Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Neurosci ; 38(3): 679-690, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29217688

RESUMEN

The MAPK/ERK pathway has a critical role in PNS development. It is required for Schwann cell (SC) differentiation and myelination; sustained embryonic MAPK/ERK activation in SCs enhances myelin growth overcoming signals that normally end myelination. Excess activation of this pathway can be maladaptive as in adulthood acute strong activation of MAPK/ERK has been shown to cause SC dedifferentiation and demyelination. We used a mouse model (including male and female animals) in which the gain-of-function MEK1DD allele produces sustained MAPK/ERK activation in adult SCs, and we determined the impact of such activation on nerve repair. In the uninjured nerve, MAPK/ERK activation neither impaired myelin nor reactivated myelination. However, in the injured nerve it was detrimental and resulted in delayed repair and functional recovery. In the early phase of injury, the rate of myelin clearance was faster. Four weeks following injury, when nerve repair is normally advanced, myelinated axons of MEK1DD mutants demonstrated higher rates of myelin decompaction, a reduced number of Cajal bands. and decreased internodal length. We noted the presence of abnormal Remak bundles with long SCs processes and reduced numbers of C-fibers/Remak bundle. Both the total number of regenerating axons and the intraepidermal nerve fiber density in the skin were reduced. Sustained activation of MAPK/ERK in adult SCs is therefore deleterious to successful nerve repair, emphasizing the differences in the signaling processes coordinating nerve development and repair. Our results also underline the key role of SCs in axon regeneration and successful target reinnervation.SIGNIFICANCE STATEMENT The MAPK/ERK pathway promotes developmental myelination and its sustained activation in SCs induced continuous myelin growth, compensating for the absence of essential myelination signals. However, the strength of activation is fundamental because acute strong induction of MAPK/ERK in adulthood induces demyelination. What has been unknown is the effect of a mild but sustained MAPK/ERK activation in SCs on nerve repair in adulthood. This promoted myelin clearance but led to abnormalities in nonmyelinating and myelinating SCs in the later phases of nerve repair, resulting in slowed axon regeneration, cutaneous reinnervation, and functional recovery. Our results emphasize the distinct role of the MAPK/ERK pathway in developmental myelination versus remyelination and the importance of signaling between SCs and axons for successful axon regeneration.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Células de Schwann/metabolismo , Animales , Activación Enzimática/fisiología , Femenino , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina , Compresión Nerviosa , Nervio Ciático/lesiones , Nervio Ciático/metabolismo
2.
Glia ; 67(10): 1990-2000, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31339187

RESUMEN

Membrane metallo-endopeptidase (MME), also known as neprilysin (NEP), has been of interest for its role in neurodegeneration and pain due to its ability to degrade ß-amyloid and substance-P, respectively. In addition to its role in the central nervous system, MME has been reported to be expressed in the peripheral system, specifically in the inner and outer border of myelinating fibers, in the Schmidt-Lantermann cleft and in the paranodes. Recently, mutations of this gene have been associated with Charcot-Marie-Tooth Type 2 (CMT2). Peripheral nerve morphometry in mice lacking MME previously showed minor abnormalities in aged animals in comparison to CMT2 patients. We found that MME expression was dysregulated after nerve injury in a Neuregulin-1 dependent fashion. We therefore explored the hypothesis that MME may have a role in remyelination. In the naïve state in adulthood we did not find any impairment in myelination in MME KO mice. After nerve injury the morphological outcome in MME KO mice was indistinguishable from WT littermates in terms of axon regeneration and remyelination. We did not find any difference in functional motor recovery. There was a significant difference in sensory function, with MME KO mice starting to recover response to mechanical stimuli earlier than WT. The epidermal reinnnervation, however, was unchanged and this altered sensitivity may relate to its known function in cleaving the peptide substance-P, known to sensitise nociceptors. In conclusion, although MME expression is dysregulated after nerve injury in a NRG1-dependent manner this gene is dispensable for axon regeneration and remyelination after injury.


