Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 40(31): 5908-5921, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32601248

RESUMEN

SORLA is a transmembrane trafficking protein associated with Alzheimer's disease risk. Although SORLA is abundantly expressed in neurons, physiological roles for SORLA remain unclear. Here, we show that cultured transgenic neurons overexpressing SORLA feature longer neurites, and accelerated neurite regeneration with wounding. Enhanced release of a soluble form of SORLA (sSORLA) is observed in transgenic mouse neurons overexpressing human SORLA, while purified sSORLA promotes neurite extension and regeneration. Phosphoproteomic analyses demonstrate enrichment of phosphoproteins related to the epidermal growth factor (EGFR)/ERK pathway in SORLA transgenic mouse hippocampus from both genders. sSORLA coprecipitates with EGFR in vitro, and sSORLA treatment increases EGFR Y1173 phosphorylation, which is involved in ERK activation in cultured neurons. Furthermore, sSORLA triggers ERK activation, whereas pharmacological EGFR or ERK inhibition reverses sSORLA-dependent enhancement of neurite outgrowth. In search for downstream ERK effectors activated by sSORLA, we identified upregulation of Fos expression in hippocampus from male mice overexpressing SORLA by RNAseq analysis. We also found that Fos is upregulated and translocates to the nucleus in an ERK-dependent manner in neurons treated with sSORLA. Together, these results demonstrate that sSORLA is an EGFR-interacting protein that activates EGFR/ERK/Fos signaling to enhance neurite outgrowth and regeneration.SIGNIFICANCE STATEMENT SORLA is a transmembrane trafficking protein previously known to reduce the levels of amyloid-ß, which is critical in the pathogenesis of Alzheimer's disease. In addition, SORLA mutations are a risk factor for Alzheimer's disease. Interestingly, the SORLA ectodomain is cleaved into a soluble form, sSORLA, which has been shown to regulate cytoskeletal signaling pathways and cell motility in cells outside the nervous system. We show here that sSORLA binds and activates the EGF receptor to induce downstream signaling through the ERK serine/threonine kinase and the Fos transcription factor, thereby enhancing neurite outgrowth. These findings reveal a novel role for sSORLA in promoting neurite regeneration through the EGF receptor/ERK/Fos pathway, thereby demonstrating a potential neuroprotective mechanism involving SORLA.


Asunto(s)
Receptores ErbB/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de Transporte de Membrana/fisiología , Regeneración Nerviosa/fisiología , Neuritas/fisiología , Receptores de LDL/fisiología , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Genes fos , Hipocampo/fisiología , Masculino , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fosforilación , Receptores de LDL/genética
2.
Am J Physiol Endocrinol Metab ; 320(2): E179-E190, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284092

RESUMEN

Adiponectin (APN) is a circulating protein specifically produced by adipocytes. Native APN specifically binds to T-cadherin, a glycosylphosphatidylinositol-anchored protein, mediating the exosome-stimulating effects of APN in endothelial, muscle, and mesenchymal stem cells. It was previously reported that APN has beneficial effects on kidney diseases, but the role of T-cadherin has not been clarified yet. Here, our immunofluorescence study indicated the existence of both T-cadherin and APN protein in pericytes, subsets of tissue-resident mesenchymal stem/progenitor cells positive for platelet-derived growth factor receptor ß (PDGFRß), surrounding peritubular capillaries. In an acute renal ischemia-reperfusion (I/R) model, T-cadherin-knockout (Tcad-KO) mice, similar to APN-KO mice, exhibited the more progressive phenotype of renal tubular damage and increased vascular permeability than wild-type mice. In addition, in response to I/R-injury, the renal PDGFRß-positive cell area increased in wild-type mice, but opposingly decreased in both Tcad-KO and APN-KO mice, suggesting severe pericyte loss. Mouse primary pericytes also expressed T-cadherin. APN promoted exosome secretion in a T-cadherin-dependent manner. Such exosome production from pericytes may play an important role in maintaining the capillary network and APN-mediated inhibition of renal tubular injury. In summary, our study suggested that APN protected the kidney in an acute renal injury model by binding to T-cadherin.NEW & NOTEWORTHY In the kidney, T-cadherin-associated adiponectin protein existed on peritubular capillary pericytes. In an acute renal ischemia-reperfusion model, deficiency of adiponectin or T-cadherin exhibited the more progressive phenotype of renal tubular damage and increased vascular permeability, accompanied by severe pericyte loss. In vitro, adiponectin promoted exosome secretion from mouse primary pericytes in a T-cadherin-dependent manner. Adiponectin plays an important role in maintaining the capillary network and amelioration of renal tubular injury by binding to T-cadherin.


Asunto(s)
Adiponectina/genética , Cadherinas/genética , Permeabilidad Capilar/genética , Enfermedades Renales/genética , Daño por Reperfusión/genética , Animales , Células Cultivadas , Enfermedades Renales/etiología , Enfermedades Renales/patología , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/complicaciones , Daño por Reperfusión/patología , Índice de Severidad de la Enfermedad
3.
Glia ; 67(12): 2248-2263, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31328333

RESUMEN

The fast and reliable propagation of action potentials along myelinated fibers relies on the clustering of voltage-gated sodium channels at nodes of Ranvier. Axo-glial communication is required for assembly of nodal proteins in the central nervous system, yet the underlying mechanisms remain poorly understood. Oligodendrocytes are known to support node of Ranvier assembly through paranodal junction formation. In addition, the formation of early nodal protein clusters (or prenodes) along axons prior to myelination has been reported, and can be induced by oligodendrocyte conditioned medium (OCM). Our recent work on cultured hippocampal neurons showed that OCM-induced prenodes are associated with an increased conduction velocity (Freeman et al., 2015). We here unravel the nature of the oligodendroglial secreted factors. Mass spectrometry analysis of OCM identified several candidate proteins (i.e., Contactin-1, ChL1, NrCAM, Noelin2, RPTP/Phosphacan, and Tenascin-R). We show that Contactin-1 combined with RPTP/Phosphacan or Tenascin-R induces clusters of nodal proteins along hippocampal GABAergic axons. Furthermore, Contactin-1-immunodepleted OCM or OCM from Cntn1-null mice display significantly reduced clustering activity, that is restored by addition of soluble Contactin-1. Altogether, our results identify Contactin-1 secreted by oligodendrocytes as a novel factor that may influence early steps of nodal sodium channel cluster formation along specific axon populations.


Asunto(s)
Contactina 1/metabolismo , Hipocampo/metabolismo , Proteína Nodal/metabolismo , Oligodendroglía/metabolismo , Animales , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Contactina 1/genética , Neuronas GABAérgicas/metabolismo , Hipocampo/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Nodal/genética , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Ratas Wistar
4.
FASEB J ; 31(4): 1571-1583, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28062540

RESUMEN

Adiponectin, an adipocyte-derived protein abundant in the circulation, is thought to be protective against atherosclerosis. However, it is not fully understood how the association of adiponectin with vascular cells and its antiatherogenic effect are connected. In this study, T-cadherin was essential for accumulation of adiponectin in the neointima and atherosclerotic plaque lesions, and the adiponectin-T-cadherin association protected against vascular injury. In the apolipoprotein E-knockout (ApoE-KO) mice, adiponectin and T-cadherin colocalized on endothelial cells and synthetic smooth muscle cells in the aortic intima. Notably, aortic adiponectin protein disappeared in T-cadherin/ApoE double-knockout (Tcad/ApoE-DKO) mice with significant elevation of blood adiponectin concentration. Furthermore, in Tcad/ApoE-DKO mice, carotid artery ligation resulted in a significant increase of neointimal thickness compared with ApoE-KO mice. Finally, on a high-cholesterol diet, Tcad/ApoE-DKO mice increased atherosclerotic plaque formation, despite a 5-fold increase in plasma adiponectin level compared with that in ApoE-KO mice. In vitro, knockdown of T-cadherin from human aortic smooth muscle cells (HASMCs) with synthetic phenotype significantly reduced adiponectin accumulation on HASMCs and negated the inhibitory effect of adiponectin on proinflammatory change. Collective evidence showed that adiponectin accumulates in the vasculature via T-cadherin, and the adiponectin-T-cadherin association plays a protective role against neointimal and atherosclerotic plaque formations.-Fujishima, Y., Maeda, N., Matsuda, K., Masuda, S., Mori, T., Fukuda, S., Sekimoto, R., Yamaoka, M., Obata, Y., Kita, S., Nishizawa, H., Funahashi, T., Ranscht, B., Shimomura, I. Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis.


Asunto(s)
Adiponectina/metabolismo , Aterosclerosis/metabolismo , Cadherinas/metabolismo , Adiponectina/sangre , Adiponectina/genética , Animales , Aterosclerosis/patología , Cadherinas/genética , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Túnica Íntima/metabolismo , Túnica Íntima/patología
5.
Proc Natl Acad Sci U S A ; 111(3): E394-403, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24385581

RESUMEN

Myelin, a multilayered membrane sheath formed by oligodendrocytes around axons in the CNS, enables rapid nerve impulse conduction and sustains neuronal health. The signals exchanged between axons and oligodendrocytes in myelin remain to be fully elucidated. Here we provide genetic evidence for multiple and critical functions of Contactin-1 in central myelin. We document dynamic Contactin-1 expression on oligodendrocytes in vivo, and progressive accumulation at nodes of Ranvier and paranodes during postnatal mouse development. Nodal and paranodal expression stabilized in mature myelin, but overall membranous expression diminished. Contactin-1-deficiency disrupted paranodal junction formation as evidenced by loss of Caspr, mislocalized potassium Kv1.2 channels, and abnormal myelin terminal loops. Reduced numbers and impaired maturation of sodium channel clusters accompanied this phenotype. Histological, electron microscopic, and biochemical analyses uncovered significant hypomyelination in Contactin-1-deficient central nerves, with up to 60% myelin loss. Oligodendrocytes were present in normal numbers, albeit a minor population of neuronal/glial antigen 2-positive (NG2(+)) progenitors lagged in maturation by postnatal day 18, when the mouse null mutation was lethal. Major contributing factors to hypomyelination were defects in the generation and organization of myelin membranes, as judged by electron microscopy and quantitative analysis of oligodendrocyte processes labeled by GFP transgenically expressed from the proteolipid protein promoter. These data reveal that Contactin-1 regulates both myelin formation and organization of nodal and paranodal domains in the CNS. These multiple roles distinguish central Contactin-1 functions from its specific role at paranodes in the periphery, and emphasize mechanistic differences in central and peripheral myelination.


Asunto(s)
Sistema Nervioso Central/crecimiento & desarrollo , Contactina 1/fisiología , Regulación del Desarrollo de la Expresión Génica , Vaina de Mielina/metabolismo , Animales , Axones/metabolismo , Diferenciación Celular , Proteínas Fluorescentes Verdes/metabolismo , Canal de Potasio Kv.1.2/metabolismo , Ratones , Ratones Noqueados , Oligodendroglía/citología , Nervio Óptico/metabolismo , Fenotipo , Factores de Tiempo
6.
Mol Med ; 22: 537-547, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27579475

RESUMEN

The cadherin 13 (CDH13) gene encodes a cell adhesion molecule likely to influence development and connections of brain circuits that modulate addiction, locomotion and cognition, including those that involve midbrain dopamine neurons. Human CDH13 mRNA expression differs by more than 80% in postmortem cerebral cortical samples from individuals with different CDH13 genotypes, supporting examination of mice with altered Cdh13 expression as models for common human variation at this locus. Constitutive cdh13 knockout mice display evidence for changed cocaine reward: shifted dose response relationship in tests of cocaine-conditioned place preference using doses that do not alter cocaine conditioned taste aversion. Reduced adult Cdh13 expression in conditional knockouts also alters cocaine reward in ways that correlate with individual differences in cortical Cdh13 mRNA levels. In control and comparison behavioral assessments, knockout mice display modestly-quicker acquisition of rotarod and water maze tasks, with a trend toward faster acquisition of 5 choice serial reaction time tasks that otherwise displayed no genotype-related differences. They display significant differences in locomotion in some settings, with larger effects in males. In assessments of brain changes that might contribute to these behavioral differences, there are selective alterations of dopamine levels, dopamine/metabolite ratios, dopaminergic fiber densities and mRNA encoding the activity dependent transcription factor npas4 in cerebral cortex of knockout mice. These novel data and previously reported human associations of CDH13 variants with addiction, individual differences in responses to stimulant administration and attention deficit hyperactivity disorder (ADHD) phenotypes suggest that levels of CDH13 expression, through mechanisms likely to include effects on mesocortical dopamine, influence stimulant reward and may contribute modestly to cognitive and locomotor phenotypes relevant to ADHD.

7.
J Biol Chem ; 288(34): 24886-97, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23824191

RESUMEN

Adipose tissue secretes protein factors that have systemic actions on cardiovascular tissues. Previous studies have shown that ablation of the adipocyte-secreted protein adiponectin leads to endothelial dysfunction, whereas its overexpression promotes wound healing. However, the receptor(s) mediating the protective effects of adiponectin on the vasculature is not known. Here we examined the role of membrane protein T-cadherin, which localizes adiponectin to the vascular endothelium, in the revascularization response to chronic ischemia. T-cadherin-deficient mice were analyzed in a model of hind limb ischemia where blood flow is surgically disrupted in one limb and recovery is monitored over 28 days by laser Doppler perfusion imaging. In this model, T-cadherin-deficient mice phenocopy adiponectin-deficient mice such that both strains display an impaired blood flow recovery compared with wild-type controls. Delivery of exogenous adiponectin rescued the impaired revascularization phenotype in adiponectin-deficient mice but not in T-cadherin-deficient mice. In cultured endothelial cells, T-cadherin deficiency by siRNA knockdown prevented the ability of adiponectin to promote cellular migration and proliferation. These data highlight a previously unrecognized role for T-cadherin in limb revascularization and show that it is essential for mediating the vascular actions of adiponectin.


Asunto(s)
Adiponectina/metabolismo , Cadherinas/metabolismo , Endotelio Vascular/metabolismo , Neovascularización Fisiológica/fisiología , Adiponectina/genética , Animales , Cadherinas/genética , Técnicas de Silenciamiento del Gen , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Isquemia/metabolismo , Ratones , Ratones Noqueados
8.
J Immunol ; 188(2): 854-63, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22156343

RESUMEN

Adiponectin (APN) is an adipose tissue-derived factor with anti-inflammatory and vascular protective properties whose levels paradoxically decrease with increasing body fat. In this study, APN's role in the early development of ALI to LPS was investigated. Intratracheal LPS elicited an exaggerated systemic inflammatory response in APN-deficient (APN(-/-)) mice compared with wild-type (wt) littermates. Increased lung injury and inflammation were observed in APN(-/-) mice as early as 4 h after delivery of LPS. Targeted gene expression profiling performed on immune and endothelial cells isolated from lung digests 4 h after LPS administration showed increased proinflammatory gene expression (e.g., IL-6) only in endothelial cells of APN(-/-) mice when compared with wt mice. Direct effects on lung endothelium were demonstrated by APN's ability to inhibit LPS-induced IL-6 production in primary human endothelial cells in culture. Furthermore, T-cadherin-deficient mice that have significantly reduced lung airspace APN but high serum APN levels had pulmonary inflammatory responses after intratracheal LPS that were similar to those of wt mice. These findings indicate the importance of serum APN in modulating LPS-induced ALI and suggest that conditions leading to hypoadiponectinemia (e.g., obesity) predispose to development of ALI through exaggerated inflammatory response in pulmonary vascular endothelium.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/prevención & control , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Tolerancia Inmunológica , Lipopolisacáridos/toxicidad , Lesión Pulmonar Aguda/metabolismo , Adiponectina/sangre , Adiponectina/deficiencia , Adiponectina/fisiología , Animales , Cadherinas/deficiencia , Cadherinas/genética , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Humanos , Tolerancia Inmunológica/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Intubación Intratraqueal , Lipopolisacáridos/antagonistas & inhibidores , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología
9.
Angiogenesis ; 16(2): 309-27, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23124902

RESUMEN

Sprouting of angiogenic perivascular cells is thought to be highly dependent upon autocrine and paracrine growth factor stimulation. Accordingly, we report that corneal angiogenesis induced by ectopic FGF implantation is strongly impaired in NG2/CSPG4 proteoglycan (PG) null mice known to harbour a putative deficit in pericyte proliferation/mobilization. Conversely, no significant differences were seen between wild type and knockout corneas when VEGF was used as an angiocrine factor. Perturbed responsiveness of NG2-deficient pericytes to paracrine and autocrine stimulation by several FGFs could be confirmed in cells isolated from NG2 null mice, while proliferation induced by other growth factors was equivalent in wild type and knockout cells. Identical results were obtained after siRNA-mediated knock-down of NG2 in human smooth muscle-like cell lines, as also demonstrated by the decreased levels of FGF receptor phosphorylation detected in these NG2 deprived cells. Binding assays with recombinant proteins and molecular interactions examined on live cells asserted that FGF-2 bound to NG2 in a glycosaminoglycan-independent, core protein-mediated manner and that the PG was alone capable of retaining FGF-2 on the cell membrane for subsequent receptor presentation. The use of dominant-negative mutant cells, engineered by combined transduction of NG2 deletion constructs and siRNA knock-down of the endogenous PG, allowed us to establish that the FGF co-receptor activity of NG2 is entirely mediated by its extracellular portion. In fact, forced overexpression of the NG2 ectodomain in human smooth muscle-like cells increased their FGF-2-induced mitosis and compensated for low levels of FGF receptor surface expression, in a manner equivalent to that produced by overexpression of the full-length NG2. Upon FGF binding, the cytoplasmic domain of NG2 is phosphorylated, but there is no evidence that this event elicits signal transductions that could bypass the FGFR-mediated ones. Pull-down experiments, protein-protein binding assays and flow cytometry FRET coherently revealed an elective ligand-independent association of NG2 with FGFR1 and FGFR3. The NG2 cooperation with these receptors was also corroborated functionally by the outcome of FGF-2 treatments of cells engineered to express diverse NG2/FGFR combinations. Comprehensively, the findings suggest that perivascular NG2 may serve as a dual modulator of the availability/accessibility of FGF at the cell membrane, as well as the resulting FGFR transducing activity.


Asunto(s)
Antígenos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Mitógenos/metabolismo , Pericitos/metabolismo , Proteoglicanos/metabolismo , Animales , Córnea/irrigación sanguínea , Transferencia Resonante de Energía de Fluorescencia , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
10.
Proc Natl Acad Sci U S A ; 107(5): 2283-8, 2010 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-20133873

RESUMEN

Voltage-gated Na(+) channel (VGSC) beta1 subunits regulate cell-cell adhesion and channel activity in vitro. We previously showed that beta1 promotes neurite outgrowth in cerebellar granule neurons (CGNs) via homophilic cell adhesion, fyn kinase, and contactin. Here we demonstrate that beta1-mediated neurite outgrowth requires Na(+) current (I(Na)) mediated by Na(v)1.6. In addition, beta1 is required for high-frequency action potential firing. Transient I(Na) is unchanged in Scn1b (beta1) null CGNs; however, the resurgent I(Na), thought to underlie high-frequency firing in Na(v)1.6-expressing cerebellar neurons, is reduced. The proportion of axon initial segments (AIS) expressing Na(v)1.6 is reduced in Scn1b null cerebellar neurons. In place of Na(v)1.6 at the AIS, we observed an increase in Na(v)1.1, whereas Na(v)1.2 was unchanged. This indicates that beta1 is required for normal localization of Na(v)1.6 at the AIS during the postnatal developmental switch to Na(v)1.6-mediated high-frequency firing. In agreement with this, beta1 is normally expressed with alpha subunits at the AIS of P14 CGNs. We propose reciprocity of function between beta1 and Na(v)1.6 such that beta1-mediated neurite outgrowth requires Na(v)1.6-mediated I(Na), and Na(v)1.6 localization and consequent high-frequency firing require beta1. We conclude that VGSC subunits function in macromolecular signaling complexes regulating both neuronal excitability and migration during cerebellar development.


Asunto(s)
Cerebelo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Sodio/metabolismo , Potenciales de Acción/fisiología , Animales , Axones/metabolismo , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Neurológicos , Complejos Multiproteicos , Canal de Sodio Activado por Voltaje NAV1.6 , Proteínas del Tejido Nervioso/química , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Transducción de Señal , Canales de Sodio/química , Canales de Sodio/deficiencia , Canales de Sodio/genética , Tetrodotoxina/toxicidad , Subunidad beta-1 de Canal de Sodio Activado por Voltaje
11.
J Neurosci ; 31(8): 2769-80, 2011 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-21414899

RESUMEN

Inhibitory interneurons play a critical role in coordinating the activity of neural circuits. To explore the mechanisms that direct the organization of inhibitory circuits, we analyzed the involvement of tropomyosin-related kinase B (TrkB) in the assembly and maintenance of GABAergic inhibitory synapses between Golgi and granule cells in the mouse cerebellar cortex. We show that TrkB acts directly within each cell-type to regulate synaptic differentiation. TrkB is required not only for assembly, but also maintenance of these synapses and acts, primarily, by regulating the localization of synaptic constituents. Postsynaptically, TrkB controls the localization of a scaffolding protein, gephyrin, but acts at a step subsequent to the localization of a cell adhesion molecule, Neuroligin-2. Importantly, TrkB is required for the localization of an Ig superfamily cell adhesion molecule, Contactin-1, in Golgi and granule cells and the absence of Contactin-1 also results in deficits in inhibitory synaptic development. Thus, our findings demonstrate that TrkB controls the assembly and maintenance of GABAergic synapses and suggest that TrkB functions, in part, through promoting synaptic adhesion.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebelosa/enzimología , Corteza Cerebelosa/crecimiento & desarrollo , Receptor trkB/fisiología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Diferenciación Celular/genética , Interneuronas/citología , Interneuronas/enzimología , Ratones , Ratones Noqueados , Ratones Transgénicos , Sinapsis/enzimología , Sinapsis/genética , Transmisión Sináptica/genética , Tropomiosina/fisiología
12.
Front Cell Neurosci ; 16: 908401, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36072569

RESUMEN

The destruction of the myelin sheath that encircles axons leads to impairments of nerve conduction and neuronal dysfunctions. A major demyelinating disorder is multiple sclerosis (MS), a progressively disabling disease in which immune cells attack the myelin. To date, there are no therapies to target selectively myelin lesions, repair the myelin or stop MS progression. Small peptides recognizing epitopes selectively exposed at sites of injury show promise for targeting therapeutics in various pathologies. Here we show the selective homing of the four amino acid peptide, cysteine-alanine-lysine glutamine (CAQK), to sites of demyelinating injuries in three different mouse models. Homing was assessed by administering fluorescein amine (FAM)-labeled peptides into the bloodstream of mice and analyzing sites of demyelination in comparison with healthy brain or spinal cord tissue. FAM-CAQK selectively targeted demyelinating areas in all three models and was absent from healthy tissue. At lesion sites, the peptide was primarily associated with the fibrous extracellular matrix (ECM) deposited in interstitial spaces proximal to reactive astrocytes. Association of FAM-CAQK was detected with tenascin-C although tenascin depositions made up only a minor portion of the examined lesion sites. In mice on a 6-week cuprizone diet, FAM-CAQK peptide crossed the nearly intact blood-brain barrier and homed to demyelinating fiber tracts. These results demonstrate the selective targeting of CAQK to demyelinating injuries under multiple conditions and confirm the previously reported association with the ECM. This work sets the stage for further developing CAQK peptide targeting for diagnostic and therapeutic applications aimed at localized myelin repair.

13.
Sci Rep ; 12(1): 4159, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35264685

RESUMEN

Adiponectin (APN), a protein abundantly secreted from adipocytes, has been reported to possess beneficial effects on cardiovascular diseases in association with its accumulation on target organs and cells by binding to T-cadherin. However, little is known about the role of APN in the development of diabetic microvascular complications, such as diabetic retinopathy (DR). Here we investigated the impact of APN on the progression of early retinal vascular damage using a streptozotocin (STZ)-induced diabetic mouse model. Our immunofluorescence results clearly showed T-cadherin-dependent localization of APN in the vascular endothelium of retinal arterioles, which was progressively decreased during the course of diabetes. Such reduction of retinal APN accompanied the early features of DR, represented by increased vascular permeability, and was prevented by glucose-lowering therapy with dapagliflozin, a selective sodium-glucose co-transporter 2 inhibitor. In addition, APN deficiency resulted in severe vascular permeability under relatively short-term hyperglycemia, together with a significant increase in vascular cellular adhesion molecule-1 (VCAM-1) and a reduction in claudin-5 in the retinal endothelium. The present study demonstrated a possible protective role of APN against the development of DR.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Adiponectina/metabolismo , Animales , Diabetes Mellitus/metabolismo , Retinopatía Diabética/metabolismo , Retinopatía Diabética/prevención & control , Endotelio Vascular/metabolismo , Glucosa/metabolismo , Ratones , Retina/metabolismo
14.
Am J Hum Genet ; 83(6): 714-24, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19026398

RESUMEN

We have previously reported a group of patients with congenital onset weakness associated with a deficiency of members of the syntrophin-alpha-dystrobrevin subcomplex and have demonstrated that loss of syntrophin and dystrobrevin from the sarcolemma of skeletal muscle can also be associated with denervation. Here, we have further studied four individuals from a consanguineous Egyptian family with a lethal congenital myopathy inherited in an autosomal-recessive fashion and characterized by a secondary loss of beta2-syntrophin and alpha-dystrobrevin from the muscle sarcolemma, central nervous system involvement, and fetal akinesia. We performed homozygosity mapping and candidate gene analysis and identified a mutation that segregates with disease within CNTN1, the gene encoding for the neural immunoglobulin family adhesion molecule, contactin-1. Contactin-1 transcripts were markedly decreased on gene-expression arrays of muscle from affected family members compared to controls. We demonstrate that contactin-1 is expressed at the neuromuscular junction (NMJ) in mice and man in addition to the previously documented expression in the central and peripheral nervous system. In patients with secondary dystroglycanopathies, we show that contactin-1 is abnormally localized to the sarcolemma instead of exclusively at the NMJ. The cntn1 null mouse presents with ataxia, progressive muscle weakness, and postnatal lethality, similar to the affected members in this family. We propose that loss of contactin-1 from the NMJ impairs communication or adhesion between nerve and muscle resulting in the severe myopathic phenotype. This disorder is part of the continuum in the clinical spectrum of congenital myopathies and congenital myasthenic syndromes.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Músculo Esquelético/patología , Mutación , Síndromes Miasténicos Congénitos/genética , Unión Neuromuscular/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Estudios de Casos y Controles , Rotura Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 12 , Estudios de Cohortes , Consanguinidad , Secuencia Conservada , Contactina 1 , Contactinas , Análisis Mutacional de ADN , Proteínas Asociadas a la Distrofina/genética , Proteínas Asociadas a la Distrofina/metabolismo , Femenino , Ligamiento Genético , Marcadores Genéticos , Haplotipos , Homocigoto , Humanos , Inmunohistoquímica , Lactante , Masculino , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Síndromes Miasténicos Congénitos/metabolismo , Unión Neuromuscular/metabolismo , Linaje , Sarcolema/metabolismo , Sarcómeros/patología , Sarcómeros/ultraestructura
15.
J Neurosci ; 29(20): 6677-90, 2009 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-19458237

RESUMEN

Although carbohydrates have been implicated in cell interactions in the nervous system, the molecular bases of their functions have remained largely obscure. Here, we show that promotion or inhibition of neurite outgrowth of cerebellar or dorsal root ganglion neurons, respectively, induced by the mucin-type adhesion molecule CD24 depends on alpha2,3-linked sialic acid and Lewis(x) present on glia-specific CD24 glycoforms. Alpha2,3-sialyl residues of CD24 bind to a structural motif in the first fibronectin type III domain of the adhesion molecule L1. Following the observation that the adhesion molecules TAG-1 and Contactin show sequence homologies with fucose-specific lectins, we obtained evidence that TAG-1 and Contactin mediate Lewis(x)-dependent CD24-induced effects on neurite outgrowth. Thus, L1, TAG-1, and Contactin function as lectin-like neuronal receptors. Their cis interactions with neighboring adhesion molecules, e.g., Caspr1 and Caspr2, and with their triggered signal transduction pathways elicit cell type-specific promotion or inhibition of neurite outgrowth induced by glial CD24 in a glycan-dependent trans interaction.


Asunto(s)
Antígeno CD24/metabolismo , Moléculas de Adhesión Celular Neuronal/fisiología , Antígeno Lewis X/farmacología , Neuritas/efectos de los fármacos , Neuronas/citología , Ácidos Siálicos/farmacología , Animales , Animales Recién Nacidos , Sitios de Unión/efectos de los fármacos , Antígeno CD24/genética , Moléculas de Adhesión Celular Neuronal/deficiencia , Células Cultivadas , Cerebelo/citología , Contactina 2 , Contactinas , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Ganglios Espinales/citología , Glicosilación/efectos de los fármacos , Inmunoprecipitación/métodos , Complejo de Antígeno L1 de Leucocito/metabolismo , Locomoción/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuritas/fisiología , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/fisiología , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Recuperación de la Función/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Transfección/métodos
16.
Am J Respir Cell Mol Biol ; 43(4): 487-97, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19915153

RESUMEN

Obese mice have increased responses to acute ozone (O(3)) exposure. T-cadherin is a binding protein for the high-molecular weight isoforms of adiponectin, an anti-inflammatory hormone that declines in obesity. The objective of the present study was to determine whether adiponectin affects pulmonary responses to O(3), and whether these effects are mediated through T-cadherin. We performed bronchoalveolar lavage (BAL) and measured pulmonary responsiveness to methacholine after acute air or O(3) exposure (2 ppm for 3 h) in adiponectin-deficient (Adipo(-/-)) or T-cadherin-deficient (T-Cad(-/-)) mice. O(3) increased pulmonary responses to methacholine and increased BAL neutrophils and protein to a greater extent in wild-type than in Adipo(-/-) mice, whereas T-cadherin deficiency had no effect. O(3)-induced increases in BAL IL-6 and keratinocyte-derived chemokine (KC), which contribute to O(3)-induced pulmonary neutrophilia, were also greater in wild-type than in Adipo(-/-) mice. In contrast, responses to O(3) were not altered by transgenic overexpression of adiponectin. To determine which adiponectin isoforms are present in the lung, Western blotting was performed. The hexameric isoform of adiponectin dominated in serum, whereas BAL was dominated by the high-molecular weight isoform of adiponectin. Interestingly, serum adiponectin was greater in T-Cad(-/-) versus wild-type mice, whereas BAL adiponectin was lower in T-Cad(-/-) versus wild-type mice, suggesting that T-cadherin may be important for transit of high-molecular weight adiponectin from the blood to the lung. Our results indicate that adiponectin deficiency inhibits pulmonary inflammation induced by acute O(3) exposure, and that T-cadherin does not mediate the effects of adiponectin responsible for these events.


Asunto(s)
Adiponectina/deficiencia , Adiponectina/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ozono/administración & dosificación , Ozono/farmacología , Adiponectina/sangre , Animales , Peso Corporal/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/citología , Cadherinas/deficiencia , Cadherinas/metabolismo , Recuento de Células , Citocinas/metabolismo , Exposición por Inhalación , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/metabolismo
17.
Mol Cell Biol ; 27(19): 6659-68, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17682066

RESUMEN

Polysialic acid, which is synthesized by two polysialyltransferases, ST8SiaII and ST8SiaIV, plays an essential role in brain development by modifying the neural cell adhesion molecule (NCAM). It is currently unclear how polysialic acid functions in different processes of neural development. Here we generated mice doubly mutant in both ST8SiaII and ST8SiaIV to determine the effects of loss of polysialic acid on brain development. In contrast to NCAM-deficient, ST8SiaII-deficient, or ST8SiaIV-deficient single mutant mice, ST8SiaII and ST8SiaIV double mutants displayed severe defects in anatomical organization of the forebrain associated with apoptotic cell death. Loss of polysialic acid affected both tangential and radial migration of neural precursors during cortical development, resulting in aberrant positioning of neuronal and glial cells. Glial cell differentiation was aberrantly increased in vivo and in vitro in the absence of polysialic acid. Consistent with these findings, polysialic acid-deficient mice exhibited increased expression of the glial cell marker glial fibrillary acidic protein and a decrease in expression of Pax6, a transcription factor regulating neural cell migration. These results indicate that polysialic acid regulates cell migration and differentiation of neural precursors crucial for brain development.


Asunto(s)
Diferenciación Celular , Movimiento Celular/fisiología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Neuronas/fisiología , Ácidos Siálicos/metabolismo , Sialiltransferasas/genética , Células Madre/fisiología , Animales , Apoptosis/fisiología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Noqueados , Neuroglía/citología , Neuroglía/fisiología , Neuronas/citología , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Sialiltransferasas/metabolismo , Células Madre/citología
18.
Mol Cell Biol ; 27(19): 6706-17, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17646384

RESUMEN

The intracellular signaling controlling neural stem/progenitor cell (NSC) self-renewal and neuronal/glial differentiation is not fully understood. We show here that Shp2, an introcellular tyrosine phosphatase with two SH2 domains, plays a critical role in NSC activities. Conditional deletion of Shp2 in neural progenitor cells mediated by Nestin-Cre resulted in early postnatal lethality, impaired corticogenesis, and reduced proliferation of progenitor cells in the ventricular zone. In vitro analyses suggest that Shp2 mediates basic fibroblast growth factor signals in stimulating self-renewing proliferation of NSCs, partly through control of Bmi-1 expression. Furthermore, Shp2 regulates cell fate decisions, by promoting neurogenesis while suppressing astrogliogenesis, through reciprocal regulation of the Erk and Stat3 signaling pathways. Together, these results identify Shp2 as a critical signaling molecule in coordinated regulation of progenitor cell proliferation and neuronal/astroglial cell differentiation.


Asunto(s)
Encéfalo , Diferenciación Celular , Proliferación Celular , Neuronas/fisiología , Proteínas Nucleares/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Astrocitos/citología , Astrocitos/fisiología , Encéfalo/citología , Encéfalo/embriología , Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Femenino , Hibridación in Situ , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Proteínas Nucleares/genética , Complejo Represivo Polycomb 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Células Madre/citología
19.
Clin Cancer Res ; 15(10): 3256-64, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19447866

RESUMEN

PURPOSE: High levels of the fat-secreted cytokine adiponectin (APN) are present in the circulation of healthy people, whereas low levels correlate with an increased incidence of breast cancer in women. The current study experimentally probes the physiologic functions of APN in mammary cancer in a newly generated genetic mouse model. EXPERIMENTAL DESIGN: We established an APN null mouse model of mammary cancer by introducing the polyoma virus middle T (PyV-mT) oncogene expressed from mouse mammary tumor virus (MMTV) regulatory elements into APN null mice. MMTV-PyV-mT-induced tumors resemble ErbB2-amplified human breast cancers. We monitored tumor onset, kinetics, and animal survival, and analyzed vascular coverage, apoptosis, and hypoxia in sections from the primary tumors. Metastatic spreading was evaluated by analyses of the lungs. RESULTS: APN prominently localized to the vasculature in human and mouse mammary tumors. In APN null mice, MMTV-PyV-mT-induced tumors appeared with delayed onset and exhibited reduced growth rates. Affected animals survived control tumor-bearing mice by an average of 21 days. Pathologic analyses revealed reduced vascularization of APN null tumors along with increased hypoxia and apoptosis. At the experimental end point, APN null transgenic mice showed increased frequency of pulmonary metastases. CONCLUSION: The current work identifies a proangiogenic contribution of APN in mammary cancer that, in turn, affects tumor progression. APN interactions with vascular receptors may be useful targets for developing therapies aimed at controlling tumor vascularization in cancer patients.


Asunto(s)
Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/genética , Neovascularización Patológica/genética , Adiponectina/genética , Adiponectina/metabolismo , Adiponectina/farmacología , Animales , Antígenos Virales de Tumores/genética , Apoptosis , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Neovascularización de la Córnea , Femenino , Immunoblotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Poliomavirus/genética , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/farmacología
20.
Front Immunol ; 11: 1268, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32676079

RESUMEN

Neuronal regulation of diverse physiological functions requires complex molecular interactions in innervated tissues to maintain proper organ function. Here we show that loss of the neuronal cell surface adhesion/recognition molecule Contactin-1 (Cntn1) directly impairs intestinal function causing wasting that subsequently results in global immune defects. Loss of Cntn1 results in hematologic alterations and changes in blood metabolites associated with malnourishment. We found thymus and spleen of Cntn1-deficient animals atrophied with severe reductions in lymphocyte populations. Elevated thymic Gilz expression indicated ongoing glucocorticoid signaling in Cntn1-deficient animals, consistent with the malnourishment phenotype. Intestinal Contactin-1 was localized to neurons in the villi and the submucosal/myenteric plexus that innervates smooth muscle. Loss of Cntn1 was associated with reduced intestinal Bdnf and Adrb2, indicating reduced neuromuscular crosstalk. Additionally, loss of Cntn1 resulted in reduced recruitment of CD3+ T cells to villi within the small intestine. Together, these data illustrate the critical role of Contactin-1 function within the gut, and how this is required for normal systemic immune functions.


Asunto(s)
Contactina 1/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/inervación , Animales , Biomarcadores , Recuento de Células Sanguíneas , Análisis Químico de la Sangre , Citometría de Flujo , Perfilación de la Expresión Génica , Glucocorticoides/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Fenotipo , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Timo/inmunología , Timo/metabolismo , Timo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA