Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G63-G73, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32538139

RESUMEN

Hyaluronic acid (HA), a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously demonstrated that both CD44 and TLR4, but predominately TLR4, mediated HA stimulation of Lgr5+ stem cell proliferation, crypt fission, and intestinal growth in postnatal mice. Here we address the questions of which cell type expresses the relevant TLR4 in driving intestinal growth and what are the downstream events from TLR4 activation. Studies were done in 14-day-old mice: wild type (WT), mice deficient in cyclooxygenase 2 (COX2), mice deficient in myeloid cell TLR4, and mice deficient in epithelial cell epidermal growth factor receptor (EGFR). Biological end points included crypt fission and Lgr5 cell proliferation. In WT mice, treatment with NS-398 (a COX2 inhibitor), clodronate (a macrophage-depleting agent), or tyrphostin (an EGFR inhibitor) resulted in 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with control mice. Mice deficient in COX2 or myeloid TLR4 or epithelial cell EGFR all had 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with WT mice. Administration of dimethyl PGE2, a stable PGE2 analog, increased crypt fission and Lgr5+ stem cell proliferation. Administration of dimethyl PGE2 reversed the effects of NS-398, clodronate, COX2 deficiency, and myeloid TLR4 deficiency but had no effect on mice treated with tyrphostin or mice deficient in epithelial cell EGFR. We conclude that, in postnatal mice, ~30% of intestinal growth as manifested by crypt fission and Lgr5+ stem cell proliferation is driven by a novel pathway: Extracellular HA binds TLR4 on pericryptal macrophages, inducing the production of PGE2 through COX2. PGE2 transactivates EGFR in Lgr5+ epithelial stem cells, resulting in Lgr5+ stem cell proliferation and crypt fission.NEW & NOTEWORTHY This study, in newborn mice, describes a novel molecular pathway regulating Lgr5+ epithelial stem cell proliferation and normal intestinal elongation, as assessed by crypt fission. In this pathway, endogenous extracellular hyaluronic acid binds to Toll-like receptor 4 on pericryptal macrophages releasing PGE2 which binds to epidermal growth factor receptor on Lgr5+ stem cells resulting in proliferation. Lgr5+ stem cell proliferation leads to crypt fission and intestinal elongation. The demonstration that normal growth requires microbial-independent Toll-like receptor activation is novel.


Asunto(s)
Dinoprostona/metabolismo , Receptores ErbB/efectos de los fármacos , Ácido Hialurónico/farmacología , Receptor Toll-Like 4/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Receptores ErbB/metabolismo , Ácido Hialurónico/antagonistas & inhibidores , Intestinos/efectos de los fármacos , Ratones Noqueados , Receptor Toll-Like 4/metabolismo , Activación Transcripcional/efectos de los fármacos
2.
Gut ; 68(6): 1003-1013, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29934438

RESUMEN

OBJECTIVE: Lactobacillus rhamnosus GG (LGG), a probiotic, given by gavage is radioprotective of the mouse intestine. LGG-induced radioprotection is toll-like receptor 2 (TLR2) and cyclooxygenase-2 (COX-2)-dependent and is associated with the migration of COX-2+mesenchymal stem cells (MSCs) from the lamina propria of the villus to the lamina propria near the crypt epithelial stem cells. Our goals were to define the mechanism of LGG radioprotection including identification of the TLR2 agonist, and the mechanism of the MSC migration and to determine the safety and efficacy of this approach in models relevant to clinical radiation therapy. DESIGN: Intestinal radioprotection was modelled in vitro with cell lines and enteroids as well as in vivo by assaying clinical outcomes and crypt survival. Fractionated abdominal and single dose radiation were used along with syngeneic CT26 colon tumour grafts to assess tumour radioprotection. RESULTS: LGG with a mutation in the processing of lipoteichoic acid (LTA), a TLR2 agonist, was not radioprotective, while LTA agonist and native LGG were. An agonist of CXCR4 blocked LGG-induced MSC migration and LGG-induced radioprotection. LGG given by gavage induced expression of CXCL12, a CXCR4 agonist, in pericryptal macrophages and depletion of macrophages by clodronate liposomes blocked LGG-induced MSC migration and radioprotection. LTA effectively protected the normal intestinal crypt, but not tumours in fractionated radiation regimens. CONCLUSIONS: LGG acts as a 'time-release capsule' releasing radioprotective LTA. LTA then primes the epithelial stem cell niche to protect epithelial stem cells by triggering a multicellular, adaptive immune signalling cascade involving macrophages and PGE2 secreting MSCs. TRIAL REGISTRATION NUMBER: NCT01790035; Pre-results.


Asunto(s)
Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus , Lipopolisacáridos/metabolismo , Probióticos/farmacología , Traumatismos por Radiación/prevención & control , Ácidos Teicoicos/metabolismo , Animales , Movimiento Celular/efectos de la radiación , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Activación de Macrófagos/efectos de la radiación , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Protectores contra Radiación , Valores de Referencia , Sensibilidad y Especificidad
3.
Muscle Nerve ; 57(2): 260-267, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28380694

RESUMEN

INTRODUCTION: Acellular nerve allografts (ANAs) yield less consistent favorable outcomes compared with autografts for long gap reconstructions. We evaluated whether a hybrid ANA can improve 6-cm gap reconstruction. METHODS: Rat sciatic nerve was transected and repaired with either 6-cm hybrid or control ANAs. Hybrid ANAs were generated using a 1-cm cellular isograft between 2.5-cm ANAs, whereas control ANAs had no isograft. Outcomes were assessed by graft gene and marker expression (n = 4; at 4 weeks) and motor recovery and nerve histology (n = 10; at 20 weeks). RESULTS: Hybrid ANAs modified graft gene and marker expression and promoted modest axon regeneration across the 6-cm defect compared with control ANA (P < 0.05), but yielded no muscle recovery. Control ANAs had no appreciable axon regeneration across the 6-cm defect. DISCUSSION: A hybrid ANA confers minimal motor recovery benefits for regeneration across long gaps. Clinically, the authors will continue to reconstruct long nerve gaps with autografts. Muscle Nerve 57: 260-267, 2018.


Asunto(s)
Regeneración Nerviosa/fisiología , Neuronas/trasplante , Envejecimiento , Aloinjertos , Animales , Axones/fisiología , Biomarcadores/análisis , Expresión Génica , Marcadores Genéticos , Masculino , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/inervación , Ratas , Ratas Endogámicas Lew , Recuperación de la Función , Nervio Ciático/lesiones , Nervio Ciático/cirugía , Estrés Fisiológico
4.
Biotechnol Bioeng ; 114(9): 2121-2130, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28481001

RESUMEN

Providing temporally regulated glial cell line-derived neurotrophic factor (GDNF) to injured nerve can promote robust axon regeneration. However, it is poorly understood why providing highly elevated levels of GDNF to nerve can lead to axon entrapment in the zone containing elevated GDNF. This limited understanding represents an obstacle to the translation of GDNF therapies to treat nerve injuries clinically. Here, we investigated how transgenic Schwann cells (SCs) overexpressing GDNF-IRES-DsRed impact nerve regeneration. Cultured primary SCs were transduced with lentiviruses (GDNF-overexpressing transgenic SCs), one of which provides the capability to express high levels of GDNF and regulate temporal GDNF expression. These SC groups were transplanted into acellular nerve allografts (ANAs) bridging a 14 mm rat sciatic nerve defect. GDNF-overexpressing transgenic SCs expressing GDNF for as little as 1 week decreased axon regeneration across ANAs and caused extensive extracellular matrix (ECM) remodeling. To determine whether additional gene expression changes beyond GDNF transgene expression occurred in GDNF-overexpressing transgenic SCs, microarray analysis of GDNF-overexpressing transgenic SCs compared to untreated SCs was performed. Microarray analysis revealed a set of common genes regulated in transgenic SC groups expressing high levels of GDNF compared to untreated SCs. A co-culture model of GDNF-overexpressing transgenic SCs with fibroblasts (FBs) revealed differential FB ECM-related gene expression compared to untreated SCs. These data suggest a component of axon entrapment is independent of GDNF's impact on axons. Biotechnol. Bioeng. 2017;114: 2121-2130. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Luminiscentes/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/fisiopatología , Traumatismos de los Nervios Periféricos/terapia , Nervio Ciático/lesiones , Nervio Ciático/trasplante , Aloinjertos , Animales , Sistema Libre de Células , Células Cultivadas , Regeneración Tisular Dirigida/métodos , Sitios Internos de Entrada al Ribosoma/fisiología , Masculino , Ratas , Ratas Endogámicas Lew , Células de Schwann/fisiología , Resultado del Tratamiento
5.
J Hand Surg Am ; 41(6): e113-21, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26880495

RESUMEN

PURPOSE: To evaluate the regenerative effect of the additional suture line when using either isografts (ISOs) or acellular nerve allografts (ANAs) placed end-to-end to span a short gap in a rat model. METHODS: Rat sciatic nerves were transected and repaired with 2-cm nerve grafts (ISO or ANA). The grafts were 2 cm in length or a 1-cm segment was connected end-to-end to a 1-cm segment to yield a 2-cm length. At 8 weeks, extensor digitorum longus (EDL) muscle force and mass were measured. Nerves were harvested for histomorphometry. In a separate parallel study, the nerves were harvested 2 weeks following graft implantation to assess gene expression changes. RESULTS: All grafts demonstrated regeneration across the 2-cm segment(s). The additional suture line did not result in statistical differences in the number of myelinated nerve fibers that reached the distal nerve. However, when the graft types were compared, there was a significant decrease in nerve fibers in the ANA groups. The EDL muscle mass was significantly greater by using nerve ISOs compared with ANAs, regardless of an additional suture line, but there were no statistical differences noted in EDL muscle force. Gene expression analysis did not differ owing to an additional suture line. CONCLUSIONS: Minimal axonal loss and no functional deficits were identified with an additional suture line in this rodent short nerve gap model. CLINICAL RELEVANCE: Placing nerve grafts in series is a viable option for treating short nerve gaps; however, the use of autografts remains preferable over the use of ANAs.


Asunto(s)
Aloinjertos , Axones/fisiología , Isoinjertos , Regeneración Nerviosa/fisiología , Nervio Ciático/cirugía , Nervio Ciático/trasplante , Animales , Modelos Animales de Enfermedad , Rechazo de Injerto , Supervivencia de Injerto , Masculino , Procedimientos Neuroquirúrgicos , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Sensibilidad y Especificidad , Factores de Tiempo , Trasplante Homólogo/métodos
6.
Gut ; 61(6): 829-38, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22027478

RESUMEN

BACKGROUND: The small intestinal epithelium is highly sensitive to radiation and is a major site of injury during radiation therapy and environmental overexposure. OBJECTIVE: To examine probiotic bacteria as potential radioprotective agents in the intestine. METHODS: 8-week-old C57BL/6 wild-type or knockout mice were administered probiotic by gavage for 3 days before 12 Gy whole body radiation. The intestine was evaluated for cell-positional apoptosis (6 h) and crypt survival (84 h). RESULTS: Gavage of 5×107 Lactobacillus rhamnosus GG (LGG) improved crypt survival about twofold (p<0.01); the effect was observed when administered before, but not after, radiation. Conditioned medium (CM) from LGG improved crypt survival (1.95-fold, p<0.01), and both LGG and LGG-CM reduced epithelial apoptosis particularly at the crypt base (33% to 18%, p<0.01). LGG was detected in the distal ileal contents after the gavage cycle, but did not lead to a detectable shift in bacterial family composition. The reduction in epithelial apoptosis and improved crypt survival offered by LGG was lost in MyD88⁻/⁻, TLR-2⁻/⁻ and cyclo-oxygenase-2⁻/⁻ (COX-2) mice but not TLR-4⁻/⁻ mice. LGG administration did not lead to increased jejunal COX-2 mRNA or prostaglandin E2 levels or a change in number of COX-2-expressing cells. However, a location shift was observed in constitutively COX-2-expressing cells of the lamina propria from the villi to a position near the crypt base (villi to crypt ratio 80:20 for control and 62:38 for LGG; p<0.001). Co-staining revealed these COX-2-expressing small intestinal lamina propria cells to be mesenchymal stem cells. CONCLUSIONS: LGG or its CM reduce radiation-induced epithelial injury and improve crypt survival. A TLR-2/MyD88 signalling mechanism leading to repositioning of constitutive COX-2-expressing mesenchymal stem cells to the crypt base is invoked.


Asunto(s)
Ciclooxigenasa 2/fisiología , Mucosa Intestinal/efectos de la radiación , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/uso terapéutico , Traumatismos Experimentales por Radiación/prevención & control , Receptor Toll-Like 2/fisiología , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/efectos de la radiación , Femenino , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Am J Physiol Gastrointest Liver Physiol ; 303(3): G377-88, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22556141

RESUMEN

Hyaluronic acid (HA), a component of the extracellular matrix, affects gastrointestinal epithelial proliferation in injury models, but its role in normal growth is unknown. We sought to determine the effects of exogenous HA on intestinal and colonic growth by intraperitoneal injection of HA twice a week into C57BL/6 mice from 3 to 8 wk of age. Similarly, to determine the effects of endogenous HA on intestinal and colonic growth, we administered PEP-1, a peptide that blocks the binding of HA to its receptors, on the same schedule. In mice treated with exogenous HA, villus height and crypt depth in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were increased. In mice treated with PEP-1, intestinal and colonic length were markedly decreased and crypt depth and villus height in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were decreased. Administration of HA was associated with increased levels of EGF (intestine) and IGF-I (colon), whereas administration of PEP-1 was associated with decreased levels of IGF-I (intestine) and epiregulin (colon). Exogenous HA increases intestinal and colonic epithelial proliferation, resulting in hyperplasia. Blocking the binding of endogenous HA to its receptors results in decreased intestinal and colonic length and a mucosal picture of hypoplasia, suggesting that endogenous HA contributes to the regulation of normal intestinal and colonic growth.


