , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Colágeno Tipo IV/deficiencia , Colágeno Tipo IV/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Predisposición Genética a la Enfermedad , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/ultraestructura , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/ultraestructura , Penetrancia , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Arteria Retiniana/metabolismo , Arteria Retiniana/patología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Factores de Tiempo
5.
J Cereb Blood Flow Metab ; 36(1): 143-57, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25853907

RESUMEN

The term matrisome refers to the ensemble of proteins constituting the extracellular matrix (ECM) (core matrisome) as well as the proteins associated with the ECM. Every organ has an ECM with a unique composition that not only provides the support and anchorage for cells, but also controls fundamental cellular processes as diverse as differentiation, survival, proliferation, and polarity. The current knowledge of the matrisome of small brain vessels is reviewed with a focus on the basement membrane (BM), a specialized form of ECM located at the interface between endothelial cells, contractile cells (smooth muscle cells and pericytes), and astrocyte endfeet­a very strategic location in the communication pathway between the cerebral microcirculation and astrocytes. We discuss some of the most recent genetic data and relevant findings from experimental models of nonamyloid cerebral small vessel disease (SVD). We propose the concept that perturbations of the cerebrovascular matrisome is a convergent pathologic pathway in monogenic forms of SVD, and is likely relevant to the sporadic disease.


Asunto(s)
Encéfalo/irrigación sanguínea , Enfermedades de los Pequeños Vasos Cerebrales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Microvasos/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Enfermedades de los Pequeños Vasos Cerebrales/genética , Enfermedades de los Pequeños Vasos Cerebrales/patología , Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/genética , Humanos , Microvasos/patología , Mutación
6.
Mol Immunol ; 62(1): 104-13, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24980869

RESUMEN

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system in which anti-aquaporin-4 (AQP4) autoantibodies (AQP4-IgG) cause damage to astrocytes by complement-dependent cytotoxicity (CDC). Various approaches have been attempted to produce NMO lesions in rodents, some involving genetically modified mice with altered immune cell function. Here, we found that mouse serum strongly inhibits complement from multiple species, preventing AQP4-IgG-dependent CDC. Effects of mouse serum on complement activation were tested in CDC assays in which AQP4-expressing cells were incubated with AQP4-IgG and complement from different species. Biochemical assays and mass spectrometry were used to characterize complement inhibitor(s) in mouse serum. Sera from different strains of mice produced almost no AQP4-IgG-dependent CDC compared with human, rat and guinea pig sera. Remarkably, addition of mouse serum prevented AQP4-IgG-dependent CDC caused by human, rat or guinea pig serum, with 50% inhibition at <5% mouse serum. Hemolysis assays indicated that the inhibitor(s) in mouse serum target the classical and not the alternative complement pathway. We found that the complement inhibitor(s) in mouse serum were contained in a serum fraction purified with protein-A resin; however, the inhibitor was not IgG as determined using serum from IgG-deficient mice. Mass spectrometry on the protein A-purified fraction produced several inhibitor candidates. The low intrinsic complement activity of mouse serum and the presence of complement inhibitor(s) limit the utility of mouse models to study disorders, such as NMO, involving the classical complement pathway.


Asunto(s)
Inactivadores del Complemento/sangre , Vía Clásica del Complemento/inmunología , Neuromielitis Óptica/inmunología , Animales , Acuaporina 4/inmunología , Autoanticuerpos/sangre , Células CHO , Inactivadores del Complemento/inmunología , Cricetulus , Modelos Animales de Enfermedad , Cobayas , Humanos , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuromielitis Óptica/sangre , Ratas , Ratas Wistar
7.
Muscle Nerve ; 50(5): 859-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24862586

RESUMEN

INTRODUCTION: Neuromyelitis optica (NMO) is characterized by inflammatory demyelinating lesions of the spinal cord and optic nerves from an autoimmune response against water channel aquaporin-4 (AQP4). We report 2 patients with transient hyperCKemia associated with NMO suggesting possible skeletal muscle damage. METHODS: Patient 1 was a 72-year-old man who presented with muscle soreness and elevated serum creatine kinase (CK) preceding an initial attack of NMO. Patient 2 was a 25-year-old woman with an established diagnosis of NMO who presented with diffuse myalgias, proximal upper extremity weakness, and hyperCKemia. Muscle biopsies were obtained for histopathologic evaluation, protein gel electrophoresis, immunofluorescence, and complement staining. RESULTS: In both patients the muscle showed only mild variation in fiber diameter. There were no inflammatory changes or muscle fiber necrosis, though there was reduced AQP4 expression and deposition of activated complement. CONCLUSIONS: Complement-mediated sarcolemmal injury may lead to hyperCKemia in NMO.


Asunto(s)
Creatina Quinasa/sangre , Neuromielitis Óptica/sangre , Anciano , Acuaporina 4/sangre , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Neuromielitis Óptica/enzimología , Médula Espinal/patología , Adulto Joven
8.
Neurosci Lett ; 574: 70-5, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24717641

RESUMEN

Astrocyte water channel aquaporin-4 (AQP4) facilitates water movement across the blood-brain barrier and into injured astrocytes. We previously showed reduced cytotoxic brain edema with improved neurological outcome in AQP4 knockout mice in water intoxication, infection and cerebral ischemia. Here, we established a 4-vessel transient occlusion model to test the hypothesis that AQP4 deficiency in mice could improve neurological outcome following severe global cerebral ischemia as occurs in cardiac arrest/resuscitation. Mice were subjected to 10-min transient bilateral carotid artery occlusion at 24h after bilateral vertebral artery cauterization. Cerebral blood flow was reduced during occlusion by >94% in both AQP4(+/+) and AQP4(-/-) mice. The primary outcome, neurological score, was remarkably better at 3 and 5 days after occlusion in AQP4(-/-) than in AQP4(+/+) mice, and survival was significantly improved as well. Brain water content was increased by 2.8±0.4% in occluded AQP4(+/+) mice, significantly greater than that of 0.3±0.6% in AQP4(-/-) mice. Histological examination and immunofluorescence of hippocampal sections at 5 days showed significantly greater neuronal loss in the CA1 region of hippocampus in AQP4(+/+) than AQP4(-/-) mice. The neuroprotection in mice conferred by AQP4 deletion following severe global cerebral ischemia provides proof-of-concept for therapeutic AQP4 inhibition to improve neurological outcome in cardiac arrest.


Asunto(s)
Acuaporina 4/metabolismo , Isquemia Encefálica/metabolismo , Animales , Acuaporina 4/genética , Arteriopatías Oclusivas/complicaciones , Astrocitos/patología , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/etiología , Isquemia Encefálica/patología , Arterias Carótidas , Inflamación/metabolismo , Ratones Noqueados , Vaina de Mielina/metabolismo , Neuronas/patología , Arteria Vertebral
9.
Exp Neurol ; 255: 145-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24636863

RESUMEN

Neuromyelitis optica (NMO) pathogenesis involves binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) present in serum to AQP4 on astrocytes, which causes complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). Human immunoglobulin G (hIgG) is effective for treatment of humorally mediated neurological autoimmune diseases and has been reported to improve disease outcome in a limited number of NMO patients. Here, we investigated hIgG actions on NMO-IgG pathogenicity using an in vivo rat model of NMO and in vitro assays. In rats administered NMO-IgG by intracerebral injection, the size of neuroinflammatory demyelinating lesions was reduced by ~50% when hIgG was administered by intraperitoneal injection to reach levels of 10-25mg/mL in rat serum, comparable with human therapeutic levels. In vitro, hIgG at 10mg/mL reduced by 90% NMO-IgG-mediated CDC following addition of NMO-IgG and human complement to AQP4-expressing cells. The hIgG effect was mainly on the classical complement pathway. hIgG at 10mg/mL also reduced by up to 90% NMO-IgG-mediated ADCC as assayed with human natural killer cells as effector cells. However, hIgG at up to 40mg/mL did not affect AQP4 cell surface expression or its supramolecular assembly in orthogonal arrays of particles, nor did it affect NMO-IgG binding to AQP4. We conclude that hIgG reduces NMO-IgG pathogenicity by inhibition of CDC and ADCC, providing a mechanistic basis to support further clinical evaluation of its therapeutic efficacy in NMO.


Asunto(s)
Acuaporina 4/inmunología , Autoanticuerpos/metabolismo , Inmunoglobulina G/uso terapéutico , Neuromielitis Óptica/inmunología , Animales , Acuaporina 4/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/farmacología , Neuromielitis Óptica/tratamiento farmacológico , Neuromielitis Óptica/patología , Ratas , Ratas Endogámicas Lew
10.
J Cell Biol ; 204(4): 559-73, 2014 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-24515349

RESUMEN

The astrocyte water channel aquaporin-4 (AQP4) is expressed as heterotetramers of M1 and M23 isoforms in which the presence of M23-AQP4 promotes formation of large macromolecular aggregates termed orthogonal arrays. Here, we demonstrate that the AQP4 aggregation state determines its subcellular localization and cellular functions. Individually expressed M1-AQP4 was freely mobile in the plasma membrane and could diffuse into rapidly extending lamellipodial regions to support cell migration. In contrast, M23-AQP4 formed large arrays that did not diffuse rapidly enough to enter lamellipodia and instead stably bound adhesion complexes and polarized to astrocyte end-feet in vivo. Co-expressed M1- and M23-AQP4 formed aggregates of variable size that segregated due to diffusional sieving of small, mobile M1-AQP4-enriched arrays into lamellipodia and preferential interaction of large, M23-AQP4-enriched arrays with the extracellular matrix. Our results therefore demonstrate an aggregation state-dependent mechanism for segregation of plasma membrane protein complexes that confers specific functional roles to M1- and M23-AQP4.


Asunto(s)
Acuaporina 4/fisiología , Astrocitos/citología , Neoplasias Encefálicas/patología , Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Glioblastoma/patología , Seudópodos/metabolismo , Animales , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Glioblastoma/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Noqueados , Modelos Teóricos , Isoformas de Proteínas , Multimerización de Proteína , Puntos Cuánticos
11.
J Neuroinflammation ; 11: 16, 2014 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-24468108

RESUMEN

BACKGROUND: Although optic neuritis (ON) is a defining feature of neuromyelitis optica (NMO), appropriate animal models of NMO ON are lacking. Most NMO patients are seropositive for immunoglobulin G autoantibodies (NMO-IgG) against the astrocyte water channel aquaporin-4 (AQP4). METHODS: Several approaches were tested to develop a robust, passive-transfer mouse model of NMO ON, including NMO-IgG and complement delivery by: (i) retrobulbar infusion; (ii) intravitreal injection; (iii) a single intracranial injection near the optic chiasm; and (iv) 3-days continuous intracranial infusion near the optic chiasm. RESULTS: Little ON or retinal pathology was seen using approaches (i) to (iii). Using approach (iv), however, optic nerves showed characteristic NMO pathology, with loss of AQP4 and glial fibrillary acidic protein immunoreactivity, granulocyte and macrophage infiltration, deposition of activated complement, demyelination and axonal injury. Even more extensive pathology was created in mice lacking complement inhibitor protein CD59, or using a genetically modified NMO-IgG with enhanced complement effector function, including significant loss of retinal ganglion cells. In control studies, optic nerve pathology was absent in treated AQP4-deficient mice, or in wild-type mice receiving control (non-NMO) IgG and complement. CONCLUSION: Passive transfer of NMO-IgG and complement by continuous infusion near the optic chiasm in mice is sufficient to produce ON with characteristic NMO pathology. The mouse model of NMO ON should be useful in further studies of NMO pathogenesis mechanisms and therapeutics.


Asunto(s)
Acuaporina 4/inmunología , Enfermedades Desmielinizantes/etiología , Inmunización Pasiva/efectos adversos , Inmunoglobulina G/inmunología , Neuromielitis Óptica/inmunología , Animales , Acuaporina 4/deficiencia , Antígenos CD59/genética , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuromielitis Óptica/complicaciones , Neuromielitis Óptica/etiología , Nervio Óptico/inmunología , Nervio Óptico/metabolismo , Retina/inmunología , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/metabolismo
12.
FASEB J ; 28(2): 705-14, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24186965

RESUMEN

Aquaporin-4 (AQP4), the principal water channel in astrocytes, is involved in brain water movement, inflammation, and neuroexcitation. In this study, there was strong neuroprotection in mice lacking AQP4 in a model of global cerebral ischemia produced by transient, bilateral carotid artery occlusion (BCAO). Survival and neurological outcome were greatly improved in the AQP4(-/-) vs. AQP4(+/+) mice after occlusion, with large and robust differences in both outbred (CD1) and inbred (C57bl/6) mouse strains without or with mechanical ventilation. Improved survival was also seen in mice lacking the scaffold protein α-syntrophin, which manifest reduced astrocyte water permeability secondary to defective AQP4 plasma membrane targeting. Intracranial pressure elevation and brain water accumulation were much reduced in the AQP4(-/-) vs. AQP4(+/+) mice after carotid artery occlusion, as were blood-brain barrier (BBB) disruption and neuronal loss. Brain slices from AQP4(-/-) mice showed significantly reduced cell swelling and cytotoxicity in response to oxygen-glucose deprivation, compared with slices from AQP4(+/+) mice. Our findings suggest that the neuroprotective effect of AQP4 deletion in global cerebral ischemia involves reduced astrocyte swelling and brain water accumulation, resulting in reduced BBB disruption, inflammation, and neuron death. AQP4 water transport inhibition may improve survival and neurological outcome after cardiac arrest and in other conditions associated with global cerebral ischemia.


Asunto(s)
Acuaporina 4/metabolismo , Isquemia Encefálica/metabolismo , Animales , Acuaporina 4/genética , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatología , Hipocampo/metabolismo , Hipocampo/patología , Inmunohistoquímica , Presión Intracraneal/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
13.
Acta Neuropathol ; 127(4): 539-51, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24190619

RESUMEN

Animal models of neuromyelitis optica (NMO) are needed for elucidation of disease mechanisms and for testing new therapeutics. Prior animal models of NMO involved administration of human anti-aquaporin-4 immunoglobulin G antibody (NMO-IgG) to rats with pre-existing neuroinflammation, or to naïve mice supplemented with human complement. We report here the development of NMO pathology following passive transfer of NMO-IgG to naïve rats. A single intracerebral infusion of NMO-IgG to adult Lewis rats produced robust lesions around the needle track in 100 % of rats; at 5 days there was marked loss of aquaporin-4 (AQP4), glial fibrillary acidic protein (GFAP) and myelin, granulocyte and macrophage infiltration, vasculocentric complement deposition, blood-brain barrier disruption, microglial activation and neuron death. Remarkably, a distinct 'penumbra' was seen around lesions, with loss of AQP4 but not of GFAP or myelin. No lesions or penumbra were seen in rats receiving control IgG. The size of the main lesion with loss of myelin was greatly reduced in rats made complement-deficient by cobra venom factor or administered NMO-IgG lacking complement-dependent cytotoxicity (CDC) effector function. However, the penumbra was seen under these conditions, suggesting a complement-independent pathogenesis mechanism. The penumbra was absent with NMO-IgG lacking both CDC and antibody-dependent cellular cytotoxicity (ADCC) effector functions. Finally, lesion size was significantly reduced after macrophage depletion with clodronate liposomes. These results: (i) establish a robust, passive-transfer model of NMO in rats that does not require pre-existing neuroinflammation or complement administration; (ii) implicate ADCC as responsible for a unique type of pathology also seen in human NMO; and (iii) support a pathogenic role of macrophages in NMO.


Asunto(s)
Acuaporina 4/inmunología , Inmunoglobulina G , Neuromielitis Óptica/inducido químicamente , Neuromielitis Óptica/patología , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/patología , Bucladesina/farmacología , Células CHO , Corteza Cerebral/citología , Cricetulus , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inmunoglobulina G/sangre , Ratones , Vaina de Mielina/patología , Proteínas del Tejido Nervioso/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Endogámicas Lew , Transfección
14.
Acta Neuropathol ; 126(5): 699-709, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23995423

RESUMEN

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system that can cause paralysis and blindness. The pathogenesis of NMO involves binding of immunoglobulin G autoantibodies to aquaporin-4 (AQP4) on astrocytes, which is thought to cause complement-dependent cytotoxicity (CDC) and a secondary inflammatory response leading to oligodendrocyte and neuronal damage. Here, we investigate in vivo the role of antibody-dependent cell-mediated cytotoxicity (ADCC) triggered by AQP4 autoantibodies (AQP4-IgG) in the development of NMO pathology. A high-affinity, human recombinant monoclonal AQP4-IgG was mutated in its Fc region to produce 'NMO superantibodies' with enhanced CDC and/or ADCC effector functions, without altered AQP4 binding. Pathological effects of these antibodies were studied in a mouse model of NMO produced by intracerebral injection of AQP4-IgG and human complement. The original (non-mutated) antibody produced large NMO lesions in this model, with loss of AQP4 and GFAP immunoreactivity, inflammation and demyelination, as did a mutated antibody with enhanced CDC and ADCC effector functions. As anticipated, a mutated AQP4-IgG lacking CDC, but having tenfold enhanced ADCC, produced little pathology. However, unexpectedly, a mutated antibody with ninefold enhanced CDC, but lacking ADCC, produced much less pathology than the original AQP4-IgG. Also, pathology was greatly reduced following administration of AQP4-IgG and complement to mice lacking the FcγIII receptor involved in effector cell activation during ADCC, and to normal mice injected with an Fcγ receptor blocking antibody. Our results provide evidence for the central involvement of ADCC in NMO pathology and suggest ADCC as a new therapeutic target in NMO.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Autoanticuerpos/inmunología , Enfermedades Desmielinizantes/inmunología , Inflamación/inmunología , Neuromielitis Óptica/inmunología , Animales , Acuaporina 4/inmunología , Autoantígenos/inmunología , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoglobulina G/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Transfección
15.
Ann Neurol ; 73(1): 77-85, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23055279

RESUMEN

OBJECTIVE: Neuromyelitis optica (NMO) is caused by binding of pathogenic autoantibodies (NMO-immunoglobulin G [IgG]) to aquaporin-4 (AQP4) on astrocytes, which initiates complement-dependent cytotoxicity (CDC) and inflammation. We recently introduced mutated antibody (aquaporumab) and small-molecule blocker strategies for therapy of NMO, based on prevention of NMO-IgG binding to AQP4. Here, we investigated an alternative strategy involving neutralization of NMO-IgG effector function by selective IgG heavy-chain deglycosylation with bacteria-derived endoglycosidase S (EndoS). METHODS: Cytotoxicity and NMO pathology were measured in cell and spinal cord slice cultures, and in mice exposed to control or EndoS-treated NMO-IgG. RESULTS: EndoS treatment of NMO patient serum reduced by >95% CDC and antibody-dependent cell-mediated cytotoxicity, without impairment of NMO-IgG binding to AQP4. Cytotoxicity was also prevented by addition of EndoS after NMO-IgG binding to AQP4. The EndoS-treated, nonpathogenic NMO-IgG competitively displaced pathogenic NMO-IgG bound to AQP4, and prevented NMO pathology in spinal cord slice culture and mouse models of NMO. INTERPRETATION: EndoS deglycosylation converts pathogenic NMO-IgG autoantibodies into therapeutic blocking antibodies. EndoS treatment of blood may be beneficial in NMO, and may be accomplished, for example, by therapeutic apheresis using surface-immobilized EndoS.


Asunto(s)
Acuaporina 4/sangre , Acuaporina 4/uso terapéutico , Inmunoglobulina G/biosíntesis , Neuromielitis Óptica/sangre , Neuromielitis Óptica/terapia , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Autoanticuerpos/uso terapéutico , Proteínas Bacterianas/fisiología , Células CHO , Cricetinae , Cricetulus , Glicósido Hidrolasas/fisiología , Glicosilación , Humanos , Inmunoglobulina G/sangre , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Médula Espinal/enzimología
16.
Glia ; 60(12): 2027-39, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22987455

RESUMEN

Neuromyelitis optica (NMO) is thought to be caused by immunoglobulin G autoantibodies (NMO-IgG) against astrocyte water channel aquaporin-4 (AQP4). A recent study (Hinson et al. (2012) Proc Natl Acad Sci USA 109:1245-1250) reported that NMO-IgG inhibits AQP4 water permeability directly and causes rapid cellular internalization of the M1 but not M23 isoform of AQP4, resulting in AQP4 clustering, enhanced complement-dependent cytotoxicity, and tissue swelling. Here, we report evidence challenging this proposed mechanism of NMO-IgG-mediated pathology. We measured osmotic water permeability by stopped-flow light scattering on plasma membrane vesicles isolated from AQP4-expressing CHO cells, an approach that can detect changes in water permeability as small as 5% and is not confounded by internalization effects. We found similar single-molecule water permeability for M1-AQP4 tetramers and M23-AQP4 clusters (orthogonal arrays of particles, OAPs). Exposure of AQP4 to high concentrations of NMO-IgG from six seropositive NMO patients, and to high-affinity recombinant monoclonal NMO antibodies, did not reduce AQP4 water permeability. Also, NMO-IgG did not reduce water permeability in AQP4-reconstituted proteoliposomes. In transfected cells expressing M1- or M23-AQP4 individually, NMO-IgG caused more rapid internalization of M23- than M1-AQP4. In cells coexpressing both isoforms, M1- and M23-AQP4 comingled in OAPs that were internalized together in response to NMO-IgG. Super-resolution imaging and native gel electrophoresis showed that the size of AQP4 OAPs was not altered by NMO sera or recombinant NMO antibodies. We conclude that NMO-IgG does not: (i) inhibit AQP4 water permeability, (ii) cause preferential internalization of M1-AQP4, or (iii) cause intramembrane AQP4 clustering.


Asunto(s)
Acuaporina 4/metabolismo , Autoanticuerpos/fisiología , Permeabilidad de la Membrana Celular/fisiología , Membrana Celular/metabolismo , Inmunoglobulina G/fisiología , Neuromielitis Óptica/inmunología , Animales , Anticuerpos Monoclonales/fisiología , Acuaporina 4/antagonistas & inhibidores , Acuaporina 4/química , Células CHO , Línea Celular Tumoral , Membrana Celular/patología , Cricetinae , Cricetulus , Humanos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Neuromielitis Óptica/metabolismo , Neuromielitis Óptica/patología , Unión Proteica/fisiología , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Agua/metabolismo
17.
Int J Biochem Cell Biol ; 44(9): 1519-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22713791

RESUMEN

Neuromyelitis optica (NMO) is an autoimmune 'aquaporinopathy' of the central nervous system that causes inflammatory demyelinating lesions primarily in spinal cord and optic nerve, leading to paralysis and blindness. NMO lesions show loss of aquaporin-4 (AQP4), GFAP and myelin, infiltration of granulocytes and macrophages, and perivascular deposition of activated complement. Most patients with NMO are seropositive for immunoglobulin autoantibodies (AQP4-IgG) against AQP4, the principal water channel of astrocytes. There is strong evidence that AQP4-IgG is pathogenic in NMO, probably by a mechanism involving complement-dependent astrocyte cytotoxicity, causing leukocyte infiltration, cytokine release and blood-brain barrier disruption, which leads to oligodendrocyte death, myelin loss and neuron death. Here, we review the evidence for this and alternative proposed NMO pathogenesis mechanisms, such as AQP4-IgG-induced internalization of AQP4 and glutamate transporters, complement-independent cell-mediated cytotoxicity, and AQP4-IgG inhibition of AQP4 water transport function. Based on the initiating pathogenic role of AQP4-IgG binding to astrocyte AQP4 in NMO, selective blocker therapies are under development in which AQP4-targeted monoclonal antibodies or small molecules block binding of AQP4-IgG to astrocytes and consequent downstream pathology.


Asunto(s)
Acuaporina 4 , Neuromielitis Óptica/etiología , Neuromielitis Óptica/terapia , Animales , Acuaporina 4/química , Acuaporina 4/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Neuromielitis Óptica/metabolismo , Neuromielitis Óptica/patología , Técnicas de Cultivo de Órganos
18.
J Cell Sci ; 125(Pt 18): 4405-12, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22718347

RESUMEN

Aquaporin-4 (AQP4) is a water channel expressed in astrocytes, skeletal muscle and epithelial cells that forms supramolecular aggregates in plasma membranes called orthogonal arrays of particles (OAPs). AQP4 is expressed as a short isoform (M23) that forms large OAPs, and a long isoform (M1) that does not form OAPs by itself but can mingle with M23 to form relatively small OAPs. AQP4 OAPs were imaged with ~20 nm spatial precision by photoactivation localization microscopy (PALM) in cells expressing chimeras of M1- or M23-AQP4 with photoactivatable fluorescent proteins. Native AQP4 was imaged by direct stochastic optical reconstruction microscopy (dSTORM) using a primary anti-AQP4 antibody and fluorescent secondary antibodies. We found that OAP area increased from 1878±747 to 3647±958 nm(2) with decreasing M1:M23 ratio from 1:1 to 1:3, and became elongated. Two-color dSTORM indicated that M1 and M23 co-assemble in OAPs with a M1-enriched periphery surrounding a M23-enriched core. Native AQP4 in astrocytes formed OAPs with an area of 2142±829 nm(2), which increased to 5137±1119 nm(2) with 2-bromopalmitate. PALM of AQP4 OAPs in live cells showed slow diffusion (average ~10(-12) cm(2)/s) and reorganization. OAP area was not altered by anti-AQP4 IgG autoantibodies (NMO-IgG) that cause the neurological disease neuromyelitis optica. Super-resolution imaging allowed elucidation of novel nanoscale structural and dynamic features of OAPs.


Asunto(s)
Acuaporina 4/química , Acuaporina 4/metabolismo , Membrana Celular/metabolismo , Imagenología Tridimensional/métodos , Animales , Astrocitos/metabolismo , Células CHO , Muerte Celular , Línea Celular Tumoral , Supervivencia Celular , Análisis por Conglomerados , Cricetinae , Cricetulus , Humanos , Proteínas Luminiscentes/metabolismo , Ratones , Microscopía Fluorescente , Isoformas de Proteínas/metabolismo , Estructura Cuaternaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/metabolismo
20.
Acta Neuropathol ; 123(6): 861-72, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22526022

RESUMEN

The pathogenesis of neuromyelitis optica (NMO) involves targeting of NMO-immunoglobulin G (NMO-IgG) to aquaporin-4 (AQP4) on astrocytes in the central nervous system. Prior work provided evidence for complement-dependent cytotoxicity (CDC) in NMO lesion development. Here, we show that antibody-dependent cellular cytotoxicity (ADCC), in the absence of complement, can also produce NMO-like lesions. Antibody-dependent cellular cytotoxicity was produced in vitro by incubation of mouse astrocyte cultures with human recombinant monoclonal NMO-IgG and human natural killer cells (NK-cells). Injection of NMO-IgG and NK-cells in mouse brain caused loss of AQP4 and GFAP, two characteristic features of NMO lesions, but little myelin loss. Lesions were minimal or absent following injection of: (1) control (non-NMO) IgG with NK-cells; (2) NMO-IgG and NK-cells in AQP4-deficient mice; or (3) NMO-IgG and NK-cells in wild-type mice together with an excess of mutated NMO-IgG lacking ADCC effector function. NK-cells greatly exacerbated NMO lesions produced by NMO-IgG and complement in an ex vivo spinal cord slice model of NMO, causing marked myelin loss. NMO-IgG can thus produce astrocyte injury by ADCC in a complement-independent and dependent manner, suggesting the potential involvement of ADCC in NMO pathogenesis.


Asunto(s)
Acuaporina 4/metabolismo , Células Asesinas Naturales/inmunología , Vaina de Mielina/metabolismo , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/patología , Animales , Anticuerpos Monoclonales/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos , Acuaporina 4/deficiencia , Astrocitos/inmunología , Astrocitos/metabolismo , Células Cultivadas , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina G/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Noqueados , Vaina de Mielina/patología , Neuromielitis Óptica/genética , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...