Asunto(s)
Vaina de Mielina/enzimología , Neprilisina/metabolismo , Regeneración Nerviosa/fisiología , Nervio Ciático/enzimología , Nervio Ciático/lesiones , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Vaina de Mielina/patología , Neprilisina/genética , Neurregulina-1/genética , Neurregulina-1/metabolismo , Nocicepción/fisiología , Recuperación de la Función/fisiología , Nervio Ciático/patología
3.
Neurobiol Dis ; 95: 145-57, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27431093

RESUMEN

Charcot-Marie-Tooth 1A (CMT1A) is a demyelinating hereditary neuropathy for which pharmacological treatments are not yet available. An abnormally high intracellular Ca(2+) concentration was observed in Schwann cells (SC) from CMT1A rats, caused by the PMP22-mediated overexpression of the P2X7 purinoceptor. The purpose of this study was to investigate the tolerability and therapeutic potential of a pharmacological antagonist of the P2X7 receptor (A438079) in CMT1A. A438079 ameliorated in vitro myelination of organotypic DRG cultures from CMT1A rats. Furthermore, we performed an experimental therapeutic trial in PMP22 transgenic and in wild-type rats. A preliminary dose-escalation trial showed that 3mg/kg A438079 administered via intraperitoneal injection every 24h for four weeks was well tolerated by wild type and CMT1A rats. Affected rats treated with 3mg/kg A438079 revealed a significant improvement of the muscle strength, when compared to placebo controls. Importantly, histologic analysis revealed a significant increase of the total number of myelinated axons in tibial nerves. Moreover, a significant decrease of the hypermyelination of small caliber axons and a significant increase of the frequency and diameter of large caliber myelinated axons was highlighted. An improved distal motor latencies was recorded, whereas compound muscle action potentials (CMAP) remained unaltered. A438079 reduced the SC differentiation defect in CMT1A rats. These results show that pharmacological inhibition of the P2X7 receptor is well tolerated in CMT1A rats and represents a proof-of-principle that antagonizing this pathway may correct the molecular derangements and improve the clinical phenotype in the CMT1A neuropathy.


Asunto(s)
Axones/patología , Enfermedad de Charcot-Marie-Tooth/patología , Enfermedades Desmielinizantes/patología , Proteínas de la Mielina/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Células de Schwann/metabolismo , Animales , Animales Modificados Genéticamente , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Enfermedades Desmielinizantes/genética , Modelos Animales de Enfermedad , Proteínas de la Mielina/genética , Fenotipo , Ratas Sprague-Dawley , Ratas Transgénicas
4.
J Neurosci ; 34(44): 14506-16, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25355205

RESUMEN

The transient receptor potential A1 (TRPA1) channel is essential for vertebrate pain. Even though TRPA1 activation by ligands has been studied extensively, the molecular machinery regulating TRPA1 is only poorly understood. Using an unbiased proteomics-based approach we uncovered the physical association of Annexin A2 (AnxA2) with native TRPA1 in mouse sensory neurons. AnxA2 is enriched in a subpopulation of sensory neurons and coexpressed with TRPA1. Furthermore, we observe an increase of TRPA1 membrane levels in cultured sensory neurons from AnxA2-deficient mice. This is reflected by our calcium imaging experiments revealing higher responsiveness upon TRPA1 activation in AnxA2-deficient neurons. In vivo these findings are associated with enhanced nocifensive behaviors specifically in TRPA1-dependent paradigms of acute and inflammatory pain, while heat and mechanical sensitivity as well as TRPV1-mediated pain are preserved in AnxA2-deficient mice. Our results support a model whereby AnxA2 limits the availability of TRPA1 channels to regulate nociceptive signaling in vertebrates.


Asunto(s)
Anexina A2/metabolismo , Canales de Calcio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nocicepción/fisiología , Nociceptores/metabolismo , Dolor/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Conducta Animal/fisiología , Células HEK293 , Calor , Humanos , Ratones , Dimensión del Dolor , Estimulación Física , Ratas , Canal Catiónico TRPA1
5.
Proc Natl Acad Sci U S A ; 109(24): 9617-22, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22645329

RESUMEN

Erythropoietin (EPO) is a neuroprotective cytokine in models of ischemic and nervous system injury, where it reduces neuronal apoptosis and inflammatory cytokines and increases neurogenesis and angiogenesis. EPO also improves cognition in healthy volunteers and schizophrenic patients. We studied the effect of EPO administration on the gene-expression profile in the ischemic cortex of rats after cerebral ischemia at early time points (2 and 6 h). EPO treatment up-regulated genes already increased by ischemia. Hierarchical clustering and analysis of overrepresented functional categories identified genes implicated in synaptic plasticity-Arc, BDNF, Egr1, and Egr2, of which Egr2 was the most significantly regulated. Up-regulation of Arc, BDNF, Dusp5, Egr1, Egr2, Egr4, and Nr4a3 was confirmed by quantitative PCR. We investigated the up-regulation of Egr2/Krox20 further because of its role in neuronal plasticity. Its elevation by EPO was confirmed in an independent in vivo experiment of cerebral ischemia in rats. Using the rat neuroblastoma B104, we found that wild-type cells that do not express EPO receptor (EPOR) do not respond to EPO by inducing Egr2. However, EPOR-expressing B104 cells induce Egr2 early upon incubation with EPO, indicating that Egr2 induction is a direct effect of EPO and that EPOR mediates this effect. Because these changes occur in vivo before decreased inflammatory cytokines or neuronal apoptosis is evident, these findings provide a molecular mechanism for the neuroreparative effects of cytokines and suggest a mechanism of neuroprotection by which promotion of a plastic phenotype results in decreased inflammation and neuronal death.


Asunto(s)
Encéfalo/metabolismo , Eritropoyetina/fisiología , Perfilación de la Expresión Génica , Plasticidad Neuronal/genética , Accidente Cerebrovascular/genética , Animales , Reacción en Cadena de la Polimerasa , Ratas
6.
Mol Med ; 19: 223-9, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23821361

RESUMEN

Erythropoietin (EPO) has protective effects in neurodegenerative and neuroinflammatory diseases, including in animal models of multiple sclerosis, where EPO decreases disease severity. EPO also promotes neurogenesis and is protective in models of toxic demyelination. In this study, we asked whether EPO could promote neurorepair by also inducing remyelination. In addition, we investigated whether the effect of EPO could be mediated by the classical erythropoietic EPO receptor (EPOR), since it is still questioned if EPOR is functional in nonhematopoietic cells. Using CG4 cells, a line of rat oligodendrocyte precursor cells, we found that EPO increases the expression of myelin genes (myelin oligodendrocyte glycoprotein [MOG] and myelin basic protein [MBP]). EPO had no effect in wild-type CG4 cells, which do not express EPOR, whereas it increased MOG and MBP expression in cells engineered to overexpress EPOR (CG4-EPOR). This was reflected in a marked increase in MOG protein levels, as detected by Western blot. In these cells, EPO induced by 10-fold the early growth response gene 2 (Egr2), which is required for peripheral myelination. However, Egr2 silencing with a siRNA did not reverse the effect of EPO, indicating that EPO acts through other pathways. In conclusion, EPO induces the expression of myelin genes in oligodendrocytes and this effect requires the presence of EPOR. This study demonstrates that EPOR can mediate neuroreparative effects.


Asunto(s)
Eritropoyetina/farmacología , Vaina de Mielina/genética , Oligodendroglía/efectos de los fármacos , Receptores de Eritropoyetina/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Básica de Mielina/genética , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/metabolismo , Oligodendroglía/metabolismo , Ratas
7.
Neurobiol Dis ; 35(2): 270-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19464369

RESUMEN

Epothilones are a novel class of microtubule-targeting anticancer agents that are neurotoxic. In this study, we investigated the epothilone B toxic effect in vitro and we characterized in vivo the general and neurological side effects of epothilone B administration in Wistar and Fischer rats. The in vitro experiments made it possible to explore a wide concentration range (0.1 nM-1 muM) and evidenced a dose-dependent effect of epothilone B exposure on neuron neurite elongation. This dose-dependent neurotoxic effect was confirmed in both in vivo studies performed on two different rat strains at the neurophysiological, behavioral and pathological levels in the dose range 0.25-1.5 mg/kg iv weekly x 4 weeks and tubulin hyper-polymerization was demonstrated in sciatic nerve specimens. These are the first studies of the neurological effects of epothilone B and they can provide a basis for extending pre-clinical investigation to other members of the epothilone family.


Asunto(s)
Epotilonas/toxicidad , Ganglios Espinales/efectos de los fármacos , Neurotoxinas/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Epotilonas/administración & dosificación , Femenino , Ganglios Espinales/citología , Técnicas In Vitro , Vaina de Mielina/metabolismo , Conducción Nerviosa/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neurotoxinas/administración & dosificación , Umbral del Dolor/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Ratas Wistar , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Nervio Ciático/ultraestructura , Piel/efectos de los fármacos , Piel/inervación , Tubulina (Proteína)/metabolismo
8.
J Peripher Nerv Syst ; 14(1): 36-44, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19335538

RESUMEN

Peripheral neurotoxicity is a frequent complication limiting docetaxel chemotherapy in patients with cancer. We developed an experimental model that closely mimics the course of neuropathy in patients, aiming to investigate both the mechanisms of neurotoxicity at biochemical, functional and morphological levels and the potential neuroprotective role of neuroactive steroids. We demonstrated that treatment with dihydroprogesterone (DHP) or progesterone (P) counteracts docetaxel-induced neuropathy, preventing nerve conduction and thermal threshold changes, and degeneration of skin nerves in the foodpad. Neuroactive steroids also counteract the changes in gene expression of several myelin proteins and calcitonin gene-related peptide induced by docetaxel in sciatic nerve and lumbar spinal cord, respectively. Most nerve abnormalities observed during the treatment with docetaxel spontaneously recovered after drug withdrawal, similarly to what occurs in patients. However, results of midterm follow-up experiments indicated that animals cotreated with DHP or P have a faster recovery of the neuropathy compared with docetaxel-treated rats. Our study confirmed that neuroactive steroids exert a protective effect on peripheral nerves at different levels, suggesting that they might represent a new therapeutic frontier for patients with chemotherapy-induced neuropathy.


Asunto(s)
20-alfa-Dihidroprogesterona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Conducción Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/prevención & control , Progesterona/farmacología , Análisis de Varianza , Animales , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Docetaxel , Masculino , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/metabolismo , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Fibras Nerviosas/patología , Conducción Nerviosa/fisiología , Umbral del Dolor/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Ratas , Ratas Endogámicas F344 , Nervio Ciático/patología , Taxoides/farmacología
9.
J Cell Biol ; 218(7): 2370-2387, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31201266

RESUMEN

RalA and RalB are small GTPases that are involved in cell migration and membrane dynamics. We used transgenic mice in which one or both GTPases were genetically ablated to investigate the role of RalGTPases in the Schwann cell (SC) response to nerve injury and repair. RalGTPases were dispensable for SC function in the naive uninjured state. Ablation of both RalA and RalB (but not individually) in SCs resulted in impaired axon remyelination and target reinnervation following nerve injury, which resulted in slowed recovery of motor function. Ral GTPases were localized to the leading lamellipodia in SCs and were required for the formation and extension of both axial and radial processes of SCs. These effects were dependent on interaction with the exocyst complex and impacted on the rate of SC migration and myelination. Our results show that RalGTPases are required for efficient nerve repair by regulating SC process formation, migration, and myelination, therefore uncovering a novel role for these GTPases.


Asunto(s)
Regeneración Nerviosa/genética , Traumatismos de los Nervios Periféricos/genética , Proteínas de Unión al GTP ral/genética , Animales , Axones/metabolismo , Movimiento Celular/genética , Humanos , Ratones , Ratones Transgénicos , Vaina de Mielina/genética , Traumatismos de los Nervios Periféricos/patología , Células de Schwann/metabolismo , Células de Schwann/patología , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Nervio Ciático/patología
10.
Mol Med ; 14(11-12): 682-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18670620

RESUMEN

Erythropoietin (EPO) is of great interest as a therapy for many of the central nervous system (CNS) diseases and its administration is protective in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Endogenous EPO is induced by hypoxic/ischemic injury, but little is known about its expression in other CNS diseases. We report here that EPO expression in the spinal cord is induced in mouse models of chronic or relapsing-remitting EAE, and is prominently localized to motoneurons. We found a parallel increase of hypoxia-inducible transcription factor (HIF)-1 alpha, but not HIF-2 alpha, at the mRNA level, suggesting a possible role of non-hypoxic factors in EPO induction. EPO mRNA in the spinal cord was co-expressed with interferon (IFN)-gamma and tumor necrosis factor (TNF), and these cytokines inhibited EPO production in vitro in both neuronal and glial cells. Given the known inhibitory effect of EPO on neuroinflammation, our study indicates that EPO should be viewed as part of the inflammatory/anti-inflammatory network in MS.


Asunto(s)
Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Eritropoyetina/metabolismo , Eritropoyetina/fisiología , Animales , Línea Celular Tumoral , Eritropoyetina/genética , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunohistoquímica , Interferón gamma/farmacología , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
11.
Neuron ; 97(4): 806-822.e10, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29429934

RESUMEN

Human autoantibodies to contactin-associated protein-like 2 (CASPR2) are often associated with neuropathic pain, and CASPR2 mutations have been linked to autism spectrum disorders, in which sensory dysfunction is increasingly recognized. Human CASPR2 autoantibodies, when injected into mice, were peripherally restricted and resulted in mechanical pain-related hypersensitivity in the absence of neural injury. We therefore investigated the mechanism by which CASPR2 modulates nociceptive function. Mice lacking CASPR2 (Cntnap2-/-) demonstrated enhanced pain-related hypersensitivity to noxious mechanical stimuli, heat, and algogens. Both primary afferent excitability and subsequent nociceptive transmission within the dorsal horn were increased in Cntnap2-/- mice. Either immune or genetic-mediated ablation of CASPR2 enhanced the excitability of DRG neurons in a cell-autonomous fashion through regulation of Kv1 channel expression at the soma membrane. This is the first example of passive transfer of an autoimmune peripheral neuropathic pain disorder and demonstrates that CASPR2 has a key role in regulating cell-intrinsic dorsal root ganglion (DRG) neuron excitability.


Asunto(s)
Ganglios Espinales/fisiopatología , Inmunoglobulina G/administración & dosificación , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Dolor Nociceptivo/inmunología , Dolor Nociceptivo/fisiopatología , Células Receptoras Sensoriales/fisiología , Animales , Células Cultivadas , Femenino , Humanos , Inmunización Pasiva , Masculino , Mecanotransducción Celular , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Células del Asta Posterior/fisiología , Canales de Potasio de la Superfamilia Shaker/fisiología
12.
Methods Mol Biol ; 982: 163-73, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23456868

RESUMEN

Erythropoietin (EPO) has neuroprotective effects in many models of damage and disease of the nervous system where neuroinflammation plays a substantial role, including experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS). Since the first pioneering studies, in which EPO was shown to protect rats with acute EAE mainly by inhibiting inflammation, many other studies have pointed out other mechanisms of protection, including oligodendrogenesis and inhibition of axonal damage.Here we review the preclinical studies in which EPO has shown therapeutic efficacy in several models of EAE in mice and rats. Moreover, we report in detail the protocol to administer EPO to mice with myelin oligodendrocyte glycoprotein (MOG)-induced chronic progressive EAE, and a representative result. In this model, EPO inihibits the clinical score of the disease when administered according to a preventive but also to a therapeutic schedule, and therefore at disease onset, suggesting that it might not only inhibit inflammation but also actively stimulate repair.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Eritropoyetina/metabolismo , Esclerosis Múltiple/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Glicoproteína Mielina-Oligodendrócito/metabolismo , Ratas
13.
Neurotox Res ; 18(2): 151-60, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19876698

RESUMEN

Taxanes, including docetaxel (DOCE), are severely neurotoxic, causing disabling peripheral neuropathy. Co-treatment with neuroprotective agents has been proposed to prevent or reverse this. Besides its hemopoietic effects, erythropoietin (EPO) has neuroprotective and neurotrophic properties and when administered systemically it has a wide range of neuroprotective action in animal models of nervous system damage, including cisplatin-induced peripheral neurotoxicity. The present study investigated the effects of EPO on chemotherapy-induced peripheral neurotoxicity (CINP) by DOCE in vivo and whether it interfered with tumor growth or antitumor activity. Female Fischer rats bearing 13762 mammary carcinoma were randomly divided into four groups: untreated, treated with EPO, DOCE, or DOCE + EPO. DOCE was given once a week (5 mg/kg, i.v.) and EPO three times a week (50 microg/kg i.p.), for 4 weeks. Three other groups of rats without tumors were left untreated or given DOCE or DOCE + EPO. The rats were observed for 4 weeks after treatment. CINP and neuroprotection were evaluated by measuring nociception, electrophysiological, and biochemical parameters. EPO protected against CINP, and tumor growth in EPO-treated rats was the same as in controls. EPO significantly improved the thermal threshold, tail nerve conduction velocity, and intra-epidermal nerve fiber density. These benefits lasted through the follow-up period and EPO speeded-up spontaneous recovery after treatment withdrawal. EPO did not impair DOCE antitumor activity. Since CINP induced by DOCE reproduces the clinical utility of taxane in humans, the findings reported might provide a basis for investigating EPO as a neuroprotective agent in patients receiving therapy with DOCE.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/efectos adversos , Eritropoyetina/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/prevención & control , Taxoides/efectos adversos , Animales , Docetaxel , Interacciones Farmacológicas , Eritropoyetina/administración & dosificación , Femenino , Humanos , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Endogámicas F344 , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda