Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Intervalo de año de publicación
1.
Am J Pathol ; 193(7): 977-994, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37037285

RESUMEN

Congenital toxoplasmosis constitutes a major cause of pre- and postnatal complications. Fetal infection with Toxoplasma gondii influences development and can lead to microcephaly, encephalitis, and neurologic abnormalities. Systematic studies concerning the effects of neural progenitor cell infection with T. gondii are unavailable. Cortical intermediate progenitor cells cultivated as neurospheres obtained from E16.5 Swiss Webster mice were infected with T. gondii (ME49 strain) tachyzoites to mimic the developing mouse cerebral cortex in vitro. Infection was associated with decreased cell proliferation, detected by Ki-67 staining at 48 and 72 hours after infection in floating neurospheres, and reduced cellularity at 96 hours. Transient decreases in the expression of the neurogenesis-related transcription factors T-box brain protein 1, mouse atonal homolog protein 1, and hairy and enhancer of split protein 1 were found in infected cultures, while the level of transcription factor SOX-2 remained unaltered. Neurogenic potential, assessed in plated neurospheres, was impaired in infected cultures, as indicated by decreased late neuronal marker neurofilament heavy chain immunoreactivity. Infected cultures exhibited decreased overall migration rates at 48 and 120 hours. These findings indicate that T. gondii infection of neural progenitor cells may lead to reduced neurogenesis due to an imbalance in cell proliferation alongside an altered migratory profile. If translated to the in vivo situation, these data could explain, in part, cortical malformations in congenitally infected individuals.


Asunto(s)
Células-Madre Neurales , Toxoplasma , Ratones , Animales , Neuronas , Neurogénesis , Proliferación Celular
2.
Exp Eye Res ; 200: 108212, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32910940

RESUMEN

PURPOSE: Cellular therapy with mesenchymal stem cells (MSC) is emerging as an effective option to treat optic neuropathies. In models of retinal degeneration, MSC injected in the vitreous body protects injured retinal ganglion cells and stimulate their regeneration, however the mechanism is still unknown. Considering the immunomodulating proprieties of MSC and the controversial role of microglial contribution on retinal regeneration, we developed an in vitro co-culture model to analyze the effect of MSC on retinal microglia population. METHODS: We used whole adult rat retinal explants in co-culture with human Wharton's jelly mesenchymal stem cells (hMSC) separated by a transwell membrane and analyzed hMSC effect on both retinal ganglion cells (RGCs) and retinal microglia. RESULTS: hMSC in co-culture protected RGCs after 3 days in vitro by paracrine signaling. In addition, hMSC reduced microglia population and inhibited the pro-inflammatory phenotype of the remaining microglia. CONCLUSIONS: Using a co-culture model, we demonstrated the paracrine effect of hMSC on RGC survival after injury concomitant with a reduction of microglial population. Paracrine signaling of hMSC also changed microglia phenotype and the expression of antiinflammatory factors in the retina. Our results are consistent with a detrimental effect of microglia on RGC survival and regeneration after injury.


Asunto(s)
Células Madre Mesenquimatosas/citología , Microglía/patología , Regeneración Nerviosa , Comunicación Paracrina/fisiología , Degeneración Retiniana/diagnóstico , Células Ganglionares de la Retina/patología , Animales , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Masculino , Microglía/metabolismo , Fenotipo , Ratas , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo
3.
Stem Cell Res Ther ; 10(1): 121, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30995945

RESUMEN

BACKGROUND: Retina and/or optic nerve injury may cause irreversible blindness, due to degeneration of retinal ganglion cells. We and others have previously shown that the intravitreal injection of mesenchymal stem cells (MSCs) protects injured retinal ganglion cells and stimulates their regeneration after optic nerve injury, but the long-term effects of this therapy are still unknown. METHODS: We injected rat MSC (rMSC) intravitreally in adult (3-5 months) Lister Hooded rats of either sex after optic nerve crush. Retinal ganglion cell survival, axonal regeneration, and reconnection were analyzed 60 and 240 days after crush by immunohistochemistry for Tuj1, anterograde labeling with cholera-toxin B and by immunohistochemistry for nerve growth factor-induced gene A (NGFI-A, driven by light stimulation) in the superior colliculus after a cycle of light deprivation-stimulation. Visual behaviors (optokinetic reflex, looming response, and preference for dark) were analyzed 70 days after crush. RESULTS: rMSC treatment doubled the number of surviving retinal ganglion cells, preferentially of a larger subtype, and of axons regenerating up to 0.5 mm. Some axons regenerated to the lateral geniculate nucleus and superior colliculus. NGFI-A+ cells were doubled in rMSC-treated animals 60 days after crush, but equivalent to vehicle-injected animals 240 days after crush, suggesting that newly formed synapses degenerated. Animals did not recover visual behaviors. CONCLUSIONS: We conclude that rMSC-induced neuroprotection is sustained at longer time points. Although rMSCs promoted long-term neuroprotection and long-distance axon regeneration, the reconnection of retinal ganglion cells with their targets was transitory, indicating that they need additional stimuli to make stable reconnections.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Regeneración Nerviosa , Traumatismos del Nervio Óptico , Nervio Óptico/fisiología , Aloinjertos , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Masculino , Células Madre Mesenquimatosas/patología , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/terapia , Ratas , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología
4.
Brain Res ; 1712: 73-81, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30735638

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a chronic degenerative disease that mainly affects motor neurons, leading to progressive paralysis and death. Recently, cell therapy has emerged as a therapeutic alternative for several neurological diseases, including ALS, and bone-marrow cells are one of the major cell sources. Considering the importance of pre-clinical trials to determine the best therapeutic protocol and the hope of translating this protocol to the clinical setting, we tested bone-marrow mononuclear cell (BMMC) therapy administered by different routes in the SOD1G93A model of ALS. BMMCs were isolated from non-transgenic, age matched animals and administered intravenously (IV), intramuscularly (IM), and intravenously and intramuscular concomitantly (IV + IM). BMMC therapy had no significant beneficial effects when injected IV or IM, but delayed disease progression when these two routes were used concomitantly. BMMC IV + IM treatment reduced the number of microglia cells in the spinal cord and partially protected of neuromuscular-junction innervation, but had no effect in preventing motor-neuron loss. This study showed that injection of BMMC IV + IM had better results when compared to each route in isolation, highlighting the importance of targeting multiple anatomical regions in the treatment of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Administración Intravenosa/métodos , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inyecciones Intramusculares/métodos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Unión Neuromuscular/metabolismo , Médula Espinal/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/metabolismo
5.
Mol Neurobiol ; 56(2): 1056-1069, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29869196

RESUMEN

Retinal ganglion cell (RGC) degeneration occurs within 2 weeks following optic nerve crush (ONC) as a consequence of reduced retro-transport of growth factors including nerve growth factor (NGF). The hypothesis that intravitreal (ivt) and eye drop (ed) administration of recombinant human NGF (rhNGF) might counteract ONC in adult rats is explored in this study. We found that both ivt- and ed-rhNGF reduced RGC loss and stimulated axonal regrowth. Chiefly, survival and regenerative effects of rhNGF were associated with a reduction of cells co-expressing Nogo-A/p75NTR at crush site borders, which contribute to glia scar formation following nerve injury, and induce further degeneration. We also found that ocular application of rhNGF reduced p75NTR and proNGF and enhanced phosphorylation of TrkA and its intracellular signals at retina level. Nogo-R and Rock2 expression was also normalized by ed-rhNGF treatment in both ONC and contralateral retina. Our findings that ocular applied NGF reaches and exerts biological actions on posterior segment of the eye give a further insight into the neurotrophin diffusion/transport through eye structures and/or their trafficking in optic nerve. In addition, the use of a highly purified NGF form in injury condition in which proNGF/p75NTR binding is favored indicates that increased availability of mature NGF restores the balance between TrkA and p75NGF, thus resulting in RGC survival and axonal growth. In conclusion, ocular applied NGF is confirmed as a good experimental paradigm to study mechanisms of neurodegeneration and regeneration, disclose biomarkers, and time windows for efficacy treatment following cell or nerve injury.


Asunto(s)
Factor de Crecimiento Nervioso/farmacología , Traumatismos del Nervio Óptico/inducido químicamente , Nervio Óptico/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Masculino , Modelos Teóricos , Proteínas Nogo/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Ratas Long-Evans , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo
6.
Front Immunol ; 9: 1173, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29896197

RESUMEN

Recombinant filamentous fd bacteriophages (rfd) expressing antigenic peptides were shown to induce cell-mediated immune responses in the absence of added adjuvant, being a promising delivery system for vaccination. Here, we tested the capacity of rfd phages to protect against infection with the human protozoan Trypanosoma cruzi, the etiologic agent of Chagas Disease. For this, C57BL/6 (B6) and Tlr9-/- mice were vaccinated with rfd phages expressing the OVA257-264 peptide or the T. cruzi-immunodominant peptides PA8 and TSKB20 and challenged with either the T. cruzi Y-OVA or Y-strain, respectively. We found that vaccination with rfd phages induces anti-PA8 and anti-TSKB20 IgG production, expansion of Ag-specific IFN-γ, TNF-α, and Granzyme B-producing CD8+ T cells, as well as in vivo Ag-specific cytotoxic responses. Moreover, the fd-TSKB20 vaccine was able to protect against mortality induced by a high-dose inoculum of the parasite. Although vaccination with rfd phages successfully reduced both parasitemia and parasite load in the myocardium of WT B6 mice, Tlr9-/- animals were not protected against infection. Thus, our data extend previous studies, demonstrating that rfd phages induce Ag-specific IgG and CD8+ T cell-mediated responses and confer protection against an important human parasite infection, through a TLR9-dependent mechanism.


Asunto(s)
Bacteriófago M13 , Enfermedad de Chagas , Regulación de la Expresión Génica , Vacunas Antiprotozoos , Receptor Toll-Like 9 , Trypanosoma cruzi , Vacunación , Animales , Bacteriófago M13/genética , Bacteriófago M13/inmunología , Enfermedad de Chagas/genética , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/prevención & control , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Noqueados , Vacunas Antiprotozoos/genética , Vacunas Antiprotozoos/inmunología , Vacunas Antiprotozoos/farmacología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Trypanosoma cruzi/genética , Trypanosoma cruzi/inmunología
7.
Stem Cells Int ; 2017: 5759490, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29270199

RESUMEN

CD60b antigens are highly expressed during development in the rat nervous system, while in the adult their expression is restricted to a few regions, including the subventricular zone (SVZ) around the lateral ventricles-a neurogenic niche in the adult brain. For this reason, we investigated whether the expression of C60b is associated with neural stem/progenitor cells in the SVZ, from development into adulthood. We performed in vitro and in vivo analyses of CD60b expression at different stages and identified the presence of these antigens in neural stem/progenitor cells. We also observed that CD60b could be used to purify and enrich a population of neurosphere-forming cells from the developing and adult brain. We showed that CD60b antigens (mainly corresponding to ganglioside 9-O-acetyl GD3, a well-known molecule expressed during central nervous system development and mainly associated with neuronal migration) are also present in less mature cells and could be used to identify and isolate neural stem/progenitor cells during development and in the adult brain. A better understanding of molecules associated with neurogenesis may contribute not only to improve the knowledge about the physiology of the mammalian central nervous system, but also to find new treatments for regenerating tissue after disease or brain injury.

8.
Int J Mol Sci ; 18(1)2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28067793

RESUMEN

Nerve growth factor (NGF) is suggested to be neuroprotective after nerve injury; however, retinal ganglion cells (RGC) degenerate following optic-nerve crush (ONC), even in the presence of increased levels of endogenous NGF. To further investigate this apparently paradoxical condition, a time-course study was performed to evaluate the effects of unilateral ONC on NGF expression and signaling in the adult retina. Visually evoked potential and immunofluorescence staining were used to assess axonal damage and RGC loss. The levels of NGF, proNGF, p75NTR, TrkA and GFAP and the activation of several intracellular pathways were analyzed at 1, 3, 7 and 14 days after crush (dac) by ELISA/Western Blot and PathScan intracellular signaling array. The progressive RGC loss and nerve impairment featured an early and sustained activation of apoptotic pathways; and GFAP and p75NTR enhancement. In contrast, ONC-induced reduction of TrkA, and increased proNGF were observed only at 7 and 14 dac. We propose that proNGF and p75NTR contribute to exacerbate retinal degeneration by further stimulating apoptosis during the second week after injury, and thus hamper the neuroprotective effect of the endogenous NGF. These findings might aid in identifying effective treatment windows for NGF-based strategies to counteract retinal and/or optic-nerve degeneration.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Traumatismos del Nervio Óptico/complicaciones , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Transducción de Señal , Animales , Apoptosis , Western Blotting , Potenciales Evocados Visuales/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Microscopía Fluorescente , Compresión Nerviosa , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Ratas , Ratas Long-Evans , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Retina/metabolismo , Retina/fisiopatología , Degeneración Retiniana/etiología , Degeneración Retiniana/fisiopatología , Factores de Tiempo
9.
Stem Cell Res Ther ; 7: 41, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26979533

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a progressive neurological disease that selectively affects the motor neurons. The details of the mechanisms of selective motor-neuron death remain unknown and no effective therapy has been developed. We investigated the therapy with bone-marrow mononuclear cells (BMMC) in a mouse model of ALS (SOD1(G93A) mice). METHODS: We injected 10(6) BMMC into the lumbar portion of the spinal cord of SOD1(G93A) mice in presymptomatic (9 weeks old) and symptomatic (14 weeks old) phases. In each condition, we analyzed the progression of disease and the lifespan of the animals. RESULTS: We observed a mild transitory delay in the disease progression in the animals injected with BMMC in the presymptomatic phase. However, we observed no increase in the lifespan. When we injected BMMC in the symptomatic phase, we observed no difference in the animals' lifespan or in the disease progression. Immunohistochemistry for NeuN showed a decrease in the number of motor neurons during the course of the disease, and this decrease was not affected by either treatment. Using different strategies to track the BMMC, we noted that few cells remained in the spinal cord after transplantation. This observation could explain why the BMMC therapy had only a transitory effect. CONCLUSION: This is the first report of intraspinal BMMC therapy in a mouse model of ALS. We conclude this cellular therapy has only a mild transitory effect when performed in the presymptomatic phase of the disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Enfermedades Asintomáticas/terapia , Trasplante de Médula Ósea , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Células del Asta Anterior/fisiología , Movimiento Celular , Supervivencia Celular , Rastreo Celular , Femenino , Inyecciones Espinales , Región Lumbosacra/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/fisiología , Actividad Motora , Mutación Missense , Recuperación de la Función , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
10.
Stem Cells Int ; 2016: 5078619, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26649049

RESUMEN

Following optic nerve injury associated with acute or progressive diseases, retinal ganglion cells (RGCs) of adult mammals degenerate and undergo apoptosis. These diseases have limited therapeutic options, due to the low inherent capacity of RGCs to regenerate and due to the inhibitory milieu of the central nervous system. Among the numerous treatment approaches investigated to stimulate neuronal survival and axonal extension, cell transplantation emerges as a promising option. This review focuses on cell therapies with bone marrow mononuclear cells and bone marrow-derived mesenchymal stem cells, which have shown positive therapeutic effects in animal models of optic neuropathies. Different aspects of available preclinical studies are analyzed, including cell distribution, potential doses, routes of administration, and mechanisms of action. Finally, published and ongoing clinical trials are summarized.

11.
BMC Cancer ; 14: 866, 2014 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-25417721

RESUMEN

BACKGROUND: Signal transducer and activator of transcription 3 (STAT3) is an important transcriptional factor frequently associated with the proliferation and survival of a large number of distinct cancer types. However, the signaling pathways and mechanisms that regulate STAT3 activation remain to be elucidated. METHODS: In this study we took advantage of existing cellular models for chronic myeloid leukemia resistance, western blot, in vitro signaling, real time PCR, flow cytometry approaches for cell cycle and apoptosis evaluation and siRNA assay in order to investigate the possible relationship between STATIP1, STAT3 and CML resistance. RESULTS: Here, we report the characterization of STAT3 protein regulation by STAT3-interacting protein (STATIP1) in the leukemia cell line K562, which demonstrates constitutive BCR-ABL TK activity. K562 cells exhibit high levels of phosphorylated STAT3 accumulated in the nucleus and enhanced BCR-ABL-dependent STAT3 transcriptional activity. Moreover, we demonstrate that STATIP1 is not involved in either BCR-ABL or STAT3 signaling but that STATIP1 is involved in the down-regulation of STAT3 transcription levels; STATIP1-depleted K562 cells display increased proliferation and increased levels of the anti-apoptosis STAT3 target genes CCND1 and BCL-XL, respectively. Furthermore, we demonstrated that Lucena, an Imatinib (IM)-resistant cell line, exhibits lower STATIP1 mRNA levels and undergoes apoptosis/cell cycle arrest in response to STAT3 inhibition together with IM treatment. We provide evidence that STATIP1 siRNA could confer therapy resistance in the K562 cells. Moreover, analysis of CML patients showed an inverse expression of STAIP1 and STAT3 mRNA levels, ratifying that IM-resistant patients present low STATIP1/high STAT3 mRNA levels. CONCLUSIONS: Our data suggest that STATIP1 may be a negative regulator of STAT3 and demonstrate its involvement in IM therapy resistance in CML.


Asunto(s)
Benzamidas/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Piperazinas/farmacología , Pirimidinas/farmacología , Factor de Transcripción STAT3/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Masculino , Persona de Mediana Edad , Fosforilación , Factor de Transcripción STAT3/genética , Regulación hacia Arriba , Adulto Joven
12.
PLoS One ; 9(10): e110722, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25347773

RESUMEN

Bone marrow-derived cells have been used in different animal models of neurological diseases. We investigated the therapeutic potential of mesenchymal stem cells (MSC) injected into the vitreous body in a model of optic nerve injury. Adult (3-5 months old) Lister Hooded rats underwent unilateral optic nerve crush followed by injection of MSC or the vehicle into the vitreous body. Before they were injected, MSC were labeled with a fluorescent dye or with superparamagnetic iron oxide nanoparticles, which allowed us to track the cells in vivo by magnetic resonance imaging. Sixteen and 28 days after injury, the survival of retinal ganglion cells was evaluated by assessing the number of Tuj1- or Brn3a-positive cells in flat-mounted retinas, and optic nerve regeneration was investigated after anterograde labeling of the optic axons with cholera toxin B conjugated to Alexa 488. Transplanted MSC remained in the vitreous body and were found in the eye for several weeks. Cell therapy significantly increased the number of Tuj1- and Brn3a-positive cells in the retina and the number of axons distal to the crush site at 16 and 28 days after optic nerve crush, although the RGC number decreased over time. MSC therapy was associated with an increase in the FGF-2 expression in the retinal ganglion cells layer, suggesting a beneficial outcome mediated by trophic factors. Interleukin-1ß expression was also increased by MSC transplantation. In summary, MSC protected RGC and stimulated axon regeneration after optic nerve crush. The long period when the transplanted cells remained in the eye may account for the effect observed. However, further studies are needed to overcome eventually undesirable consequences of MSC transplantation and to potentiate the beneficial ones in order to sustain the neuroprotective effect overtime.


Asunto(s)
Axones/metabolismo , Células Madre Mesenquimatosas/metabolismo , Regeneración Nerviosa , Neuronas/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/terapia , Nervio Óptico , Adipocitos/citología , Animales , Antígenos de Superficie/metabolismo , Diferenciación Celular , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Condrocitos/citología , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Expresión Génica , Inmunofenotipificación , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Imagen por Resonancia Magnética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Compresión Nerviosa , Traumatismos del Nervio Óptico/diagnóstico , Osteocitos/citología , Ratas , Células Ganglionares de la Retina/metabolismo
13.
Brain Res ; 1587: 54-68, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25204691

RESUMEN

In adult mammals, the regeneration of the optic nerve is very limited and at the moment there are several groups trying different approaches to increase retinal ganglion cell (RGC) survival and axonal outgrowth. One promising approach is cell therapy. In previous work, we performed intravitreal transplantation of bone-marrow mononuclear cells (BMMCs) after optic nerve crush in adult rats and we demonstrated an increase in RGC survival and axon outgrowth 14 days after injury. In the present work, we investigated if these results could be sustained for a longer period of time. Optic nerve crush was performed in Lister-hooded adult rats and BMMC or saline injections were performed shortly after injury. Neuronal survival and regeneration were evaluated in rats׳ retina and optic nerve after 28 days. We demonstrated an increase of 5.2 fold in the axon outgrowth 28 days after lesion, but the BMMCs had no effect on RGC survival. In an attempt to prolong RGC survival, we established a new protocol with two BMMC injections, the second one 7 days after the injury. Untreated animals received two injections of saline. We observed that although the axonal outgrowth was still increased after the second BMMC injection, the RGC survival was not significantly different from untreated animals. These results demonstrate that BMMCs transplantation promotes neuroregeneration at least until 28 days after injury. However, the effects on RGC survival previously observed by us at 14 days were not sustained at 28 days and could not be prolonged with a second dose of BMMC.


Asunto(s)
Axones/fisiología , Trasplante de Médula Ósea , Regeneración Nerviosa , Traumatismos del Nervio Óptico/terapia , Animales , Transporte Axonal , Supervivencia Celular , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Perfilación de la Expresión Génica , Supervivencia de Injerto , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Compresión Nerviosa , Traumatismos del Nervio Óptico/fisiopatología , Ratas , Células Ganglionares de la Retina/patología
14.
ASN Neuro ; 4(6): 357-69, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22894715

RESUMEN

Previous studies indicated that a ganglioside 9acGD3 (9-O-acetyl GD3) antibody [the J-Ab (Jones antibody)] reduces GCP (granule cell progenitor) migration in vitro and in vivo. We here investigated, using cerebellar explants of post-natal day (P) 6 mice, the mechanism by which 9acGD3 reduces GCP migration. We found that immunoblockade of the ganglioside with the J-Ab or the lack of GD3 synthase reduced GCP in vitro migration and the frequency of Ca(2+) oscillations. Immunocytochemistry and pharmacological assays indicated that GCPs expressed P2Y(1)Rs (P2Y(1) receptors) and that deletion or blockade of these receptors decreased the migration rate of GCPs and the frequency of Ca(2+) oscillations. The reduction in P2Y(1)-mediated calcium signals seen in Jones-treated and GD3 synthase-null GCPs were paralleled by P2Y(1)R internalization. We conclude that 9acGD3 controls GCP migration by influencing P2Y(1)R cellular distribution and function.


Asunto(s)
Señalización del Calcio/genética , Movimiento Celular/fisiología , Cerebelo/citología , Gangliósidos/metabolismo , Células-Madre Neurales/fisiología , Receptores Purinérgicos P2Y1/metabolismo , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Gangliósidos/deficiencia , Gangliósidos/inmunología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Técnicas de Cultivo de Órganos , Agonistas del Receptor Purinérgico P2Y/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/deficiencia , Receptores Purinérgicos P2Y1/genética , Transfección , Tubulina (Proteína)/metabolismo
15.
Invest Ophthalmol Vis Sci ; 53(8): 4720-9, 2012 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-22695963

RESUMEN

PURPOSE: Bone marrow mononuclear cells (BMMCs) have been used with considerable success to improve regeneration and/or functional recovery in animal models of neurologic diseases. Injected into the host, they migrate to the damaged areas and release cytokines and/or trophic factors, which are capable of altering the genetic program of the injured tissue cells. In this study, there was a search for genes with altered expression in a model of optic nerve crush and cell therapy. METHODS: Optic nerve crush was followed by an intravitreous injection of BMMCs or vehicle in adult rats. After 14 days, we obtained a transcriptome screening of the retinas using differential display and automatic sequencing, followed by q-PCR, Western blot, and immunohistochemistry of selected genes and proteins. RESULTS: Among the differentially displayed genes, transcription of the antiapoptotic Tax1-binding protein 1 (Tax1BP1) and Synaptotagmin IV (Syt IV), an immediate early gene, is increased in the treated group. Tax1BP1 expression is robust in the ganglion cell layer and is significantly increased by cell therapy. Syt IV is expressed by activated Müller cells and astrocytes in the retina and optic nerve, without changes in protein levels among the groups. CONCLUSIONS: Tax1BP1 and Syt IV transcription and/or expression are differently modulated by optic nerve crush and BMMC treatment, and might be related to neuronal damage and cell-therapy effects in the retina. The increased expression of Tax1BP1 in the treated eyes could be involved in the neuroprotective effects of BMMCs that were described previously by our group.


Asunto(s)
Células de la Médula Ósea/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Compresión Nerviosa , Traumatismos del Nervio Óptico/metabolismo , Sinaptotagminas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Western Blotting , Modelos Animales de Enfermedad , Proteínas de Neoplasias/metabolismo , Nervio Óptico/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Ratas , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo
16.
PLoS One ; 6(9): e25178, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949881

RESUMEN

Imbalance of the excitatory neurotransmitter glutamate and of the inhibitory neurotransmitter GABA is one of several causes of seizures. ATP has also been implicated in epilepsy. However, little is known about the mechanisms involved in the release of ATP from cells and the consequences of the altered ATP signaling during seizures. Pannexin1 (Panx1) is found in astrocytes and in neurons at high levels in the embryonic and young postnatal brain, declining in adulthood. Panx1 forms large-conductance voltage sensitive plasma membrane channels permeable to ATP that are also activated by elevated extracellular K(+) and following P2 receptor stimulation. Based on these properties, we hypothesized that Panx1 channels may contribute to seizures by increasing the levels of extracellular ATP. Using pharmacological tools and two transgenic mice deficient for Panx1 we show here that interference with Panx1 ameliorates the outcome and shortens the duration of kainic acid-induced status epilepticus. These data thus indicate that the activation of Panx1 in juvenile mouse hippocampi contributes to neuronal hyperactivity in seizures.


Asunto(s)
Conducta Animal/efectos de los fármacos , Conexinas/fisiología , Epilepsia/prevención & control , Proteínas del Tejido Nervioso/fisiología , Convulsiones/prevención & control , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Western Blotting , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Epilepsia/inducido químicamente , Epilepsia/metabolismo , Técnica del Anticuerpo Fluorescente , Hipocampo/citología , Hipocampo/metabolismo , Ácido Kaínico/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Potasio/metabolismo , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Estado Epiléptico/metabolismo
17.
Cell Transplant ; 20(3): 391-406, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20719093

RESUMEN

The central nervous system (CNS) of adult mammals generally does not regenerate, and many studies have attempted to identify factors that could increase neuroprotection and/or axonal outgrowth after CNS lesions. Using the optic nerve crush of rats as a model for CNS injury, we investigated the effect of intravitreal transplantation of syngeneic bone-marrow mononuclear cells (BMMCs) on the survival of retinal ganglion cells (RGC) and on the regeneration of optic axons. Control animals received intravitreal saline injections after lesion. Injections of BMMCs resulted in a 1.6-fold increase in the number of RGCs surviving 14 days after injury. The BMMC-treated animals also had increased numbers of axons, which grew up to 1.5 mm from the crush site, and also had reduced Müller glia activation. Analysis of mRNAs in all conditions revealed an increase in levels of fibroblast growth factor 2 (FGF-2) mRNA in treated animals 14 days after injury. To investigate whether the regenerated axons could reach the brain, we retrograde labeled the RGCs by injecting a lipophilic tracer into the superior colliculus. We also analyzed the expression of NGFI-A in the superficial layers of the superior colliculus as a possible marker of synaptic input from RGC axons. We found evidence that more RGCs were able to reach the brain after treatment and we showed that NGFI-A expression was higher in the treated animals 60 days after injury. These results demonstrate that transplant of BMMCs can increase neuroprotection and neuroregeneration after injury in a model of optic nerve crush, and these effects could be mediated by FGF-2.


Asunto(s)
Axones/fisiología , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Regeneración Nerviosa , Células Ganglionares de la Retina/citología , Animales , Supervivencia Celular , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Ratas , Proteínas Represoras/metabolismo , Colículos Superiores/metabolismo
18.
Epilepsia ; 51 Suppl 3: 71-5, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20618405

RESUMEN

Most of the gamma-aminobutyric acid (GABA)ergic interneurons in the cerebral cortex originate from restricted regions of the ventral telencephalon known as the caudal and medial ganglionic eminence (MGE) and from the preoptic area. It is well established that dysfunction of GABAergic interneurons can lead to epilepsy. During the last decade new approaches to prevent, reduce, or reverse the epileptic condition have been studied, including cell-based therapy from different sources. Recent studies have shown that transplanted neuronal precursor cells derived from MGE have the ability to migrate, differentiate into inhibitory GABAergic interneurons, and integrate into cortical and hippocampal networks, modifying the inhibitory tone in the host brain. Therefore, transplantation of neuronal precursors derived from MGE into the postnatal central nervous system (CNS) could modify the neuronal circuitry in neurologic diseases in which inhibitory synaptic function is altered, such as in epilepsy. Here, we evaluated the seizure susceptibility of mice transplanted with MGE-derived cells in the maximum electroconvulsive shock (MES) model and we review some data from different studies using GABAergic precursor or GABA-releasing cell grafts in animal models of seizure and epilepsy.


Asunto(s)
Células Madre Embrionarias/trasplante , Epilepsia/cirugía , Telencéfalo/citología , Animales , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Epilepsia/etiología , Epilepsia/fisiopatología , Hipocampo/fisiopatología , Interneuronas/fisiología , Ratones , Ratas , Receptores de GABA/fisiología , Sinapsis/fisiología , Telencéfalo/trasplante
19.
Eur J Neurosci ; 32(3): 311-21, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20646049

RESUMEN

During retinal development, cell proliferation and exit from the cell cycle must be precisely regulated to ensure the generation of the appropriate numbers and proportions of the various retinal cell types. Previously, we showed that pituitary adenylyl cyclase-activating polypeptide (PACAP) exerts a neuroprotective effect in the developing retina of rats, through the cAMP-cAMP-dependent protein kinase (protein kinase A) (PKA) pathway. Here, we show that PACAP also regulates the proliferation of retinal progenitor cells. PACAP, PACAP-specific receptor (PAC1), and the receptors activated by both PACAP and vasoactive intestinal peptide (VIP), VPAC1 and VPAC2, are expressed during embryonic and postnatal development of the rat retina. Treatment of retinal explants with PACAP38 reduced the incorporation of [(3)H]thymidine as well as the number of 5-bromo-2'-deoxyuridine-positive and cyclin D1-positive cells. Pharmacological experiments indicated that PACAP triggers this antiproliferative effect through the activation of both PAC1 and VPACs, and the cAMP-PKA pathway. In addition, PACAP receptor activation decreased both cyclin D1 mRNA and protein content. Altogether, the data support the hypothesis that PACAP is a cell-extrinsic regulator with multiple roles during retinal development, including the regulation of proliferation in a subpopulation of retinal progenitor cells.


Asunto(s)
Proliferación Celular , Ciclina D1/metabolismo , Regulación hacia Abajo , Neurogénesis/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Retina/metabolismo , Neuronas Retinianas/metabolismo , Análisis de Varianza , Animales , Western Blotting , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclina D1/genética , Inmunohistoquímica , Microscopía Confocal , Fosforilación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Péptido Intestinal Vasoactivo/genética , Receptores de Péptido Intestinal Vasoactivo/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre , Técnicas de Cultivo de Tejidos , Péptido Intestinal Vasoactivo/metabolismo
20.
PLoS Pathog ; 6(4): e1000870, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20442858

RESUMEN

The murine model of T. cruzi infection has provided compelling evidence that development of host resistance against intracellular protozoans critically depends on the activation of members of the Toll-like receptor (TLR) family via the MyD88 adaptor molecule. However, the possibility that TLR/MyD88 signaling pathways also control the induction of immunoprotective CD8+ T cell-mediated effector functions has not been investigated to date. We addressed this question by measuring the frequencies of IFN-gamma secreting CD8+ T cells specific for H-2K(b)-restricted immunodominant peptides as well as the in vivo Ag-specific cytotoxic response in infected animals that are deficient either in TLR2, TLR4, TLR9 or MyD88 signaling pathways. Strikingly, we found that T. cruzi-infected Tlr2(-/-), Tlr4(-/-), Tlr9(-/) (-) or Myd88(-/-) mice generated both specific cytotoxic responses and IFN-gamma secreting CD8+ T cells at levels comparable to WT mice, although the frequency of IFN-gamma+CD4+ cells was diminished in infected Myd88(-/-) mice. We also analyzed the efficiency of TLR4-driven immune responses against T. cruzi using TLR4-deficient mice on the C57BL genetic background (B6 and B10). Our studies demonstrated that TLR4 signaling is required for optimal production of IFN-gamma, TNF-alpha and nitric oxide (NO) in the spleen of infected animals and, as a consequence, Tlr4(-/-) mice display higher parasitemia levels. Collectively, our results indicate that TLR4, as well as previously shown for TLR2, TLR9 and MyD88, contributes to the innate immune response and, consequently, resistance in the acute phase of infection, although each of these pathways is not individually essential for the generation of class I-restricted responses against T. cruzi.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad de Chagas/inmunología , Inmunidad Innata , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Animales , Enfermedad de Chagas/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Trypanosoma cruzi/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA