Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Front Mol Neurosci ; 10: 116, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28515678

RESUMEN

Synaptic cell adhesion molecules represent important targets for neuronal activity-dependent proteolysis. Postsynaptic neuroligins (NLs) form trans-synaptic complexes with presynaptic neurexins (NXs). Both NXs and NLs are cleaved from the cell surface by metalloproteases in an activity-dependent manner, releasing a soluble extracellular fragment and membrane-tethered C-terminal fragment. The cleavage of NL1 depresses synaptic transmission, but the mechanism by which this occurs is unknown. Metabotropic glutamate receptor 2 (mGluR2) are located primarily at the periphery of presynaptic terminals, where they inhibit the formation of cyclic adenosine monophosphate (cAMP) and consequently suppress the release of glutamate and decrease synaptic transmission. In the present study, we found that the soluble ectodomain of NL1 binds to and activates mGluR2 in both neurons and heterologous cells, resulting in a decrease in cAMP formation. In a slice preparation from the hippocampus of mice, NL1 inhibited the release of glutamate from mossy fibers that project to CA3 pyramidal neurons. The presynaptic effect of NL1 was abolished in the presence of a selective antagonist for mGluR2. Thus, our data suggest that the soluble extracellular domain of NL1 functionally interacts with mGluR2 and thereby decreases synaptic strength.

2.
J Obstet Gynaecol Res ; 42(12): 1846-1853, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27748558

RESUMEN

AIM: The aim of this study was to develop a novel optical imaging system for detecting protoporphyrin IX (PpIX) autofluorescence, to prove that PpIX autofluorescence is as useful as 5-aminolevulinic acid (5-ALA)-induced fluorescence for detecting and localizing cervical cancer, and to monitor the change in PpIX autofluorescence or induced PpIX fluorescence before, during, and after photodynamic therapy (PDT). METHODS: TC-1 cells - highly tumorigenic cells immortalized using human papillomavirus type 16 proteins E6 and E7 - were subcutaneously grafted into the thighs of nude mice. The suspected tumor tissues were visualized using autofluorescence imaging and induced fluorescence imaging under 5-ALA administration. When the 5-ALA-induced PpIX was sufficiently accumulated in tumor tissues, PDT was performed using a 635-nm laser. We observed the change in fluorescence intensity during PDT. For 3 weeks after PDT, we monitored tumor remission by using white-light imaging and fluorescence imaging. RESULTS: The transplanted cells were visualized by PpIX autofluorescence, which was induced by heme synthesis. After 5-ALA administration, PpIX could be targeted by using PDT, which decreased PpIX autofluorescence. Photobleaching is useful for monitoring PDT dosimetry and for determining the photodynamic response to therapy. CONCLUSION: PpIX autofluorescence clearly differentiated the tumor from adjacent normal tissues. The results of PpIX autofluorescence imaging and 5-ALA-induced fluorescence imaging were identical. PpIX autofluorescence imaging is a simple and cost-effective cervical cancer screening method that could be performed during or after PDT to ensure effective treatment or remission as a change in fluorescence intensity can be observed in real time without a blinding effect.


Asunto(s)
Ácido Aminolevulínico/farmacocinética , Imagen Óptica/métodos , Protoporfirinas/farmacocinética , Neoplasias del Cuello Uterino/diagnóstico por imagen , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Papillomavirus Humano 16 , Humanos , Ratones , Ratones Desnudos , Fotoquimioterapia , Neoplasias del Cuello Uterino/tratamiento farmacológico
3.
Virusdisease ; 26(3): 123-32, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26396978

RESUMEN

Respiratory viruses are a major public health problem because of their prevalence and high morbidity rate leading to considerable social and economic implications. Cranberry has therapeutic potential attributed to a comprehensive list of phytochemicals including anthocyanins, flavonols, and unique A-type proanthocyanidins. Soy flavonoids, including isoflavones, have demonstrated anti-viral effects in vitro and in vivo. Recently, it was demonstrated that edible proteins can efficiently sorb and concentrate cranberry polyphenols, including anthocyanins and proanthocyanins, providing greatly stabilized matrices suitable for food products. The combination of cranberry and soy phytoactives may be an effective dietary anti-viral resource. Anti-viral properties of both cranberry juice-enriched and cranberry pomace polyphenol-enriched soy protein isolate (CB-SPI and CBP-SPI) were tested against influenza viruses (H7N1, H5N3, H3N2), Newcastle disease virus and Sendai virus in vitro and in ovo. In our experiments, preincubation with CB-SPI or CBP-SPI resulted in inhibition of virus adsorption to chicken red blood cells and reduction in virus nucleic acid content up to 16-fold, however, CB-SPI and CBP-SPI did not affect hemagglutination. Additionally, CB-SPI and CBP-SPI inhibited viral replication and infectivity more effectively than the commercially available anti-viral drug Amizon. Results suggest CB-SPI and CBP-SPI may have preventative and therapeutic potential against viral infections that cause diseases of the respiratory and gastro-intestinal tract.

4.
Prog Brain Res ; 214: 353-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25410365

RESUMEN

Extracellular matrix (ECM) molecules, their receptors at the cell surface, and cell adhesion molecules (CAMs) involved in cell-cell or cell-ECM interactions are implicated in processes related to major diseases of the central nervous system including Alzheimer's disease (AD), epilepsy, schizophrenia, addiction, multiple sclerosis, Parkinson's disease, and cancer. There are multiple strategies for targeting the ECM molecules and their metabolizing enzymes and receptors with antibodies, peptides, glycosaminoglycans, and other natural and synthetic compounds. ECM-targeting treatments include chondroitinase ABC, heparin/heparan sulfate-mimicking oligosaccharides, ECM cross-linking antibodies, and drugs stimulating expression of ECM molecules. The amount or activity of ECM-degrading enzymes like matrix metalloproteinases can be modulated indirectly via the regulation of endogenous inhibitors like TIMPs and RECK or at the transcriptional and translational levels using, e.g., histone deacetylase inhibitors, synthetic inhibitors like Periostat, microRNA-interfering drugs like AC1MMYR2, and natural compounds like flavonoids, epigallocatechin-3-gallate, anacardic acid, and erythropoietin. Among drugs targeting the major ECM receptors, integrins, are the anticancer peptide cilengitide and anti-integrin antibodies, which have a potential for treatment of stroke, multiple sclerosis, and AD. The latter can be also potentially treated with modulators of CAMs, such as peptide mimetics derived from L1-CAM and NCAM1.


Asunto(s)
Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/terapia , Matriz Extracelular/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Humanos
5.
J Neuroinflammation ; 11: 27, 2014 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-24490798

RESUMEN

BACKGROUND: Interleukin 1 (IL-1) is implicated in neuroinflammation, an essential component of neurodegeneration. We evaluated the potential anti-inflammatory effect of a novel peptide antagonist of IL-1 signaling, Ilantide. METHODS: We investigated the binding of Ilantide to IL-1 receptor type I (IL-1RI) using surface plasmon resonance, the inhibition of Il-1ß-induced activation of nuclear factor κB (NF-κB) in HEK-Blue cells that contained an IL-1ß-sensitive reporter, the secretion of TNF-α in macrophages, protection against IL-1-induced apoptosis in neonatal pancreatic islets, and the penetration of Ilantide through the blood-brain barrier using competitive enzyme-linked immunosorbent assay (ELISA). We studied the effects of the peptide on social behavior and memory in rat models of lipopolysaccharide (LPS)- and amyloid-induced neuroinflammation, respectively, and its effect in a rat model of experimental autoimmune enchephalomyelitis. RESULTS: Ilantide bound IL-1RI, inhibited the IL-1ß-induced activation of NF-κB, and inhibited the secretion of TNF-α in vitro. Ilantide protected pancreatic islets from apoptosis in vitro and reduced inflammation in an animal model of arthritis. The peptide penetrated the blood-brain barrier. It reduced the deficits in social activity and memory in LPS- and amyloid-treated animals and delayed the development of experimental autoimmune enchephalomyelitis. CONCLUSIONS: These findings indicate that Ilantide is a novel and potent IL-1RI antagonist that is able to reduce inflammatory damage in the central nervous system and pancreatic islets.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Animales , Animales Recién Nacidos , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Artritis/inducido químicamente , Células Cultivadas , Cerebelo/citología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Humanos , Proteína Antagonista del Receptor de Interleucina 1/química , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Lipopolisacáridos , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar , Conducta Social , Transfección , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
6.
Neuropsychopharmacology ; 39(5): 1148-58, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24213355

RESUMEN

Neuroligins (NLGNs) are cell adhesion molecules that are important for proper synaptic formation and functioning, and are critical regulators of the balance between neural excitation/inhibition (E/I). Mutations in NLGNs have been linked to psychiatric disorders in humans involving social dysfunction and are related to similar abnormalities in animal models. Chronic stress increases the likelihood for affective disorders and has been shown to induce changes in neural structure and function in different brain regions, with the hippocampus being highly vulnerable to stress. Previous studies have shown evidence of chronic stress-induced changes in the neural E/I balance in the hippocampus. Therefore, we hypothesized that chronic restraint stress would lead to reduced hippocampal NLGN-2 levels, in association with alterations in social behavior. We found that rats submitted to chronic restraint stress in adulthood display reduced sociability and increased aggression. This occurs along with a reduction of NLGN-2, but not NLGN-1 expression (as shown with western blot, immunohistochemistry, and electron microscopy analyses), throughout the hippocampus and detectable in different layers of the CA1, CA3, and DG subfields. Furthermore, using synthetic peptides that comprise sequences in either NLGN-1 (neurolide-1) or NLGN-2 (neurolide-2) involved in the interaction with their presynaptic partner neurexin (NRXN)-1, intra-hippocampal administration of neurolide-2 led also to reduced sociability and increased aggression. These results highlight hippocampal NLGN-2 as a key molecular substrate regulating social behaviors and underscore NLGNs as promising targets for the development of novel drugs for the treatment of dysfunctional social behaviors.


Asunto(s)
Agresión/fisiología , Moléculas de Adhesión Celular Neuronal/metabolismo , Hipocampo/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Conducta Social , Estrés Psicológico/fisiopatología , Animales , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Enfermedad Crónica , Corticosterona/sangre , Hipocampo/patología , Masculino , Proteínas del Tejido Nervioso/genética , Neuritas/fisiología , Neuronas/fisiología , Tamaño de los Órganos , Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Superficie Celular/metabolismo , Restricción Física , Estrés Psicológico/patología
7.
Neurochem Res ; 38(12): 2550-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24132641

RESUMEN

ErbB receptors not only function in cancer, but are also key developmental regulators in the nervous system. We previously identified an ErbB1 peptide antagonist, Inherbin3, that is capable of inhibiting tumor growth in vitro and in vivo. In this study, we found that inhibition of ErbB1 kinase activity and activation of ErbB4 by NRG-1ß induced neurite extension, suggesting that ErbB1 and ErbB4 act as negative and positive regulators, respectively, of the neuritogenic response. Inherbin3, inhibited activation not only of ErbB1 but also of ErbB4 in primary neurons, strongly induced neurite outgrowth in rat cerebellar granule neurons, indicating that this effect mainly was due to inhibition of ErbB1 activation.


Asunto(s)
Cerebelo/efectos de los fármacos , Gránulos Citoplasmáticos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Neuritas/efectos de los fármacos , Péptidos/farmacología , Animales , Secuencia de Bases , Células Cultivadas , Cerebelo/citología , Cartilla de ADN , Receptores ErbB/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Fosforilación , Reacción en Cadena de la Polimerasa , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Cytokine ; 64(1): 112-21, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23972727

RESUMEN

Interleukin-4 (IL-4) is a potent antiinflammatory cytokine. However its use in the clinic is hampered by side effects. We here describe the identification of a novel synthetic peptide, termed Ph8, derived from α-helix C of IL-4, which interacts with IL-4 receptor α (IL-4Rα). Employing various cultured genetically engineered cell lines and primary lymphocytes, surface plasmon resonance, qPCR, ELISA and immunoblotting techniques we found that Ph8 bound IL-4Rα and mimicked the anti-inflammatory effects of IL-4 by inhibiting TNF-α production by macrophages in vitro. It induced phosphorylation of STAT6 65kD but inhibited phosphorylation of STAT6 110 kD induced by IL-4 in a B-cell line that expressed the type I receptor. It also inhibited the IL-4-stimulated expression of a STAT6-inducible reporter gene in cells that expressed the type II receptor. Ph8 inhibited the proliferation of Th1/2 cells and downregulated the production of IFN-γ in stimulated Th1 cells. Moreover, Ph8 did not induce any shift in Th1/Th2 profile. This is a favorable effect and it is indicating that Ph8 could block general T cell activation and inflammatory responses without further inducing the side effects generally associated with IL-4 signaling. These data collectively show that Ph8 is only a partial agonist of IL-4 mimicking its desirable properties. In agreement, Ph8 treatment of rats with collagen-induced arthritis, a Th1- and antibody- mediated disease of joint, delayed the manifestation of chronic inflammation and reduced acute inflammation in carrageenan-induced edema. Our findings indicate that Ph8 is a promising potential drug candidate for the treatment of inflammatory diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Interleucina-4/farmacología , Fragmentos de Péptidos/farmacología , Animales , Artritis Experimental/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Edema/tratamiento farmacológico , Células HEK293 , Humanos , Interferón gamma/metabolismo , Interleucina-4/análogos & derivados , Interleucina-4/química , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Fosforilación/efectos de los fármacos , Unión Proteica , Ratas , Ratas Wistar , Factor de Transcripción STAT6/metabolismo , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Cell Oncol (Dordr) ; 36(3): 201-11, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23580313

RESUMEN

BACKGROUND: Receptors of the ErbB family are involved in the development of various cancers, and the inhibition of these receptors represents an attractive therapeutic concept. Upon ligand binding, ErbB receptors become activated as homo- or heterodimers, leading to the activation of downstream signaling cascades that result in the facilitation of cell proliferation and migration. A region of the extracellular part of the receptor, termed the 'dimerization arm', is important for the formation of receptor dimers and represents an attractive target for the design of ErbB inhibitors. METHODS: An ErbB1 targeting peptide, termed Herfin-1, was designed based on a model of the tertiary structure of the EGF-EGFR ternary complex. The binding kinetics of this peptide were determined employing surface plasmon resonance analyses. ErbB1-4 expression and phosphorylation in human glioblastoma cell lines U87 and U118 were determined by Western blotting using specific antibodies. Cell proliferation was determined by MTS staining. Cell migration was examined using a Chemotaxis Migration Kit. Neurite outgrowth from primary cerebellar granule neurons was evaluated by fluorescence microscopy and image processing. RESULTS: The present study shows that Herfin-1 functions as an ErbB1 antagonist. It binds to the extracellular domain of ErbB1 with a KD value of 361 nM. In U87 and U118 cells, both expressing high levels of ErbB1, Herfin-1 inhibits EGF-induced ErbB1 phosphorylation and cell migration. Additionally, Herfin-1 was found to increase neurite outgrowth in cerebellar granule neurons, likely through the inhibition of a sustained weak ErbB1 activation. CONCLUSIONS: Targeting the ErbB1 receptor dimerization interface is a promising strategy to inhibit receptor activation in ErbB1-expressing glioma cells.


Asunto(s)
Neoplasias Encefálicas/patología , Movimiento Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Glioblastoma/patología , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Diseño de Fármacos , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/química , Receptores ErbB/metabolismo , Glioblastoma/tratamiento farmacológico , Humanos , Datos de Secuencia Molecular , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neurogénesis/efectos de los fármacos , Péptidos/química , Péptidos/uso terapéutico , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Ratas Wistar
10.
Mol Med ; 19: 43-53, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23508572

RESUMEN

We recently found that S100A4, a member of the multifunctional S100 protein family, protects neurons in the injured brain and identified two sequence motifs in S100A4 mediating its neurotrophic effect. Synthetic peptides encompassing these motifs stimulated neuritogenesis and survival in vitro and mimicked the S100A4-induced neuroprotection in brain trauma. Here, we investigated a possible function of S100A4 and its mimetics in the pathologies of the peripheral nervous system (PNS). We found that S100A4 was expressed in the injured PNS and that its peptide mimetic (H3) affected the regeneration and survival of myelinated axons. H3 accelerated electrophysiological, behavioral and morphological recovery after sciatic nerve crush while transiently delaying regeneration after sciatic nerve transection and repair. On the basis of the finding that both S100A4 and H3 increased neurite branching in vitro, these effects were attributed to the modulatory effect of H3 on initial axonal sprouting. In contrast to the modest effect of H3 on the time course of regeneration, H3 had a long-term neuroprotective effect in the myelin protein P0 null mice, a model of dysmyelinating neuropathy (Charcot-Marie-Tooth type 1 disease), where the peptide attenuated the deterioration of nerve conduction, demyelination and axonal loss. From these results, S100A4 mimetics emerge as a possible means to enhance axonal sprouting and survival, especially in the context of demyelinating neuropathies with secondary axonal loss, such as Charcot-Marie-Tooth type 1 disease. Moreover, our data suggest that S100A4 is a neuroprotectant in PNS and that other S100 proteins, sharing high homology in the H3 motif, may have important functions in PNS pathologies.


Asunto(s)
Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Proteínas S100/farmacología , Nervio Ciático/efectos de los fármacos , Animales , Células Cultivadas , Enfermedad de Charcot-Marie-Tooth/tratamiento farmacológico , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Hipocampo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína P0 de la Mielina/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/uso terapéutico , Péptidos/uso terapéutico , Ratas , Ratas Wistar , Proteínas S100/uso terapéutico , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología , Nervio Tibial/efectos de los fármacos , Nervio Tibial/fisiopatología
11.
Nat Commun ; 3: 1197, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23149742

RESUMEN

Identification of novel pro-survival factors in the brain is paramount for developing neuroprotective therapies. The multifunctional S100 family proteins have important roles in many human diseases and are also upregulated by brain injury. However, S100 functions in the nervous system remain unclear. Here we show that the S100A4 protein, mostly studied in cancer, is overexpressed in the damaged human and rodent brain and released from stressed astrocytes. Genetic deletion of S100A4 exacerbates neuronal loss after brain trauma or excitotoxicity, increasing oxidative cell damage and downregulating the neuroprotective protein metallothionein I+II. We identify two neurotrophic motifs in S100A4 and show that these motifs are neuroprotective in animal models of brain trauma. Finally, we find that S100A4 rescues neurons via the Janus kinase/STAT pathway and, partially, the interleukin-10 receptor. Our data introduce S100A4 as a therapeutic target in neurodegeneration, and raise the entire S100 family as a potentially important factor in central nervous system injury.


Asunto(s)
Citoprotección , Metástasis de la Neoplasia/patología , Neuronas/patología , Proteínas S100/metabolismo , Secuencias de Aminoácidos , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Muerte Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Quinasas Janus/metabolismo , Ácido Kaínico , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Neurotoxinas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Péptidos/farmacología , Péptidos/uso terapéutico , Ratas , Receptores de Interleucina-10/metabolismo , Proteína de Unión al Calcio S100A4 , Proteínas S100/química , Factores de Transcripción STAT/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/patología , Regulación hacia Arriba/efectos de los fármacos
12.
J Neurochem ; 121(6): 915-23, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22469063

RESUMEN

Apart from its hematopoietic activity, erythropoietin (EPO) is also known as a tissue-protective cytokine. In the brain, EPO and its receptor are up-regulated in response to insult and exert pro-survival effects. EPO binds to its receptor (EPOR) via high- and low-affinity binding sites (Sites 1 and 2, respectively), inducing conformational changes in the receptor, followed by the activation of downstream signaling cascades. Based on the crystal structure of the EPO:EPOR(2) complex, we designed a peptide, termed Epobis, whose sequence encompassed amino acids from binding Site 1. The present study shows that the Epobis peptide specifically binds to EPOR and induces neurite outgrowth from primary neurons in an EPOR-expression dependent manner. Furthermore, Epobis promoted the survival of hippocampal and cerebellar neuronal cultures after kainate treatment and KCl deprivation, respectively. Thus, we identified a new functional agonist of EPOR with the potential to promote neuroregeneration and neuroprotection.


Asunto(s)
Neuritas/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , Eritropoyetina/química , Eritropoyetina/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Modelos Moleculares , Fármacos Neuroprotectores/metabolismo , Péptidos/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Resonancia por Plasmón de Superficie , Transfección
13.
Int J Biochem Cell Biol ; 44(3): 441-6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155300

RESUMEN

Neural cell adhesion molecules 2 (NCAM2/OCAM/RNCAM), is a paralog of NCAM1. The protein exists in a transmembrane and a lipid-anchored isoform, and has an ectodomain consisting of five immunoglobulin modules and two fibronectin type 3 homology modules. Structural models of the NCAM2 ectodomain reveal that it facilitates cell adhesion through reciprocal interactions between the membrane-distal immunoglobulin modules. There are no known heterophilic NCAM2 binding partners, and NCAM2 is not glycosylated with polysialic acid, a posttranslational modification known to be a major modulator of NCAM1-mediated processes. This suggests that NCAM2 has a function or mode of action distinctly different from that of NCAM1. NCAM2 is primarily expressed in the brain, where it is believed to stimulate neurite outgrowth and to facilitate dendritic and axonal compartmentalization.


Asunto(s)
Enfermedad de Alzheimer/genética , Trastorno Autístico/genética , Síndrome de Down/genética , Neoplasias/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/metabolismo , Vías Olfatorias/patología , Animales , Axones/fisiología , Adhesión Celular , Terapia Genética , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Molécula L1 de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa , Vías Olfatorias/embriología , Vías Olfatorias/crecimiento & desarrollo , Organogénesis
14.
Eur J Cell Biol ; 90(12): 990-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22000729

RESUMEN

Ciliary neurotrophic factor (CNTF) induces neuronal differentiation and promotes the survival of various neuronal cell types by binding to a receptor complex formed by CNTF receptor α (CNTFRα), gp130, and the leukemia inhibitory factor (LIF) receptor (LIFR). The CD loop-D helix region of CNTF has been suggested to be important for the cytokine interaction with LIFR. We designed a peptide, termed cintrofin, that encompasses this region. Surface plasmon resonance analysis demonstrated that cintrofin bound to LIFR and gp130, but not to CNTFRα, with apparent KD values of 35 nM and 1.1 nM, respectively. Cintrofin promoted the survival of cerebellar granule neurons (CGNs), in which cell death was induced either by potassium withdrawal or H2O2 treatment. Cintrofin induced neurite outgrowth from CGNs, and this effect was inhibited by specific antibodies against both gp130 and LIFR, indicating that these receptors are involved in the effects of cintrofin. The C-terminal part of the peptide, corresponding to the D helix region of CNTF, was shown to be essential for the neuritogenic action of the peptide. CNTF and LIF induced neurite outgrowth in CGNs plated on laminin-coated slides. On uncoated slides, CNTF and LIF had no neuritogenic effect but were able to inhibit cintrofin-induced neuronal differentiation, indicating that cintrofin and cytokines compete for the same receptors. In addition, cintrofin induced the phosphorylation of STAT3, Akt, and ERK, indicating that it exerts cell signaling properties similar to those induced by CNTF and may be a valuable survival agent with possible therapeutic potential.


Asunto(s)
Factor Neurotrófico Ciliar/farmacología , Factor Inhibidor de Leucemia/metabolismo , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Citocinas/metabolismo , Secuencia de Aminoácidos , Antígenos CD/metabolismo , Diferenciación Celular/efectos de los fármacos , Factor Neurotrófico Ciliar/metabolismo , Humanos , Interleucina-6/metabolismo , Microscopía Confocal , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Unión Proteica , Transducción de Señal
15.
EMBO Mol Med ; 3(8): 480-94, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21739604

RESUMEN

Epithelial ovarian carcinoma (EOC) is an aggressive neoplasm, which mainly disseminates to organs of the peritoneal cavity, an event mediated by molecular mechanisms that remain elusive. Here, we investigated the expression and functional role of neural cell adhesion molecule (NCAM), a cell surface glycoprotein involved in brain development and plasticity, in EOC. NCAM is absent from normal ovarian epithelium but becomes highly expressed in a subset of human EOC, in which NCAM expression is associated with high tumour grade, suggesting a causal role in cancer aggressiveness. We demonstrate that NCAM stimulates EOC cell migration and invasion in vitro and promotes metastatic dissemination in mice. This pro-malignant function of NCAM is mediated by its interaction with fibroblast growth factor receptor (FGFR). Indeed, not only FGFR signalling is required for NCAM-induced EOC cell motility, but targeting the NCAM/FGFR interplay with a monoclonal antibody abolishes the metastatic dissemination of EOC in mice. Our results point to NCAM-mediated stimulation of FGFR as a novel mechanism underlying EOC malignancy and indicate that this interplay may represent a valuable therapeutic target.


Asunto(s)
Carcinoma/patología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neoplasias Ováricas/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Carcinoma/secundario , Movimiento Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunohistoquímica , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia/patología , Neoplasias Ováricas/secundario
16.
J Neurochem ; 117(6): 984-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21480899

RESUMEN

Neuroplastin-65 (Np65) is a brain-specific cell adhesion molecule belonging to the immunoglobulin superfamily. Homophilic trans-interaction of Np65 mediates adhesion between cells and modulates synaptic plasticity. This interaction solely occurs through the first immunoglobulin (Ig) module of Np65, but the exact binding mechanism has not yet been elucidated. In this study, we identify the homophilic binding motif of Np65 and show that a synthetic peptide modeled after this motif, termed enplastin, binds to Np65. We demonstrate that both Np65- and enplastin-induced intracellular signaling depends on fibroblast growth factor receptor, p38 mitogen-activated protein kinase, Ca(2+) /calmodulin-dependent protein kinase, and cytoplasmic Ca(2+) concentration. In addition, we show that interference with Np65 homophilic binding by enplastin has an inhibitory effect on Np65-mediated neurite outgrowth in vitro and on the initial phase of spatial learning in rats.


Asunto(s)
Glicoproteínas de Membrana/fisiología , Plasticidad Neuronal , Neuronas/fisiología , Péptidos/farmacología , Animales , Sitios de Unión , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Cerebelo/citología , Cerebelo/embriología , Cerebelo/metabolismo , Activación Enzimática , Hipocampo/citología , Hipocampo/embriología , Hipocampo/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Glicoproteínas de Membrana/genética , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Imitación Molecular , Neuritas/efectos de los fármacos , Neuritas/fisiología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Péptidos/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Factores de Crecimiento de Fibroblastos/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
17.
BMC Cancer ; 10: 383, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20663132

RESUMEN

BACKGROUND: The anti-epileptic drug valproic acid (VPA) has attracted attention as an anti-cancer agent. METHODS: The present study investigated effects of VPA exposure on histone deacetylase (HDAC) inhibition, cell growth, cell speed, and the degree of Erk1/2 phosphorylation in 10 cell lines (BT4C, BT4Cn, U87MG, N2a, PC12-E2, CSML0, CSML100, HeLa, L929, Swiss 3T3). RESULTS: VPA induced significant histone deacetylase (HDAC) inhibition in most of the cell lines, but the degree of inhibition was highly cell type-specific. Moreover, cell growth, motility and the degree of Erk1/2 phosphorylation were inhibited, activated, or unaffected by VPA in a cell type-specific manner. Importantly, no relationship was found between the effects of VPA on HDAC inhibition and changes in the degree of Erk1/2 phosphorylation, cell growth, or motility. In contrast, VPA-induced modulation of the MAPK pathway downstream of Ras but upstream of MEK (i.e., at the level of Raf) was important for changes in cell speed. CONCLUSIONS: These results suggest that VPA can modulate the degree of Erk1/2 phosphorylation in a manner unrelated to HDAC inhibition and emphasize that changes in the degree of Erk1/2 phosphorylation are also important for the anti-cancer properties of VPA.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Histona Desacetilasas/química , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Ácido Valproico/farmacología , Acetilación/efectos de los fármacos , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Histona Desacetilasas/metabolismo , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Ratas
18.
Neurosci Res ; 68(1): 35-43, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20562017

RESUMEN

Hexafins are recently identified low-molecular-weight peptide agonists of the fibroblast growth factor receptor (FGFR), derived from the beta6-beta7 loop region of various FGFs. Synthetic hexafin peptides have been shown to bind to and induce tyrosine phosphorylation of FGFR1, stimulate neurite outgrowth, and promote neuronal survival in vitro. Thus, the pronounced biological activities of hexafins in vitro make them attractive compounds for pharmacological studies in vivo. The present study investigated the effects of subcutaneous administration of hexafin1 and hexafin2 (peptides derived from FGF1 and FGF2, respectively) on social memory, exploratory activity, and anxiety-like behavior in adult rats. Treatment with hexafin1 and hexafin2 resulted in prolonged retention of social memory. Furthermore, rats treated with hexafin2 exhibited decreased anxiety-like behavior in the elevated plus maze. Employing an R6/2 mouse model of Huntington's disease (HD), we found that although hexafin2 did not affect the progression of motor symptoms, it alleviated deficits in activity related to social behavior, including sociability and social novelty. Thus, hexafin2 may have therapeutic potential for the treatment of HD.


Asunto(s)
Conducta Animal/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/agonistas , Péptidos/agonistas , Péptidos/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/agonistas , Animales , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Factor 1 de Crecimiento de Fibroblastos/síntesis química , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/síntesis química , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Inyecciones Subcutáneas/métodos , Masculino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/farmacología , Péptidos/síntesis química , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento de Fibroblastos/fisiología
19.
Cell Oncol ; 32(4): 259-74, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20364069

RESUMEN

The epidermal growth factor family of receptor tyrosine kinases (ErbBs) plays essential roles in tumorigenesis and cancer disease progression, and therefore has become an attractive target for structure-based drug design. ErbB receptors are activated by ligand-induced homo- and heterodimerization. Structural studies have revealed that ErbB receptor dimers are stabilized by receptor-receptor interactions, primarily mediated by a region in the second extracellular domain, termed the "dimerization arm". The present study is the first biological characterization of a peptide, termed Inherbin3, which constitutes part of the dimerization arm of ErbB3. Inherbin3 binds to the extracellular domains of all four ErbB receptors, with the lowest peptide binding affinity for ErbB4. Inherbin3 functions as an antagonist of epidermal growth factor (EGF)-ErbB1 signaling. We show that Inherbin3 inhibits EGF-induced ErbB1 phosphorylation, cell growth, and migration in two human tumor cell lines, A549 and HN5, expressing moderate and high ErbB1 levels, respectively. Furthermore, we show that Inherbin3 inhibits tumor growth in vivo and induces apoptosis in a tumor xenograft model employing the human non-small cell lung cancer cell line A549. The Inherbin3 peptide may be a useful tool for investigating the mechanisms of ErbB receptor homo- and heterodimerization. Moreover, the here described biological effects of Inherbin3 suggest that peptide-based targeting of ErbB receptor dimerization is a promising anti-cancer therapeutic strategy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Mucosa Respiratoria/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Trasplante de Neoplasias , Fragmentos de Péptidos/química , Agregación de Receptores/efectos de los fármacos , Receptor ErbB-3/química , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Carga Tumoral/efectos de los fármacos
20.
J Neurochem ; 114(1): 74-86, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20374425

RESUMEN

Basic fibroblast growth factor (FGF2, bFGF) is the most extensively studied member of the FGF family and is involved in neurogenesis, differentiation, neuroprotection, and synaptic plasticity in the CNS. FGF2 executes its pleiotropic biologic actions by binding, dimerizing, and activating FGF receptors (FGFRs). The present study reports the physiologic impact of various FGF2-FGFR1 contact sites employing three different synthetic peptides, termed canofins, designed based on structural analysis of the interactions between FGF2 and FGFR1. Canofins mimic the cognate ligand interaction with the receptor and preserve the neuritogenic and neuroprotective properties of FGF2. Canofins were shown by surface plasmon resonance analysis to bind to FGFR1 and promote receptor activation. However, FGF2-induced receptor phosphorylation was inhibited by canofins, indicating that canofins are partial FGFR agonists. Furthermore, canofins were demonstrated to induce neuronal differentiation determined by neurite outgrowth from cerebellar granule neurons, and this effect was dependent on FGFR activation. Additionally, canofins acted as neuroprotectants, promoting survival of cerebellar granule neurons induced to undergo apoptosis. Our results suggest that canofins mirror the effect of specific interaction sites in FGF2 for FGFR. Thus, canofins are valuable pharmacological tools to study the functional roles of specific molecular interactions of FGF2 with FGFR.


Asunto(s)
Dendrímeros/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neuronas/efectos de los fármacos , Oligopéptidos/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cerebelo/citología , Dendrímeros/química , Agonismo Parcial de Drogas , Factor 2 de Crecimiento de Fibroblastos/química , Ligandos , Modelos Moleculares , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/citología , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Oligopéptidos/química , Fosforilación , Ratas , Ratas Wistar , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/agonistas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA