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












Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 6(1): 583, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37258606

RESUMEN

The ability to image cell chemistry at the nanoscale is key for understanding cell biology, but many optical microscopies are restricted by the ~(200-250)nm diffraction limit. Electron microscopy and super-resolution fluorescence techniques beat this limit, but rely on staining and specialised labelling to generate image contrast. It is challenging, therefore, to obtain information about the functional chemistry of intracellular components. Here we demonstrate a technique for intracellular label-free chemical mapping with nanoscale (~30 nm) resolution. We use a probe-based optical microscope illuminated with a mid-infrared laser whose wavelengths excite vibrational modes of functional groups occurring within biological molecules. As a demonstration, we chemically map intracellular structures in human multiple myeloma cells and compare the morphologies with electron micrographs of the same cell line. We also demonstrate label-free mapping at wavelengths chosen to target the chemical signatures of proteins and nucleic acids, in a way that can be used to identify biochemical markers in the study of disease and pharmacology.


Asunto(s)
Luz , Microscopía , Humanos , Microscopía/métodos , Proteínas , Orgánulos
3.
ACS Appl Mater Interfaces ; 14(42): 47445-47460, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36218307

RESUMEN

A challenge in neurology is the lack of efficient brain-penetrable neuroprotectants targeting multiple disease mechanisms. Plasmonic gold nanostars are promising candidates to deliver standard-of-care drugs inside the brain but have not been trialed as carriers for neuroprotectants. Here, we conjugated custom-made peptide dendrimers (termed H3/H6), encompassing motifs of the neurotrophic S100A4-protein, onto star-shaped and spherical gold nanostructures (H3/H6-AuNS/AuNP) and evaluated their potential as neuroprotectants and interaction with neurons. The H3/H6 nanostructures crossed a model blood-brain barrier, bound to plasma membranes, and induced neuritogenesis with the AuNS, showing higher potency/efficacy than the AuNP. The H3-AuNS/NP protected neurons against oxidative stress, the H3-AuNS being more potent, and against Parkinson's or Alzheimer's disease (PD/AD)-related cytotoxicity. Unconjugated S100A4 motifs also decreased amyloid beta-induced neurodegeneration, introducing S100A4 as a player in AD. Using custom-made dendrimers coupled to star-shaped nanoparticles is a promising route to activate multiple neuroprotective pathways and increase drug potency to treat neurodegenerative disorders.


Asunto(s)
Enfermedad de Alzheimer , Dendrímeros , Fármacos Neuroprotectores , Humanos , Fármacos Neuroprotectores/química , Péptidos beta-Amiloides , Dendrímeros/farmacología , Dendrímeros/uso terapéutico , Neuronas , Oro/química , Enfermedad de Alzheimer/tratamiento farmacológico
5.
ACS Nano ; 14(6): 6866-6877, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32510204

RESUMEN

N-methyl-d-aspartate receptors (NMDARs) are ionotropic glutamate receptors controlling fundamental physiological processes in the central nervous system, such as learning and memory. Excessive activation of NMDARs causes excitotoxicity and results in neurodegeneration, which is observed in a number of pathological conditions. Because of their dichotomous role, therapeutic targeting of NMDAR is difficult. However, several lines of evidence suggest that excitotoxicity is predominantly linked to extrasynaptically located NMDARs. Here, we report on a nanoparticle-based strategy to inhibit extrasynaptic NMDARs exclusively and subtype selectively, while allowing synaptic NMDARs activity. We designed gold nanoparticles (AuNPs) carrying conopeptide derivatives conjugated on their poly(ethylene glycol) coating as allosteric NMDAR inhibitors and show that these nanoparticles antagonize exclusively extrasynaptic NMDAR-mediated currents in cultured hippocampal neurons. Additionally, we show that conopeptide-functionalized AuNPs are neuroprotective in an in vitro model of excitotoxicity. By using AuNPs carrying different allosteric inhibitors with distinct NMDAR subtype selectivity such as peptide conantokin-G or peptide conantokin-R, we suggest activation of extrasynaptic GluN2B-containing diheteromeric NMDARs as the main cause of excitotoxicity.


Asunto(s)
Nanopartículas del Metal , Receptores de N-Metil-D-Aspartato , Oro , Hipocampo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo
6.
Nanoscale ; 11(45): 22054-22069, 2019 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-31720664

RESUMEN

Nanoparticles capable of penetrating the blood-brain barrier (BBB) will greatly advance the delivery of therapies against brain disorders. Carbon nanotubes hold great potential as delivery vehicles due to their high aspect-ratio and cell-penetrating ability. Studies have shown multiwalled carbon nanotubes (MWCNT) cross the BBB, however they have largely relied on labelling methods to track and quantify transport, or on individual electron microscopy images to qualitatively assess transcytosis. Therefore, new direct and quantitative methods, using well-defined and unlabelled MWCNT, are needed to compare BBB translocation of different MWCNT types. Using highly controlled anionic (-), cationic (+) and non-ionic (0) functionalized MWCNT (fMWCNT), we correlate UV-visible spectroscopy with quantitative transmission electron microscopy, quantified from c. 270 endothelial cells, to examine cellular uptake, BBB transport and neurotoxicity of unlabelled fMWCNT. Our results demonstrate that: (i) a large fraction of cationic and non-ionic, but not anionic fMWCNT become trapped at the luminal brain endothelial cell membrane; (ii) despite high cell association, fMWCNT uptake by brain endothelial cells is low (<1.5% ID) and does not correlate with BBB translocation, (iii) anionic fMWCNT have highest transport levels across an in vitro model of the human BBB compared to non-ionic or cationic nanotubes; and (iv) fMWCNT are not toxic to hippocampal neurons at relevant abluminal concentrations; however, fMWCNT charge has an effect on carbon nanotube neurotoxicity at higher fMWCNT concentrations. This quantitative combination of microscopy and spectroscopy, with cellular assays, provides a crucial strategy to predict brain penetration efficiency and neurotoxicity of unlabelled MWCNT and other nanoparticle technologies relevant to human health.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Membrana Celular/metabolismo , Células Endoteliales/metabolismo , Ensayo de Materiales , Nanotubos de Carbono/química , Transcitosis/efectos de los fármacos , Animales , Transporte Biológico , Barrera Hematoencefálica/ultraestructura , Línea Celular Transformada , Membrana Celular/ultraestructura , Células Endoteliales/ultraestructura , Humanos , Nanotubos de Carbono/ultraestructura , Ratas
7.
ACS Appl Mater Interfaces ; 11(40): 36307-36315, 2019 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-31513373

RESUMEN

Molecular mobility in neuronal plasma membranes is a crucial factor in brain function. Microscopic viscosity is an important parameter that determines molecular mobility. This study presents the first direct measurement of the microviscosity of plasma membranes of live neurons. Microviscosity maps were obtained using fluorescence lifetime imaging of environment-sensing dyes termed "molecular rotors". Neurons were investigated both in the basal state and following common neurodegenerative stimuli, excitotoxicity, or oxidative stress. Both types of neurotoxic challenges induced microviscosity decrease in cultured neurons, and oxidant-induced membrane fluidification was counteracted by the wide-spectrum neuroprotectant, the H3 peptide. These results provide new insights into molecular mobility in neuronal membranes, paramount for basic brain function, and suggest that preservation of membrane stability may be an important aspect of neuroprotection in brain insults and neurodegenerative disorders.


Asunto(s)
Membrana Celular/fisiología , Colorantes Fluorescentes/metabolismo , Neuronas/citología , Neuroprotección , Estrés Oxidativo , Animales , Compuestos de Boro/química , Membrana Celular/efectos de los fármacos , Colorantes Fluorescentes/química , Células HeLa , Humanos , Peróxido de Hidrógeno/toxicidad , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fenómenos Ópticos , Estrés Oxidativo/efectos de los fármacos , Ratas , Viscosidad
8.
Nanoscale ; 11(27): 12858-12870, 2019 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-31157349

RESUMEN

There is a need for novel strategies to treat aggressive breast cancer subtypes and overcome drug resistance. ZnO nanoparticles (NPs) have potential in cancer therapy due to their ability to potently and selectively induce cancer cell apoptosis. Here, we tested the in vitro chemotherapeutic efficacy of ZnONPs loaded via a mesoporous silica nanolayer (MSN) towards drug-sensitive breast cancer cells (MCF-7: estrogen receptor-positive, CAL51: triple-negative) and their drug-resistant counterparts (MCF-7TX, CALDOX). ZnO-MSNs were coated on to gold nanostars (AuNSs) for future imaging capabilities in the NIR-II range. Electron and confocal microscopy showed that MSN-ZnO-AuNSs accumulated close to the plasma membrane and were internalized by cells. High-resolution electron microscopy showed that MSN coating degraded outside the cells, releasing ZnONPs that interacted with cell membranes. MSN-ZnO-AuNSs efficiently reduced the viability of all cell lines, and CAL51/CALDOX cells were more susceptible than MCF7/MCF-7-TX cells. MSN-ZnO-AuNSs were then conjugated with the antibody to Frizzled-7 (FZD-7), the receptor upregulated by several breast cancer cells. We used the disulphide (S-S) linker that could be cleaved with a high concentration of glutathione normally observed within cancer cells, releasing Zn2+ into the cytoplasm. FZD-7 targeting resulted in approximately three-fold amplified toxicity of MSN-ZnO-AuNSs towards the MCF-7TX drug-resistant cell line with the highest FZD-7 expression. This study shows that ZnO-MSs are promising tools to treat triple-negative and drug-resistant breast cancers and highlights the potential clinical utility of FZD-7 for delivery of nanomedicines and imaging probes specifically to these cancer types.


Asunto(s)
Antineoplásicos Inmunológicos , Neoplasias de la Mama/tratamiento farmacológico , Portadores de Fármacos , Receptores Frizzled/antagonistas & inhibidores , Nanopartículas , Óxido de Zinc , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Supervivencia Celular , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Liberación de Fármacos , Femenino , Receptores Frizzled/metabolismo , Humanos , Células MCF-7 , Nanopartículas/química , Nanopartículas/uso terapéutico , Óxido de Zinc/química , Óxido de Zinc/farmacología
9.
Soft Matter ; 14(46): 9466-9474, 2018 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-30427370

RESUMEN

Amyloid deposits of aggregated beta-amyloid Aß(1-42) peptides are a pathological hallmark of Alzheimer's disease. Aß(1-42) aggregates are known to induce biophysical alterations in cells, including disruption of plasma membranes. We investigated the microviscosity of plasma membranes upon interaction with oligomeric and fibrillar forms of Aß(1-42). Viscosity-sensing fluorophores termed molecular rotors were utilised to directly measure the microviscosities of giant plasma membrane vesicles (GPMVs) and plasma membranes of live SH-SY5Y and HeLa cells. The fluorescence lifetimes of membrane-inserting BODIPY-based molecular rotors revealed a decrease in bilayer microviscosity upon incubation with Aß(1-42) oligomers, while fibrillar Aß(1-42) did not significantly affect the microviscosity of the bilayer. In addition, we demonstrate that the neuroprotective peptide H3 counteracts the microviscosity change induced by Aß(1-42) oligomers, suggesting the utility of H3 as a neuroprotective therapeutic agent in neurodegenerative disorders and indicating that ligand-induced membrane stabilisation may be a possible mechanism of neuroprotection during neurodegenerative disorders such as Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Compuestos de Boro/farmacología , Membrana Celular/efectos de los fármacos , Colorantes Fluorescentes/farmacología , Fragmentos de Péptidos/farmacología , Línea Celular Tumoral , Membrana Celular/fisiología , Humanos , Neuropéptidos/farmacología , Viscosidad
10.
Theranostics ; 8(14): 3977-3990, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30083275

RESUMEN

Understanding the mechanisms of neurodegeneration is crucial for development of therapies to treat neurological disorders. S100 proteins are extensively expressed in the injured brain but S100's role and signalling in neural cells remain elusive. We recently demonstrated that the S100A4 protein protects neurons in brain injury and designed S100A4-derived peptides mimicking its beneficial effects. Here we show that neuroprotection by S100A4 involves the growth factor family receptor ErbB4 and its ligand Neuregulin 1 (NRG), key regulators of neuronal plasticity and implicated in multiple brain pathologies. The neuroprotective effect of S100A4 depends on ErbB4 expression and the ErbB4 signalling partners ErbB2/Akt, and is reduced by functional blockade of NRG/ErbB4 in cell models of neurodegeneration. We also detect binding of S100A4 with ErbB1 (EGFR) and ErbB3. S100A4-derived peptides interact with, and signal through ErbB, are neuroprotective in primary and immortalized dopaminergic neurons, and do not affect cell proliferation/motility - features which make them promising as potential neuroprotectants. Our data suggest that the S100-ErbB axis may be an important mechanism regulating neuronal survival and plasticity.


Asunto(s)
Neuronas/fisiología , Receptor ErbB-4/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Animales , Supervivencia Celular , Neurregulina-1/metabolismo , Plasticidad Neuronal , Ratas
11.
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
12.
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
13.
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
14.
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
15.
Brain ; 133(Pt 8): 2281-94, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20435631

RESUMEN

Erythropoietin, a member of the type 1 cytokine superfamily, controls proliferation and differentiation of erythroid progenitor cells through binding to and dimerization of the erythropoietin receptor. Both erythropoietin and its receptor are also expressed in the central nervous system, where they are involved in tissue protection. However, the use of erythropoietin as a neuroprotective agent may be hampered by its erythropoietic activity. Therefore, developing non-haematopoietic erythropoietin mimetics is important. Based on the crystal structure of the complex of erythropoietin and its receptor, we designed a peptide, termed Epotris, corresponding to the C α-helix region (amino-acid residues 92-111) of human erythropoietin. The peptide specifically bound to the erythropoietin receptor and promoted neurite outgrowth and survival of primary neurons with the same efficiency as erythropoietin, but with 10(3)-fold lower potency. Knockdown of the erythropoietin receptor or interference with its downstream signalling inhibited the Epotris-induced neuritogenic and pro-survival effect. Similarly to erythropoietin, Epotris penetrated the blood-brain barrier. Moreover, treatment with the peptide attenuated seizures, decreased mortality and reduced neurodegeneration in an in vivo model of kainic acid-induced neurotoxicity. In contrast to erythropoietin, Epotris did not stimulate erythropoiesis upon chronic administration. Thus, Epotris is a novel neuroprotective non-haematopoietic erythropoietin mimetic that may offer new opportunities for the treatment of neurological disorders.


Asunto(s)
Eritropoyetina/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Eritropoyetina/agonistas , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Células Cultivadas , Eritropoyesis/efectos de los fármacos , Eritropoyesis/fisiología , Eritropoyetina/química , Eritropoyetina/metabolismo , Eritropoyetina/farmacocinética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neuritas/efectos de los fármacos , Neuritas/fisiología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/mortalidad , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacocinética , Ratas , Ratas Wistar , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/mortalidad
16.
FASEB J ; 24(4): 1139-50, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19952283

RESUMEN

Neuroplastin (Np) is a glycoprotein belonging to the immunoglobulin superfamily of cell adhesion molecules (CAMs) and existing in two isoforms, Np55 and Np65, named according to their molecular weights. The extracellular part of Np65 contains three immunoglobulin (Ig)-like modules (Ig1, Ig2, and Ig3), whereas Np55 lacks the Ig1 module. Of these two isoforms, only Np65 is involved in homophilic interactions resulting in cell adhesion, whereas the role of Np55 is poorly understood. The present study reports for the first time the crystal structure of the ectodomain of Np55 at 1.95-A resolution and demonstrates that Np55 binds to and activates the fibroblast growth factor receptor 1 (FGFR1). Furthermore, we identify a sequence motif in the Ig2 module of Np55 interacting with FGFR1 and show that a synthetic peptide encompassing this motif, termed narpin, binds to and activates FGFR1. We show that both Np55 and the narpin peptide induce neurite outgrowth through FGFR1 activation and that Np55 increases synaptic calcium concentration in an FGFR1-dependent manner. Moreover, we demonstrate that narpin has an antidepressive-like effect in rats subjected to the forced swim test, suggesting that Np55-induced signaling may be involved in synaptic plasticity in vivo. Owczarek, S., Kiryushko, D., Larsen, M. H., Kastrup, J. S., Gajhede, M., Sandi, C., Berezin, V., Bock, E., Soroka, V. Neuroplastin-55 binds to and signals through the fibroblast growth factor receptor.


Asunto(s)
Calcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Péptidos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Sinapsis/metabolismo , Secuencias de Aminoácidos , Animales , Antidepresivos/química , Antidepresivos/metabolismo , Antidepresivos/farmacología , Línea Celular , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/farmacología , Neuritas/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Péptidos/química , Péptidos/farmacología , Condicionamiento Físico Animal , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Natación
17.
J Neurosci Res ; 88(5): 1074-82, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19937811

RESUMEN

Metallothioneins I and II (MTI/II) are metal-binding proteins overexpressed in response to brain injury. Recently, we have designed a peptide, termed EmtinB, which is modeled after the beta-domain of MT-II and mimics the biological effects of MTI/II in vitro. Here, we demonstrate the neuroprotective effect of EmtinB in the in vitro and in vivo models of kainic acid (KA)-induced neurotoxicity. We show that EmtinB passes the blood-brain barrier and is detectable in plasma for up to 24 hr. Treatment with EmtinB significantly attenuates seizures in C57BL/6J mice exposed to moderate (20 mg/kg) and high (30 mg/kg) KA doses and tends to decrease mortality induced by the high KA dose. Histopathological evaluation of hippocampal (CA3 and CA1) and cortical areas of mice treated with 20 mg/kg KA shows that EmtinB treatment reduces KA-induced neurodegeneration in the CA1 region. These findings establish EmtinB as a promising target for therapeutic development.


Asunto(s)
Ácido Kaínico/antagonistas & inhibidores , Metalotioneína/agonistas , Fármacos Neuroprotectores/farmacocinética , Neurotoxinas/antagonistas & inhibidores , Péptidos/farmacocinética , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiopatología , Péptidos y Proteínas de Señalización Intercelular , Ácido Kaínico/toxicidad , Masculino , Metalotioneína/metabolismo , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Péptidos/sangre , Péptidos/uso terapéutico , Ratas , Ratas Wistar , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología
18.
FEBS J ; 276(20): 5936-48, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19740107

RESUMEN

The function of S100A4, a member of the calcium-binding S100 protein family, has been associated with tumor invasion and metastasis. Although an essential pro-metastatic role of extracellular S100A4 in tumor progression has been demonstrated, the identification of the precise underlying mechanisms and protein partners (receptors) has remained elusive. To identify putative targets for extracellular S100A4, we screened a phage display peptide library using S100A4 as bait. We identified three independent peptide motifs with varying affinities for the S100A4 protein. Sequence analyses indicated that the most abundant peptide mimicked the F/YCC motif present in the epidermal growth factor domain of ErbB receptor ligands. S100A4 selectively interacted with a number of epidermal growth factor receptor (EGFR) ligands, demonstrating highest affinity for amphiregulin. Importantly, we found that S100A4 stimulated EGFR/ErbB2 receptor signaling and enhanced the amphiregulin-mediated proliferation of mouse embryonic fibroblasts. S100A4-neutralizing antibodies, as well as EGFR- and ErbB2 receptor-specific tyrosine kinase inhibitors, blocked these effects. The present results suggest that extracellular S100A4 regulates tumor progression by interacting with EGFR ligands, thereby enhancing EGFR/ErbB2 receptor signaling and cell proliferation. Structured digital abstract: * MINT-7256556: EGF (uniprotkb:P01133) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256512: BC (uniprotkb:P35070) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256485, MINT-7256618, MINT-7256636: AR (uniprotkb:P15514) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256494: HB-EGF (uniprotkb:Q99075) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256502: P53 (uniprotkb:P04637) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256654: S100A2 (uniprotkb:P29034) binds (MI:0407) to AR (uniprotkb:P15514) by far western blotting (MI:0047) * MINT-7256693: S100A5 (uniprotkb:P33763) binds (MI:0407) to AR (uniprotkb:P15514) by far western blotting (MI:0047) * MINT-7256593: S100A4 (uniprotkb:P26447) binds (MI:0407) to BC (uniprotkb:P35070) by pull down (MI:0096) * MINT-7256567: S100A4 (uniprotkb:P26447) binds (MI:0407) to AR (uniprotkb:P15514) by pull down (MI:0096).


Asunto(s)
Receptores ErbB/metabolismo , Péptidos/metabolismo , Proteínas S100/química , Proteínas S100/metabolismo , Secuencia de Aminoácidos , Anfirregulina , Animales , Sitios de Unión , Far-Western Blotting , Western Blotting , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatografía en Gel , Familia de Proteínas EGF , Ensayo de Inmunoadsorción Enzimática , Glicoproteínas/metabolismo , Glicoproteínas/farmacología , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Datos de Secuencia Molecular , Biblioteca de Péptidos , Péptidos/química , Péptidos/genética , Unión Proteica , Receptor ErbB-2/metabolismo , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Proteínas S100/farmacología , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
19.
J Neurochem ; 100(6): 1599-612, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17212696

RESUMEN

The neural cell adhesion molecule (NCAM), and the growth-associated protein (GAP-43), play pivotal roles in neuronal development and plasticity and possess interdependent functions. However, the mechanisms underlying the functional association of GAP-43 and NCAM have not been elucidated. In this study we show that (over)expression of GAP-43 in PC12E2 cells and hippocampal neurons strongly potentiates neurite extension, both in the absence and in the presence of homophilic NCAM binding. This potentiation is crucially dependent on the membrane association of GAP-43. We demonstrate that phosphorylation of GAP-43 by protein kinase C (PKC) as well as by casein kinase II (CKII) is important for the NCAM-induced neurite outgrowth. Moreover, our results indicate that in the presence of GAP-43, NCAM-induced neurite outgrowth requires functional association of NCAM-180/spectrin/GAP-43, whereas in the absence of GAP-43, the NCAM-140/non-receptor tyrosine kinase (Fyn)-associated signaling pathway is pivotal. Thus, expression of GAP-43 presumably acts as a functional switch for NCAM-180-induced signaling. This suggests that under physiological conditions, spatial and/or temporal changes of the localization of GAP-43 and NCAM on the cell membrane may determine the predominant signaling mechanism triggered by homophilic NCAM binding: NCAM-180/spectrin-mediated modulation of the actin cytoskeleton, NCAM-140-mediated activation of Fyn, or both.


Asunto(s)
Proteína GAP-43/fisiología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuritas/fisiología , Neuronas/citología , Animales , Células Cultivadas , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Ratones , Modelos Biológicos , Mutagénesis/fisiología , Moléculas de Adhesión de Célula Nerviosa/genética , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Ratas , Sinaptosomas/metabolismo , Transfección/métodos
20.
Curr Neuropharmacol ; 5(4): 253-67, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19305742

RESUMEN

Cell adhesion molecules (CAMs) play a pivotal role in the development and maintenance of the nervous system under normal conditions. They also are involved in numerous pathological processes such as inflammation, degenerative disorders, and cancer, making them attractive targets for drug development. The majority of CAMs are signal transducing receptors. CAM-induced intracellular signalling is triggered via homophilic (CAM-CAM) and heterophilic (CAM - other counter-receptors) interactions, which both can be targeted pharmacologically. We here describe the progress in the CAM pharmacology focusing on cadherins and CAMs of the immunoglobulin (Ig) superfamily, such as NCAM and L1. Structural basis of CAM-mediated cell adhesion and CAM-induced signalling are outlined. Different pharmacological approaches to study functions of CAMs are presented including the use of specific antibodies, recombinant proteins, and synthetic peptides. We also discuss how unravelling of the 3D structure of CAMs provides novel pharmacological tools for dissection of CAM-induced signalling pathways and offers therapeutic opportunities for a range of neurological disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...