Asunto(s)
Colon/crecimiento & desarrollo , Ácido Hialurónico/farmacología , Intestinos/crecimiento & desarrollo , Animales , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Epirregulina , Células Epiteliales/citología , Glucuronosiltransferasa/biosíntesis , Receptores de Hialuranos/efectos de los fármacos , Hialuronano Sintasas , Hialuronoglucosaminidasa/biosíntesis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología , ARN Mensajero/metabolismo
8.
Plast Reconstr Surg ; 142(4): 952-961, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29994844

RESUMEN

BACKGROUND: Nerve grafting with an autograft is considered the gold standard. However, the functional outcomes of long (>3 cm) nerve autografting are often poor. The authors hypothesized that a factor contributing to these outcomes is the graft microenvironment, where long compared to short autografts support axon regeneration to different extents. METHODS: A rat sciatic nerve defect model was used to compare regeneration in short (2 cm) and long (6 cm) isografts. Axon regeneration and cell populations within grafts were assessed using histology, retrograde labeling of neurons regenerating axons, immunohistochemistry, quantitative reverse transcriptase polymerase chain reaction, and electron microscopy at 4 and/or 8 weeks. RESULTS: At 8 weeks, for distances of both 1 and 2 cm from the proximal coaptation (equivalent regenerative distance), long isografts had reduced numbers of regenerated fibers compared with short isografts. Similarly, the number of motoneurons regenerating axons was reduced in the presence of long isografts compared with short isografts. Considering the regenerative microenvironments between short and long isografts, cell densities and general populations within both short and long isografts were similar. However, long isografts had significantly greater expression of senescence markers, which included senescence-associated ß-galactosidase, p21, and p16, and distinct chromatin changes within Schwann cells. CONCLUSIONS: This study shows that axon regeneration is reduced in long compared with short isografts, where long isografts contained an environment with an increased accumulation of senescent markers. Although autografts are considered the gold standard for grafting, these results demonstrate that we must continue to strive for improvements in the autograft regenerative environment.


Asunto(s)
Regeneración Nerviosa/fisiología , Nervio Ciático/fisiología , Animales , Autoinjertos , Senescencia Celular/fisiología , Masculino , Distribución Aleatoria , Ratas Endogámicas Lew , Nervio Ciático/cirugía , Trasplante Autólogo/métodos
9.
Free Radic Biol Med ; 101: 85-92, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27693326

RESUMEN

Peripheral nerve injury evokes a complex cascade of chemical reactions including generation of molecular radicals. Conversely, the reactions within nerve induced by stress are difficult to directly detect or measure to establish causality. Monitoring these reactions in vivo would enable deeper understanding of the nature of the injury and healing processes. Here, we utilized near-infrared fluorescence molecular probes delivered via intra-neural injection technique to enable live, in vivo imaging of tissue response associated with nerve injury and stress. These initially quenched fluorescent probes featured specific sensitivity to hydroxyl radicals and become fluorescent upon encountering reactive oxygen species (ROS). Intraneurally delivered probes demonstrated rapid activation in injured rat sciatic nerve but minimal activation in normal, uninjured nerve. In addition, these probes reported activation within sciatic nerves of living rats after a stress caused by a pinprick stimulus to the abdomen. This imaging approach was more sensitive to detecting changes within nerves due to the induced stress than other techniques to evaluate cellular and molecular changes. Specifically, neither histological analysis of the sciatic nerves, nor the expression of pain and stress associated genes in dorsal root ganglia could provide statistically significant differences between the control and stressed groups. Overall, the results demonstrate a novel imaging approach to measure ROS in addition to the impact of ROS within nerve in live animals.


Asunto(s)
Carbocianinas/química , Colorantes Fluorescentes/química , Radical Hidroxilo/análisis , Imagen Óptica/métodos , Traumatismos de los Nervios Periféricos/diagnóstico , Animales , Carbocianinas/síntesis química , Colorantes Fluorescentes/síntesis química , Ganglios Espinales/lesiones , Ganglios Espinales/metabolismo , Radical Hidroxilo/metabolismo , Inyecciones Intralesiones , Masculino , Traumatismos de los Nervios Periféricos/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones , Nervio Ciático/metabolismo
10.
Tissue Eng Part A ; 22(13-14): 949-61, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27297909

RESUMEN

Acellular nerve allografts (ANAs) and other nerve constructs do not reliably facilitate axonal regeneration across long defects (>3 cm). Causes for this deficiency are poorly understood. In this study, we determined what cells are present within ANAs before axonal growth arrest in nerve constructs and if these cells express markers of cellular stress and senescence. Using the Thy1-GFP rat and serial imaging, we identified the time and location of axonal growth arrest in long (6 cm) ANAs. Axonal growth halted within long ANAs by 4 weeks, while axons successfully regenerated across short (3 cm) ANAs. Cellular populations and markers of senescence were determined using immunohistochemistry, histology, and senescence-associated ß-galactosidase staining. Both short and long ANAs were robustly repopulated with Schwann cells (SCs) and stromal cells by 2 weeks. Schwann cells (S100ß(+)) represented the majority of cells repopulating both ANAs. Overall, both ANAs demonstrated similar cellular populations with the exception of increased stromal cells (fibronectin(+)/S100ß(-)/CD68(-) cells) in long ANAs. Characterization of ANAs for markers of cellular senescence revealed that long ANAs accumulated much greater levels of senescence markers and a greater percentage of Schwann cells expressing the senescence marker p16 compared to short ANAs. To establish the impact of the long ANA environment on axonal regeneration, short ANAs (2 cm) that would normally support axonal regeneration were generated from long ANAs near the time of axonal growth arrest ("stressed" ANAs). These stressed ANAs contained mainly S100ß(+)/p16(+) cells and markedly reduced axonal regeneration. In additional experiments, removal of the distal portion (4 cm) of long ANAs near the time of axonal growth arrest and replacement with long isografts (4 cm) rescued axonal regeneration across the defect. Neuronal culture derived from nerve following axonal growth arrest in long ANAs revealed no deficits in axonal extension. Overall, this evidence demonstrates that long ANAs are repopulated with increased p16(+) Schwann cells and stromal cells compared to short ANAs, suggesting a role for these cells in poor axonal regeneration across nerve constructs.


Asunto(s)
Axones/metabolismo , Senescencia Celular , Regeneración Nerviosa , Células de Schwann/metabolismo , Andamios del Tejido/química , Animales , Axones/patología , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Células de Schwann/patología , Células del Estroma/metabolismo , Células del Estroma/patología
11.
Tissue Eng Part A ; 21(23-24): 2852-64, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26466815

RESUMEN

The use of growth factors, such as glial cell line-derived neurotrophic factor (GDNF), for the treatment of peripheral nerve injury has been useful in promoting axon survival and regeneration. Unfortunately, finding a method that delivers the appropriate spatial and temporal release profile to promote functional recovery has proven difficult. Some release methods result in burst release profiles too short to remain effective over the regeneration period; however, prolonged exposure to GDNF can result in axonal entrapment at the site of release. Thus, GDNF was delivered in both a spatially and temporally controlled manner using a two-phase system comprised of an affinity-based release system and conditional lentiviral GDNF overexpression from Schwann cells (SCs). Briefly, SCs were transduced with a tetracycline-inducible (Tet-On) GDNF overexpressing lentivirus before transplantation. Three-centimeter acellular nerve allografts (ANAs) were modified by injection of a GDNF-releasing fibrin scaffold under the epineurium and then used to bridge a 3 cm sciatic nerve defect. To encourage growth past the ANA, GDNF-SCs were transplanted into the distal nerve and doxycycline was administered for 4, 6, or 8 weeks to determine the optimal duration of GDNF expression in the distal nerve. Live imaging and histomorphometric analysis determined that 6 weeks of doxycycline treatment resulted in enhanced regeneration compared to 4 or 8 weeks. This enhanced regeneration resulted in increased gastrocnemius and tibialis anterior muscle mass for animals receiving doxycycline for 6 weeks. The results of this study demonstrate that strategies providing spatial and temporal control of delivery can improve axonal regeneration and functional muscle reinnervation.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Lentivirus , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/terapia , Células de Schwann , Transducción Genética , Animales , Modelos Animales de Enfermedad , Masculino , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/metabolismo , Ratas , Ratas Sprague-Dawley , Células de Schwann/metabolismo , Células de Schwann/patología , Células de Schwann/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA