Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Res Notes ; 16(1): 348, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38007440

RESUMEN

OBJECTIVES: Animal models of skin disease are used to evaluate therapeutics to alleviate disease. One common clinical dermatological complaint is pruritus (itch), but there is a lack of standardization in the characterization of pre-clinical models and scratching behavior, a key itch endpoint, is often neglected. One such model is the widely used imiquimod (IMQ) mouse model of psoriasis. However, it lacks characterized behavioral attributes like scratching, nor has widely expanded to other species like rats. Given these important attributes, this study was designed to broaden the characterization beyond the expected IMQ-induced psoriasis-like skin inflammatory skin changes and to validate the role of a potential therapeutic agent for pruritus in our genetic rat model. The study included female Wistar rats and genetically modified knockin (humanized proteinase-activated receptor 2 (F2RL1) female rats, with the widely used C57BL/6 J mice as a methodology control for typical IMQ dosing. RESULTS: We demonstrate that the IMQ model can be reproduced in rats, including their genetically modified derivatives, and how scratching can be used as a key behavioral endpoint. We systemically delivered an anti-PAR2 antibody (P24E1102) which reversed scratching bouts-validating this behavioral methodology and have shown its feasibility and value in identifying effective antipruritic drugs.


Asunto(s)
Antipruriginosos , Psoriasis , Ratones , Ratas , Femenino , Animales , Antipruriginosos/farmacología , Antipruriginosos/uso terapéutico , Imiquimod/efectos adversos , Ratas Wistar , Ratones Endogámicos C57BL , Prurito/inducido químicamente , Prurito/tratamiento farmacológico , Prurito/genética , Piel , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Modelos Animales de Enfermedad
2.
MAbs ; 15(1): 2229098, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37381177

RESUMEN

The blood-brain barrier (BBB) largely excludes antibodies from entering the central nervous system, thus limiting the potential of therapeutic antibodies to treat conditions such as neurodegenerative diseases and neuro-psychiatric disorders. Here, we demonstrate that the transport of human antibodies across the BBB in mice can be enhanced by modulating their interactions with the neonatal Fc receptor (FcRn). When M252Y/S254T/T246E substitutions are introduced on the antibody Fc domain, immunohistochemical assays reveal widespread distribution of the engineered antibodies throughout the mouse brain. These engineered antibodies remain specific for their antigens and retain pharmacological activity. We propose that novel brain-targeted therapeutic antibodies can be engineered to differentially engage FcRn for receptor-mediated transcytosis across the BBB in order to improve neurological disease therapeutics in the future.


Asunto(s)
Anticuerpos , Barrera Hematoencefálica , Animales , Humanos , Ratones , Encéfalo , Transcitosis
3.
BMC Neurosci ; 23(1): 30, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35614392

RESUMEN

BACKGROUND: Therapeutic agents stimulating the process of myelination could be beneficial for the treatment of demyelinating diseases, such as multiple sclerosis. The efficient translation of compounds promoting myelination in vitro to efficacy in vivo is inherently time-consuming and expensive. Thyroid hormones accelerate the differentiation and maturation of oligodendrocytes, thereby promoting myelination. Systemic administration of the thyroid hormone thyroxine (T4) accelerates brain maturation, including myelination, during early postnatal development. The objective of this study was to validate an animal model for rapid testing of promyelinating therapeutic candidates for their effects on early postnatal development by using T4 as a reference compound. METHODS: Daily subcutaneous injections of T4 were given to Sprague Dawley rat pups from postnatal day (PND) 2 to PND10. Changes in white matter were determined at PND10 using diffusion tensor magnetic resonance imaging (DTI). Temporal changes in myelination from PND3 to PND11 were also assessed by quantifying myelin basic protein (MBP) expression levels in the brain using the resonance Raman spectroscopy/enzyme-linked immunosorbent assay (RRS-ELISA) and quantitative immunohistochemistry. RESULTS: DTI of white matter tracts showed significantly higher fractional anisotropy in the internal capsule of T4-treated rat pups. The distribution of total FA values in the forebrain was significantly shifted towards higher values in the T4-treated group, suggesting increased myelination. In vivo imaging data were supported by in vitro observations, as T4 administration significantly potentiated the developmental increase in MBP levels in brain lysates starting from PND8. MBP levels in the brain of animals that received treatment for 9 days correlated with the FA metric determined in the same pups in vivo a day earlier. Furthermore, accelerated developmental myelination following T4 administration was confirmed by immunohistochemical staining for MBP in coronal brain sections of treated rat pups. CONCLUSIONS: T4-treated rat pups had increased MBP expression levels and higher MRI fractional anisotropy values, both indications of accelerated myelination. This simple developmental myelination model affords a rapid test of promyelinating activity in vivo within several days, which could facilitate in vivo prescreening of candidate therapeutic compounds for developmental hypomyelinating diseases. Further research will be necessary to assess the utility of this platform for screening promyelination compounds in more complex demyelination disease models, such us multiple sclerosis.


Asunto(s)
Esclerosis Múltiple , Sustancia Blanca , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Esclerosis Múltiple/metabolismo , Oligodendroglía/metabolismo , Ratas , Ratas Sprague-Dawley , Sustancia Blanca/patología
4.
Neurotherapeutics ; 16(3): 808-827, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30815844

RESUMEN

The development of neuroprotective therapies is a sought-after goal. By screening combinatorial chemical libraries using in vitro assays, we identified the small molecule BN201 that promotes the survival of cultured neural cells when subjected to oxidative stress or when deprived of trophic factors. Moreover, BN201 promotes neuronal differentiation, the differentiation of precursor cells to mature oligodendrocytes in vitro, and the myelination of new axons. BN201 modulates several kinases participating in the insulin growth factor 1 pathway including serum-glucocorticoid kinase and midkine, inducing the phosphorylation of NDRG1 and the translocation of the transcription factor Foxo3 to the cytoplasm. In vivo, BN201 prevents axonal and neuronal loss, and it promotes remyelination in models of multiple sclerosis, chemically induced demyelination, and glaucoma. In summary, we provide a new promising strategy to promote neuroaxonal survival and remyelination, potentially preventing disability in brain diseases.


Asunto(s)
Amidas/uso terapéutico , Axones/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Vaina de Mielina/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Peptoides/uso terapéutico , Pirrolidinonas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Técnica del Anticuerpo Fluorescente , Glaucoma/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Nervio Óptico/efectos de los fármacos , Proguanil , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Triazinas
5.
Methods Mol Biol ; 1791: 179-192, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30006710

RESUMEN

The formation of new myelin in persistent multiple sclerosis (MS) lesions is compromised, leading to a reduction in neuron function and subject to degeneration and death. Current MS therapies can control autoimmune-mediated demyelination, but none directly promote the regeneration of myelin in the central nervous system (CNS). To identify new drugs that stimulate remyelination, we established a high-throughput cell-based assay to identify compounds that promote myelination. Methods were developed for initiating myelination in vitro using a preparation of primary embryonic rat cortical cells. We developed an immunofluorescent phenotypic image analysis method to quantify the morphological alignment of myelin characteristic of the initiation of myelination. The assay scalability and consistency was validated by screening the NIH clinical collection library of 727 compounds and identified ten compounds that promote myelination (Lariosa-Willingham et al., BMC Neurosci 17:16, 2016). Here, we present the detailed methods for a high capacity in vitro assay that assesses myelination of live axons.


Asunto(s)
Axones , Sistema Nervioso Central/fisiología , Ensayos Analíticos de Alto Rendimiento , Vaina de Mielina/fisiología , Neurogénesis , Animales , Encéfalo/metabolismo , Técnicas de Cultivo de Célula , Descubrimiento de Drogas , Inmunohistoquímica/métodos , Imagen Molecular/métodos , Vaina de Mielina/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Oligodendroglía/fisiología , Ratas
6.
Curr Protoc Cell Biol ; 79(1): e49, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29924487

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease that involves an immune-mediated inflammatory response in the central nervous system and optic nerve resulting in demyelination and neural degeneration, the cause of which is unknown. The adult central nervous system has the capacity to remyelinate axons by generating new oligodendrocytes (OLs). To identify clinical candidate compounds that may promote remyelination, we have developed a high-throughput screening (HTS) assay to identify compounds that promote the differentiation of oligodendrocyte precursor cells (OPCs) into OLs. Using acutely dissociated and purified rat OPCs coupled with immunofluorescent image quantification, we have developed an OL differentiation assay. Building on OPC culturing techniques developed over the past 30 years, we have scaled up the isolation and purification process to generate sufficient quantities for HTS. We then describe the use of these acutely derived OPCs in an assay designed to identify compounds that promote differentiation into OLs. We have validated this assay with a known promoter of differentiation, thyroid hormone, and subsequently used the assay to screen the NIH clinical collection library (Lariosa-Willingham, et al., 2016). © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Diferenciación Celular , Separación Celular/métodos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/citología , Animales , Disección , Proteína Básica de Mielina/metabolismo , Oxígeno/farmacología , Perfusión , Ratas Sprague-Dawley , Tripsina/metabolismo
7.
Mediators Inflamm ; 2017: 8302636, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28473732

RESUMEN

Glial activation and subsequent release of neurotoxic proinflammatory factors are believed to play an important role in the pathogenesis of several neurological disorders including Parkinson's disease (PD). Inhibition of glial activation and inflammatory processes may represent a therapeutic target to alleviate neurodegeneration. Securinine, a major natural alkaloid product from the root of the plant Securinega suffruticosa, has been reported to have potent biological activity and is used in the treatment of neurological conditions such as amyotrophic lateral sclerosis, poliomyelitis, and multiple sclerosis. In this study, we explored the underlying mechanisms of neuroprotection elicited by securinine, particularly its anti-inflammatory effects in glial cells. Our results demonstrate that securinine significantly and dose-dependently suppressed the nitric oxide production in microglia and astrocytic cultures. In addition, securinine inhibited the activation of the inflammatory mediator NF-κB, as well as mitogen-activated protein kinases in lipopolysaccharide- (LPS-) stimulated BV2 cells. Additionally, securinine also inhibited interferon-γ- (IFN-γ-) induced nitric oxide levels and iNOS mRNA expression. Furthermore, conditioned media (CM) from securinine pretreated BV2 cells significantly reduced mesencephalic dopaminergic neurotoxicity compared with CM from LPS stimulated microglia. These findings suggest that securinine may be a potential candidate for the treatment of neurodegenerative diseases related to neuroinflammation.


Asunto(s)
Azepinas/uso terapéutico , Compuestos Heterocíclicos de Anillo en Puente/uso terapéutico , Lactonas/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Piperidinas/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Astrocitos/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Enfermedad de Parkinson/inmunología , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa
8.
BMC Res Notes ; 9(1): 444, 2016 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-27629829

RESUMEN

BACKGROUND: Newly proliferated oligodendrocyte precursor cells (OPCs) migrate and surround lesions of patients with multiple sclerosis (MS) and other demyelinating diseases, but fail to differentiate into oligodendrocytes (OLs) and remyelinate remaining viable axons. The abundance of secreted inflammatory factors within and surrounding these lesions likely plays a major inhibitory role, promoting cell death and preventing OL differentiation and axon remyelination. To identify clinical candidate compounds that may protect existing and differentiating OLs in patients, we have developed a high throughput screening (HTS) assay that utilizes purified rat OPCs. RESULTS: Using a fluorescent indicator of cell viability coupled with image quantification, we developed an assay to allow the identification of compounds that promote OL viability and differentiation in the presence of the synergistic inflammatory cytokines, tumor necrosis factor α and interferon-γ. We have utilized this assay to screen the NIH clinical collection library and identify compounds that protect OLs and promote OL differentiation in the presence of these inflammatory cytokines. CONCLUSION: This primary OL-based cytokine protection assay is adaptable for HTS and may be easily modified for profiling of compounds in the presence of other potentially inhibitory molecules found in MS lesions. This assay should be of use to those interested in identifying drugs for the treatment of MS and other demyelinating diseases.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Oligodendroglía/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Inflamación , Interferón gamma/metabolismo , Masculino , Esclerosis Múltiple/patología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
9.
BMC Res Notes ; 9(1): 419, 2016 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-27592856

RESUMEN

BACKGROUND: Multiple sclerosis is caused by an autoimmune response resulting in demyelination and neural degeneration. The adult central nervous system has the capacity to remyelinate axons in part through the generation of new oligodendrocytes (OLs). To identify clinical candidate compounds that may promote remyelination, we have developed a high throughput screening (HTS) assay to identify compounds that promote the differentiation of oligodendrocyte precursor cells (OPCs) into OLs. RESULTS: Using acutely dissociated and purified rat OPCs coupled with immunofluorescent image quantification, we have developed an OL differentiation assay. We have validated this assay with a known promoter of differentiation, thyroid hormone, and subsequently used the assay to screen the NIH clinical collection library. We have identified twenty-seven hit compounds which were validated by dose response analysis and the generation of half maximal effective concentration (EC50) values allowed for the ranking of efficacy. The assay identified novel promoters of OL differentiation which we attribute to (1) the incorporation of an OL toxicity pre-screen to allow lowering the concentrations of toxic compounds and (2) the utilization of freshly purified, non-passaged OPCs. These features set our assay apart from other OL differentiation assays used for drug discovery efforts. CONCLUSIONS: This acute primary OL-based differentiation assay should be of use to those interested in screening large compound libraries for the identification of drugs for the treatment of MS and other demyelinating diseases.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Técnicas In Vitro , Esclerosis Múltiple/tratamiento farmacológico , Ratas , Células Madre/citología
10.
BMC Neurosci ; 17: 16, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-27103572

RESUMEN

BACKGROUND: Regeneration of new myelin is impaired in persistent multiple sclerosis (MS) lesions, leaving neurons unable to function properly and subject to further degeneration. Current MS therapies attempt to ameliorate autoimmune-mediated demyelination, but none directly promote the regeneration of lost and damaged myelin of the central nervous system (CNS). Development of new drugs that stimulate remyelination has been hampered by the inability to evaluate axonal myelination in a rapid CNS culture system. RESULTS: We established a high throughput cell-based assay to identify compounds that promote myelination. Culture methods were developed for initiating myelination in vitro using primary embryonic rat cortical cells. We developed an immunofluorescent phenotypic image analysis method to quantify the morphological alignment of myelin characteristic of the initiation of myelination. Using γ-secretase inhibitors as promoters of myelination, the optimal growth, time course and compound treatment conditions were established in a 96 well plate format. We have characterized the cortical myelination assay by evaluating the cellular composition of the cultures and expression of markers of differentiation over the time course of the assay. We have validated the assay scalability and consistency by screening the NIH clinical collection library of 727 compounds and identified ten compounds that promote myelination. Half maximal effective concentration (EC50) values for these compounds were determined to rank them according to potency. CONCLUSIONS: We have designed the first high capacity in vitro assay that assesses myelination of live axons. This assay will be ideal for screening large compound libraries to identify new drugs that stimulate myelination. Identification of agents capable of promoting the myelination of axons will likely lead to the development of new therapeutics for MS patients.


Asunto(s)
Axones/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Esclerosis Múltiple/tratamiento farmacológico , Vaina de Mielina/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Secretasas de la Proteína Precursora del Amiloide/farmacología , Animales , Axones/fisiología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Corteza Cerebral/fisiología , Medios de Cultivo Condicionados/farmacología , Técnica del Anticuerpo Fluorescente/métodos , Esclerosis Múltiple/fisiopatología , Vaina de Mielina/fisiología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Ratas
11.
Dev Cell ; 34(2): 152-67, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26166300

RESUMEN

Myelin is essential in vertebrates for the rapid propagation of action potentials, but the molecular mechanisms driving its formation remain largely unknown. Here we show that the initial stage of process extension and axon ensheathment by oligodendrocytes requires dynamic actin filament assembly by the Arp2/3 complex. Unexpectedly, subsequent myelin wrapping coincides with the upregulation of actin disassembly proteins and rapid disassembly of the oligodendrocyte actin cytoskeleton and does not require Arp2/3. Inducing loss of actin filaments drives oligodendrocyte membrane spreading and myelin wrapping in vivo, and the actin disassembly factor gelsolin is required for normal wrapping. We show that myelin basic protein, a protein essential for CNS myelin wrapping whose role has been unclear, is required for actin disassembly, and its loss phenocopies loss of actin disassembly proteins. Together, these findings provide insight into the molecular mechanism of myelin wrapping and identify it as an actin-independent form of mammalian cell motility.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Complejo 2-3 Proteico Relacionado con la Actina/genética , Actinas/metabolismo , Animales , Axones/fisiología , Membrana Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/embriología , Cofilina 1/genética , Gelsolina/genética , Gelsolina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Nervio Óptico/metabolismo , Nervio Óptico/fisiología , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley
12.
Mol Pharmacol ; 88(2): 357-67, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26018904

RESUMEN

Erythropoietin (EPO) and its receptor are expressed in a wide variety of tissues, including the central nervous system. Local expression of both EPO and its receptor is upregulated upon injury or stress and plays a role in tissue homeostasis and cytoprotection. High-dose systemic administration or local injection of recombinant human EPO has demonstrated encouraging results in several models of tissue protection and organ injury, while poor tissue availability of the protein limits its efficacy. Here, we describe the discovery and characterization of the nonpeptidyl compound STS-E412 (2-[2-(4-chlorophenoxy)ethoxy]-5,7-dimethyl-[1,2,4]triazolo[1,5-a]pyrimidine), which selectively activates the tissue-protective EPO receptor, comprising an EPO receptor subunit (EPOR) and the common ß-chain (CD131). STS-E412 triggered EPO receptor phosphorylation in human neuronal cells. STS-E412 also increased phosphorylation of EPOR, CD131, and the EPO-associated signaling molecules JAK2 and AKT in HEK293 transfectants expressing EPOR and CD131. At low nanomolar concentrations, STS-E412 provided EPO-like cytoprotective effects in primary neuronal cells and renal proximal tubular epithelial cells. The receptor selectivity of STS-E412 was confirmed by a lack of phosphorylation of the EPOR/EPOR homodimer, lack of activity in off-target selectivity screening, and lack of functional effects in erythroleukemia cell line TF-1 and CD34(+) progenitor cells. Permeability through artificial membranes and Caco-2 cell monolayers in vitro and penetrance across the blood-brain barrier in vivo suggest potential for central nervous system availability of the compound. To our knowledge, STS-E412 is the first nonpeptidyl, selective activator of the tissue-protective EPOR/CD131 receptor. Further evaluation of the potential of STS-E412 in central nervous system diseases and organ protection is warranted.


Asunto(s)
Encéfalo/embriología , Eritropoyetina/metabolismo , Neuronas/metabolismo , Pirimidinas/farmacología , Pirimidinas/farmacocinética , Receptores de Eritropoyetina/agonistas , Triazoles/farmacología , Triazoles/farmacocinética , Animales , Disponibilidad Biológica , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/citología , Células CACO-2 , Células Cultivadas , Subunidad beta Común de los Receptores de Citocinas/metabolismo , Células HEK293 , Humanos , Ratas , Transducción de Señal/efectos de los fármacos
13.
J Cell Sci ; 127(Pt 22): 4894-903, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25217627

RESUMEN

The dynamic interactions between cells and basement membranes serve as essential regulators of tissue architecture and function in metazoans, and perturbation of these interactions contributes to the progression of a wide range of human diseases, including cancers. Here, we reveal the pathway and mechanism for the endocytic trafficking of a prominent basement membrane protein, laminin-111 (referred to here as laminin), and their disruption in disease. Live-cell imaging of epithelial cells revealed pronounced internalization of laminin into endocytic vesicles. Laminin internalization was receptor mediated and dynamin dependent, and laminin proceeded to the lysosome through the late endosome. Manipulation of laminin receptor expression revealed that the dominant regulator of laminin internalization is dystroglycan, a laminin receptor that is functionally perturbed in muscular dystrophies and in many cancers. Correspondingly, laminin internalization was found to be deficient in aggressive cancer cells displaying non-functional dystroglycan, and restoration of dystroglycan function strongly enhanced the endocytosis of laminin in both breast cancer and glioblastoma cells. These results establish previously unrecognized mechanisms for the modulation of cell-basement-membrane communication in normal cells and identify a profound disruption of endocytic laminin trafficking in aggressive cancer subtypes.


Asunto(s)
Distroglicanos/metabolismo , Laminina/metabolismo , Neoplasias/metabolismo , Animales , Membrana Basal/metabolismo , Endocitosis , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Ratones , Ratones Noqueados , Embarazo
14.
Cancer Res ; 72(10): 2578-88, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22589276

RESUMEN

Perturbations in the composition and assembly of extracellular matrices (ECM) contribute to progression of numerous diseases, including cancers. Anchoring of laminins at the cell surface enables assembly and signaling of many ECMs, but the possible contributions of altered laminin anchoring to cancer progression remain undetermined. In this study, we investigated the prominence and origins of defective laminin anchoring in cancer cells and its association with cancer subtypes and clinical outcomes. We found loss of laminin anchoring to be widespread in cancer cells. Perturbation of laminin anchoring originated from several distinct defects, which all led to dysfunctional glycosylation of the ECM receptor dystroglycan. In aggressive breast and brain cancers, defective laminin anchoring was often due to suppressed expression of the glycosyltransferase LARGE. Reduced expression of LARGE characterized a broad array of human tumors in which it was associated with aggressive cancer subtypes and poor clinical outcomes. Notably, this defect robustly predicted poor survival in patients with brain cancers. Restoring LARGE expression repaired anchoring of exogenous and endogenous laminin and modulated cell proliferation and tumor growth. Together, our findings suggest that defects in laminin anchoring occur commonly in cancer cells, are characteristic of aggressive cancer subtypes, and are important drivers of disease progression.


Asunto(s)
Membrana Celular/metabolismo , Laminina/metabolismo , Neoplasias/metabolismo , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Distroglicanos/metabolismo , Matriz Extracelular/metabolismo , Glicosilación , Humanos , N-Acetilglucosaminiltransferasas/metabolismo
15.
J Cell Sci ; 123(Pt 21): 3683-92, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20940259

RESUMEN

Receptors for basement membrane (BM) proteins, including dystroglycan (DG), coordinate tissue development and function by mechanisms that are only partially defined. To further elucidate these mechanisms, we generated a conditional knockout of DG in the epithelial compartment of the mouse mammary gland. Deletion of DG caused an inhibition of mammary epithelial outgrowth and a failure of lactation. Surprisingly, loss of DG in vivo did not disrupt normal tissue architecture or BM formation, even though cultured Dag1-null epithelial cells failed to assemble laminin-111 at the cell surface. The absence of DG was, however, associated with a marked loss in activity of signal transducer and activator of transcription 5 (STAT5). Loss of DG perturbed STAT5 signaling induced by either prolactin or growth hormone. We found that DG regulates signaling by both hormones in a manner that is dependent on laminin-111 binding, but independent of the DG cytoplasmic domain, suggesting that it acts via a co-receptor mechanism reliant on DG-mediated laminin assembly. These results demonstrate a requirement for DG in the growth and function of a mammalian epithelial tissue in vivo. Moreover, we reveal a selective role for DG in the control of multiple STAT5-dependent hormone signaling pathways, with implications for numerous diseases in which DG function is compromised.


Asunto(s)
Membrana Basal/metabolismo , Distroglicanos/metabolismo , Laminina/biosíntesis , Glándulas Mamarias Animales/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Membrana Basal/crecimiento & desarrollo , Membrana Basal/patología , Distroglicanos/genética , Epitelio/patología , Femenino , Hormona del Crecimiento/biosíntesis , Hormona del Crecimiento/genética , Lactancia/genética , Laminina/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/genética , Embarazo , Prolactina/metabolismo , Unión Proteica/genética , Factor de Transcripción STAT5/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
16.
Neuropharmacology ; 58(2): 551-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19654014

RESUMEN

After injury or during neurodegenerative disease in the central nervous system (CNS), the concentration of tumor necrosis factor alpha (TNFalpha) rises above normal during the inflammatory response. In vitro and in vivo, addition of exogenous TNFalpha to neurons has been shown to induce rapid plasma membrane-delivery of AMPA-type glutamate receptors (AMPARs) potentiating glutamatergic excitotoxicity. Thus the discovery of drug targets reducing excess TNFalpha-induced AMPAR surface expression may help protect neurons after injury. In this study, we investigate the neuroprotective role of the CB1 cannabinoid receptor using quantitative immunofluorescent and real-time video microscopy to measure the steady-state plasma membrane AMPAR distribution and rate of AMPAR exocytosis after TNFalpha exposure in the presence or absence of CB1 agonists. The neuroprotective potential of CB1 activation with TNFalpha was measured in hippocampal neuron cultures challenged by an in vitro kainate (KA)-mediated model of Excitotoxic Neuroinflammatory Death (END). Here, we demonstrate that CB1 activation blocks the TNFalpha-induced increase in surface AMPARs and protects neurons from END. Thus, neuroprotective strategies which increase CB1 activity may help to reduce the END that occurs as a result of a majority of CNS insults.


Asunto(s)
Membrana Celular/fisiología , Hipocampo/fisiología , Neuronas/fisiología , Receptores AMPA/metabolismo , Receptores de Cannabinoides/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Benzoxazinas/farmacología , Agonistas de Receptores de Cannabinoides , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Membrana Celular/efectos de los fármacos , Células Cultivadas , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Hipocampo/efectos de los fármacos , Ácido Kaínico/toxicidad , Morfolinas/farmacología , Naftalenos/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Neurotoxinas/toxicidad , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Factores de Tiempo , Grabación en Video
17.
J Neurosci ; 28(9): 2119-30, 2008 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-18305246

RESUMEN

The postinjury inflammatory response in the CNS leads to neuronal excitotoxicity. Our previous studies show that a major component of this response, the inflammatory cytokine tumor necrosis factor alpha (TNFalpha), causes a rapid increase in AMPA glutamate receptors (AMPARs) on the plasma membrane of cultured hippocampal neurons. This may potentiate neuron death through an increased vulnerability to AMPAR-dependent excitotoxic stress. Here, we test this hypothesis with an in vitro lactose dehydrogenase death assay and examine in detail the AMPAR surface delivery time course, receptor subtype, and synaptic and extrasynaptic distribution after TNFalpha exposure. These data demonstrate that surface levels of glutamate receptor 2 (GluR2)-lacking Ca2+-permeable AMPARs peak at 15 min after TNFalpha treatment, and the majority are directed to extrasynaptic sites. TNFalpha also induces an increase in GluR2-containing surface AMPARs but with a slower time course. We propose that this activity contributes to excitotoxic neuron death because TNFalpha potentiation of kainate excitotoxicity is blocked by a Ca2+-permeable AMPAR antagonist [NASPM (1-naphthyl acetyl spermine)] and a specific phosphoinositide 3 kinase (PI3 kinase) inhibitor (LY294,002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one]) previously shown to block the TNFalpha-induced increase in AMPAR surface delivery. This information forms the basis for future in vivo studies examining AMPAR-dependent potentiation of excitotoxic neuron death and dysfunction caused by TNFalpha after acute injury and during neurodegenerative or neuropsychiatric disorders.


Asunto(s)
Exocitosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores AMPA/metabolismo , Sinapsis/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Biotinilación/métodos , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Células Cultivadas , Interacciones Farmacológicas , Embrión de Mamíferos , Inhibidores Enzimáticos , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/citología , Inmunoprecipitación/métodos , Microscopía Confocal/métodos , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
18.
J Neurosci ; 27(41): 11112-21, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17928453

RESUMEN

We directly resolved discrete exocytic fusion events mediating insertion of AMPA-type glutamate receptors (AMPARs) to the somatodendritic surface of rat hippocampal pyramidal neurons, in slice and dissociated cultures, using protein tagging with a pH-sensitive GFP (green fluorescent protein) variant and rapid (10 frames/s) fluorescence microscopy. AMPAR-containing exocytic events occurred under basal culture conditions in both the cell body and dendrites; potentiating chemical stimuli produced an NMDA receptor-dependent increase in the frequency of individual exocytic events. The number of AMPARs inserted per exocytic event, estimated using single-molecule analysis, was quite uniform but individual events differed significantly in kinetic properties affecting the subsequent surface distribution of receptors. "Transient" events, from which AMPARs dispersed laterally immediately after surface insertion, generated a pronounced but short-lived (dissipating within approximately 1 s) increase in surface AMPAR fluorescence extending locally (2-5 microm) from the site of exocytosis. "Persistent" events, from which inserted AMPARs dispersed slowly (typically over 5-10 s), affected local surface receptor concentration to a much smaller degree. Both modes of exocytic insertion occurred throughout the dendritic shaft, but remarkably, neither mode of insertion was observed directly into synaptic spines. AMPARs entered spines preferentially from transient events occurring in the adjoining dendritic shaft, driven apparently by mass action and short-range lateral diffusion, and locally delivered AMPARs remained mostly in the mobile fraction. These results suggest a highly dynamic mechanism for both constitutive and activity-dependent surface delivery of AMPARs, mediated by kinetically distinct exocytic modes that differ in propensity to drive lateral entry of receptors to nearby synapses.


Asunto(s)
Membrana Celular/fisiología , Sistemas de Computación , Potenciales Postsinápticos Excitadores/fisiología , Exocitosis/fisiología , Receptores AMPA/fisiología , Animales , Animales Recién Nacidos , Hipocampo/fisiología , Técnicas de Cultivo de Órganos , Ratas , Receptores de Superficie Celular/fisiología
19.
J Biol Chem ; 279(21): 22331-46, 2004 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-15024025

RESUMEN

Inward rectifier potassium (Kir) channels play important roles in the maintenance and control of cell excitability. Both intracellular trafficking and modulation of Kir channel activity are regulated by protein-protein interactions. We adopted a proteomics approach to identify proteins associated with Kir2 channels via the channel C-terminal PDZ binding motif. Detergent-solubilized rat brain and heart extracts were subjected to affinity chromatography using a Kir2.2 C-terminal matrix to purify channel-interacting proteins. Proteins were identified with multidimensional high pressure liquid chromatography coupled with electrospray ionization tandem mass spectrometry, N-terminal microsequencing, and immunoblotting with specific antibodies. We identified eight members of the MAGUK family of proteins (SAP97, PSD-95, Chapsyn-110, SAP102, CASK, Dlg2, Dlg3, and Pals2), two isoforms of Veli (Veli-1 and Veli-3), Mint1, and actin-binding LIM protein (abLIM) as Kir2.2-associated brain proteins. From heart extract purifications, SAP97, CASK, Veli-3, and Mint1 also were found to associate with Kir2 channels. Furthermore, we demonstrate for the first time that components of the dystrophin-associated protein complex, including alpha1-, beta1-, and beta2-syntrophin, dystrophin, and dystrobrevin, interact with Kir2 channels, as demonstrated by immunoaffinity purification and affinity chromatography from skeletal and cardiac muscle and brain. Affinity pull-down experiments revealed that Kir2.1, Kir2.2, Kir2.3, and Kir4.1 all bind to scaffolding proteins but with different affinities for the dystrophin-associated protein complex and SAP97, CASK, and Veli. Immunofluorescent localization studies demonstrated that Kir2.2 co-localizes with syntrophin, dystrophin, and dystrobrevin at skeletal muscle neuromuscular junctions. These results suggest that Kir2 channels associate with protein complexes that may be important to target and traffic channels to specific subcellular locations, as well as anchor and stabilize channels in the plasma membrane.


Asunto(s)
Proteínas Asociadas a la Distrofina , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Western Blotting , Encéfalo/embriología , Encéfalo/metabolismo , Células COS , Membrana Celular/metabolismo , Cerebelo/metabolismo , Cromatografía de Afinidad , Cromatografía Líquida de Alta Presión , ADN Complementario/metabolismo , Detergentes/farmacología , Glutatión Transferasa/metabolismo , Guanilato-Quinasas , Immunoblotting , Espectrometría de Masas , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Péptidos/química , Pruebas de Precipitina , Isoformas de Proteínas , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteoma , Proteómica/métodos , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Tinción con Nitrato de Plata , Espectrometría de Masa por Ionización de Electrospray
20.
J Biol Chem ; 279(18): 19051-63, 2004 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-14960569

RESUMEN

Strong inward rectifier potassium (Kir2) channels are important in the control of cell excitability, and their functions are modulated by interactions with intracellular proteins. Here we identified a complex of scaffolding/trafficking proteins in brain that associate with Kir2.1, Kir2.2, and Kir2.3 channels. By using a combination of affinity interaction pulldown assays and co-immunoprecipitations from brain and transfected cells, we demonstrated that a complex composed of SAP97, CASK, Veli, and Mint1 associates with Kir2 channels via the C-terminal PDZ-binding motif. We further demonstrated by using in vitro protein interaction assays that SAP97, Veli-1, or Veli-3 binds directly to the Kir2.2 C terminus and recruits CASK. Co-immunoprecipitations indicated that specific Veli isoforms participate in forming distinct protein complexes in brain, where Veli-1 stably associates with CASK and SAP97, Veli-2 associates with CASK and Mint1, and Veli-3 associates with CASK, SAP97, and Mint1. Additionally, immunocytochemistry of rat cerebellum revealed overlapping expression of Kir2.2, SAP97, CASK, Mint1, with Veli-1 in the granule cell layer and Veli-3 in the molecular layer. We propose a model whereby Kir2.2 associates with distinct SAP97-CASK-Veli-Mint1 complexes. In one complex, SAP97 interacts directly with the Kir2 channels and recruits CASK, Veli, and Mint1. Alternatively, Veli-1 or Veli-3 interacts directly with the Kir2 channels and recruits CASK and SAP97; association of Mint1 with the complex requires Veli-3. Expression of Kir2.2 in polarized epithelial cells resulted in targeting of the channels to the basolateral membrane and co-localization with SAP97 and CASK, whereas a dominant interfering form of CASK caused the channels to mislocalize. Therefore, CASK appears to be a central protein of a macromolecular complex that participates in trafficking and plasma membrane localization of Kir2 channels.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Polaridad Celular , Cerebelo/química , Homólogo 1 de la Proteína Discs Large , Células Epiteliales/metabolismo , Glutatión Transferasa , Guanilato-Quinasas , Corazón , Sustancias Macromoleculares , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos , Unión Proteica , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Ratas , Proteínas Recombinantes de Fusión/aislamiento & purificación , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...