Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(12)2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37373396

RESUMEN

The spinal cord has a poor ability to regenerate after an injury, which may be due to cell loss, cyst formation, inflammation, and scarring. A promising approach to treating a spinal cord injury (SCI) is the use of biomaterials. We have developed a novel hydrogel scaffold fabricated from oligo(poly(ethylene glycol) fumarate) (OPF) as a 0.08 mm thick sheet containing polymer ridges and a cell-attractive surface on the other side. When the cells are cultured on OPF via chemical patterning, the cells attach, align, and deposit ECM along the direction of the pattern. Animals implanted with the rolled scaffold sheets had greater hindlimb recovery compared to that of the multichannel scaffold control, which is likely due to the greater number of axons growing across it. The immune cell number (microglia or hemopoietic cells: 50-120 cells/mm2 in all conditions), scarring (5-10% in all conditions), and ECM deposits (Laminin or Fibronectin: approximately 10-20% in all conditions) were equal in all conditions. Overall, the results suggest that the scaffold sheets promote axon outgrowth that can be guided across the scaffold, thereby promoting hindlimb recovery. This study provides a hydrogel scaffold construct that can be used in vitro for cell characterization or in vivo for future neuroprosthetics, devices, or cell and ECM delivery.


Asunto(s)
Organofosfonatos , Traumatismos de la Médula Espinal , Ratas , Animales , Hidrogeles/química , Organofosfonatos/metabolismo , Cicatriz/patología , Ratas Sprague-Dawley , Regeneración Nerviosa , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/metabolismo , Axones/patología , Andamios del Tejido/química
2.
J Neuropathol Exp Neurol ; 82(7): 595-610, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37244652

RESUMEN

Machine learning is a powerful tool that is increasingly being used in many research areas, including neuroscience. The recent development of new algorithms and network architectures, especially in the field of deep learning, has made machine learning models more reliable and accurate and useful for the biomedical research sector. By minimizing the effort necessary to extract valuable features from datasets, they can be used to find trends in data automatically and make predictions about future data, thereby improving the reproducibility and efficiency of research. One application is the automatic evaluation of micrograph images, which is of great value in neuroscience research. While the development of novel models has enabled numerous new research applications, the barrier to use these new algorithms has also decreased by the integration of deep learning models into known applications such as microscopy image viewers. For researchers unfamiliar with machine learning algorithms, the steep learning curve can hinder the successful implementation of these methods into their workflows. This review explores the use of machine learning in neuroscience, including its potential applications and limitations, and provides some guidance on how to select a fitting framework to use in real-life research projects.


Asunto(s)
Microscopía , Motivación , Reproducibilidad de los Resultados , Algoritmos , Aprendizaje Automático
3.
Int J Neurosci ; : 1-23, 2022 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-36371721

RESUMEN

Background: Activated microglia release harmful substances to retinal ganglion cells (RGCs), but may also benefit by removing cellular debris and secreting neurotrophic factors. These paradoxical roles remain controversial because the nature and time-course of the injury that defines their role is unknown. The aim of this study was to determine if pharmacological manipulation of microglia to acquire a pro-inflammatory or pro-survival phenotype will exacerbate or enhance neuronal survival after injury.Material and methods: Treated HAP I (highly aggressively proliferating immortalized) microglia were injected into the vitreous or tail vein (T V) of female Sprague-Dawley rats. Retinas were examined at 4-14 days following optic nerve crush (ONC) and the number of surviving RGCs was determined.Results: Injection of untreated HAP I cells resulted in the greater loss of RGCs early after ONC when injected into the vitreous and later after ONC when injected into the T V. LP S activated HAP I cells injected into the vitreous resulted in greater RGC loss with and without injury. When injected into the T V with ONC there was no loss of RGCs 4 days after ONC but greater loss afterwards. Minocycline treated HAP I cells injected into the vitreous resulted in greater RGC survival than untreated HAP I cells. However, when injected into the T V with ONC there was greater loss of RGCs. These results suggest that optic nerve signals attract extrinsic microglia to the retina, resulting in a proinflammatory response.Conclusion: Neuroprotection or cytotoxicity of microglia depends on the type of activation, time course of the injury, and if they act on the axon or cell body.


We show here that neuroprotection is not solely determined by the microglial activation state but factors such as the environment and time-course of the injury.Culture microglia can be treated in vitro and then injected in vivo.The cells migrate to the site of injury, cell body of retinal ganglion cells if in the vitreous or to the optic nerve if injected in the tail vein.Retinal ganglion cell death is dependent on the location the microglia act, time-course of injury, and activation state.Proinflammatory microglia can be neuroprotective early in the injury when the primary site of action is on the axons whereas hypoactivated microglia are neuroprotective early in injury when they act on the soma. Later in the injury, both become detrimental.

4.
Neuroscientist ; 28(2): 163-179, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33089762

RESUMEN

Evidence from preclinical and clinical research suggest that neuromodulation technologies can facilitate the sublesional spinal networks, isolated from supraspinal commands after spinal cord injury (SCI), by reestablishing the levels of excitability and enabling descending motor signals via residual connections. Herein, we evaluate available evidence that sublesional and supralesional spinal circuits could form a translesional spinal network after SCI. We further discuss evidence of translesional network reorganization after SCI in the presence of sensory inputs during motor training. In this review, we evaluate potential mechanisms that underlie translesional circuitry reorganization during neuromodulation and rehabilitation in order to enable motor functions after SCI. We discuss the potential of neuromodulation technologies to engage various components that comprise the translesional network, their functional recovery after SCI, and the implications of the concept of translesional network in development of future neuromodulation, rehabilitation, and neuroprosthetics technologies.


Asunto(s)
Traumatismos de la Médula Espinal , Médula Espinal , Humanos , Recuperación de la Función
5.
NPJ Regen Med ; 6(1): 66, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34671050

RESUMEN

Here, we report the effect of newly regenerated axons via scaffolds on reorganization of spinal circuitry and restoration of motor functions with epidural electrical stimulation (EES). Motor recovery was evaluated for 7 weeks after spinal transection and following implantation with scaffolds seeded with neurotrophin producing Schwann cell and with rapamycin microspheres. Combined treatment with scaffolds and EES-enabled stepping led to functional improvement compared to groups with scaffold or EES, although, the number of axons across scaffolds was not different between groups. Re-transection through the scaffold at week 6 reduced EES-enabled stepping, still demonstrating better performance compared to the other groups. Greater synaptic reorganization in the presence of regenerated axons was found in group with combined therapy. These findings suggest that newly regenerated axons through cell-containing scaffolds with EES-enabled motor training reorganize the sub-lesional circuitry improving motor recovery, demonstrating that neuroregenerative and neuromodulatory therapies cumulatively enhancing motor function after complete SCI.

6.
Biomedicines ; 9(5)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925613

RESUMEN

Spinal cord injury (SCI) results in cell death, demyelination, and axonal loss. The spinal cord has a limited ability to regenerate, and current clinical therapies for SCI are not effective in helping promote neurologic recovery. We have developed a novel scaffold biomaterial that is fabricated from the biodegradable hydrogel oligo(poly(ethylene glycol)fumarate) (OPF). We have previously shown that positively charged OPF scaffolds (OPF+) in an open spaced, multichannel design can be loaded with Schwann cells to support axonal generation and functional recovery following SCI. We have now developed a hybrid OPF+ biomaterial that increases the surface area available for cell attachment and that contains an aligned microarchitecture and extracellular matrix (ECM) proteins to better support axonal regeneration. OPF+ was fabricated as 0.08 mm thick sheets containing 100 µm high polymer ridges that self-assemble into a spiral shape when hydrated. Laminin, fibronectin, or collagen I coating promoted neuron attachment and axonal outgrowth on the scaffold surface. In addition, the ridges aligned axons in a longitudinal bipolar orientation. Decreasing the space between the ridges increased the number of cells and neurites aligned in the direction of the ridge. Schwann cells seeded on laminin coated OPF+ sheets aligned along the ridges over a 6-day period and could myelinate dorsal root ganglion neurons over 4 weeks. This novel scaffold design, with closer spaced ridges and Schwann cells, is a novel biomaterial construct to promote regeneration after SCI.

7.
Tissue Eng Part A ; 27(11-12): 648-664, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33764164

RESUMEN

Positively charged oligo(poly(ethylene glycol) fumarate) (OPF+) hydrogel scaffolds, implanted into a complete transection spinal cord injury (SCI), facilitate a permissive regenerative environment and provide a platform for controlled observation of repair mechanisms. Axonal regeneration after SCI is critically dependent upon nutrients and oxygen from a newly formed blood supply. Our objective was to investigate fundamental characteristics of revascularization in association with the ingrowth of axons into hydrogel scaffolds, thereby defining spatial relationships between axons and the neovasculature. A novel combination of stereologic estimates and precision image analysis techniques quantitate neurovascular regeneration in rats. Multichannel hydrogel scaffolds containing Matrigel-only (MG), Schwann cells (SCs), or SCs with rapamycin-eluting poly(lactic co-glycolic acid) microspheres (RAPA) were implanted for 6 weeks following complete spinal cord transection. Image analysis of 72 scaffold channels identified a total of 2494 myelinated and 4173 unmyelinated axons at 10 µm circumferential intervals centered around 708 individual blood vessel profiles. Blood vessel number, density, volume, diameter, intervessel distances, total vessel surface and cross-sectional areas, and radial diffusion distances were compared. Axon number and density, blood vessel surface area, and vessel cross-sectional areas in the SC group exceeded that in the MG and RAPA groups. Individual axons were concentrated within a concentric radius of 200-250 µm from blood vessel walls, in Gaussian distributions, which identified a peak axonal number (Mean Peak Amplitude) corresponding to defined distances (Mean Peak Distance) from each vessel, the highest concentrations of axons were relatively excluded from a 25-30 µm zone immediately adjacent to the vessel, and from vessel distances >150 µm. Higher axonal densities correlated with smaller vessel cross-sectional areas. A statistical spatial algorithm was used to generate cumulative distribution F- and G-functions of axonal distribution in the reference channel space. Axons located around blood vessels were definitively organized as clusters and were not randomly distributed. A scoring system stratifies 5 direct measurements and 12 derivative parameters influencing regeneration outcomes. By providing methods to quantify the axonal-vessel relationships, these results may refine spinal cord tissue engineering strategies to optimize the regeneration of complete neurovascular bundles in their relevant spatial relationships after SCI. Impact statement Vascular disruption and impaired neovascularization contribute critically to the poor regenerative capacity of the spinal cord after injury. In this study, hydrogel scaffolds provide a detailed model system to investigate the regeneration of spinal cord axons as they directly associate with individual blood vessels, using novel methods to define their spatial relationships and the physiologic implications of that organization. These results refine future tissue engineering strategies for spinal cord repair to optimize the re-development of complete neurovascular bundles in their relevant spatial architectures.


Asunto(s)
Hidrogeles , Traumatismos de la Médula Espinal , Animales , Axones , Regeneración Nerviosa , Ratas , Células de Schwann , Médula Espinal , Andamios del Tejido
8.
Mayo Clin Proc ; 96(6): 1426-1437, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33678411

RESUMEN

OBJECTIVE: To provide precise description of the dorsal and ventral roots orientation along with the main spinal cord anatomical measurements and their segment-specific variations. PATIENTS AND METHODS: We collected and analyzed the measurements of the spines, spinal cords, and dorsal and ventral roots (C2-L5) of nine adult cadavers (five males and four females). RESULTS: This study for the first time provides analysis of the dorsal and ventral roots orientation along with spinal cord anatomical measurements and their segment-specific distribution. The results of this study showed less variability in rostral root angles compared with the caudal. Dorsal and ventral rootlets were oriented mostly perpendicular to the spinal cord at the cervical level and had more parallel orientation to the spinal cord at the thoracic and lumbar segments. The number of rootlets per root was greatest at dorsal cervical and lumbar segments. Spinal cord transverse diameter and width of the dorsal columns were largest at cervical segments. The strongest correlation between the spinal cord and vertebrae structures was found between the length of intervertebral foramen to rostral rootlet distance and vertebral bone length. CONCLUSION: These results demonstrate consistent variation in spinal cord anatomical features across all tested subjects. The results of this study can be used to locate spinal roots and main spinal cord landmarks based on bone marks on computed tomography or X-rays. These results could improve stereotactic surgical procedures and electrode positioning for neuromodulation procedures.


Asunto(s)
Médula Espinal/anatomía & histología , Raíces Nerviosas Espinales/anatomía & histología , Anciano de 80 o más Años , Puntos Anatómicos de Referencia/anatomía & histología , Vértebras Cervicales , Femenino , Humanos , Vértebras Lumbares , Masculino , Vértebras Torácicas
9.
Spinal Cord ; 59(3): 319-327, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33139846

RESUMEN

STUDY DESIGN: Animal study. OBJECTIVES: Umbilical cord-derived mesenchymal stem cells (UC-MSCs) have recently been shown to hold great therapeutic potential for spinal cord injury (SCI). However, majority of the studies have been done using human cells transplanted into the rat with immunosuppression; this may not represent the outcomes that occur in humans. Herein, we present the therapeutic effect of using rat UC-MSCs (rUC-MSC) without immunosuppression in a rat model of SCI. SETTING: Mayo Clinic, Rochester, MN, USA. METHODS: Twelve female rats were randomly divided into two groups, control, and rUC-MSC group, and then subjected to a T9 moderate contusion SCI. Next, 2 × 106 rUC-MSCs or ringer-lactate solution were injected through the tail vein at 7 days post injury. Rats were assessed for 14 weeks by an open-field Basso, Beattie, and Bresnahan (BBB) motor score as well as postmortem quantification of axonal sparing/regeneration, cavity volume, and glial scar. RESULTS: Animals treated with rUC-MSCs were found to have early and sustained motor improvement (BBB score of 14.6 ± 1.9 compared to 10.1 ± 1.7 in the control group) at 14 weeks post injury (mean difference: 4.55, 95% CI: 2.04 to 7.06; p value < 0.001). Total cavity volume in the injury epicenter was significantly reduced in the rUC-MSC group; control: 33.0% ± 2.1, rUC-MSC: 25.3% ± 3.8 (mean difference: -7.7% (95% CI: -12.3 to -2.98); p value < 0.05). In addition, spinal cords from rats treated with rUC-MSCs were found to have a significantly greater number of myelinated axons, decreased astrogliosis, and reduced glial scar formation compared to control rats. CONCLUSIONS: Our study indicates that intravenous injection of allogenic UC-MSCs without immunosuppression exert beneficial effects in subacute SCI and thus could be a useful therapy to improve the functional capacity among patients with SCI.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Traumatismos de la Médula Espinal , Animales , Femenino , Humanos , Ratas , Recuperación de la Función , Médula Espinal , Traumatismos de la Médula Espinal/terapia , Cordón Umbilical
10.
PLoS One ; 15(10): e0241285, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33125407

RESUMEN

Adaptive immunity is critical for controlling infections, which are a leading cause of morbidity and mortality in patients with spinal cord injury (SCI). In rats and mice, compromised peripheral adaptive immune responses, as shown by splenic atrophy and lowered frequencies of peripheral lymphocytes, were shown to result from high-level thoracic SCI. However, whether cervical SCI, which is the most common level of SCI in humans, impairs adaptive immunity remains largely unknown. In the present study, we induced cervical SCI in rats at the C7/T1 level by clip compression and looked at changes in peripheral adaptive immunity at 2-, 10- and 20-weeks post-injury. Specifically, we quantified changes in the frequencies of T- and B- lymphocytes in the blood and the mandibular and deep cervical lymph nodes, which drain the cervical spinal cord. We also assessed changes in serum IgG and IgM immunoglobulin levels, as well as spleen size. We found a significant decline in circulating T- and B- cell frequencies at 10 weeks post-SCI, which returned to normal at 20 weeks after injury. We found no effect of cervical SCI on T- and B- cell frequencies in the draining lymph nodes. Moreover, cervical SCI had no effect on net spleen size, although injured rats had a higher spleen/body weight ratio than sham controls at all time points of the study. Lastly, IgG and IgM immunoglobulin declined at 2 weeks, followed by a significant increase in IgM levels at 10 weeks of injury. These data indicate that cervical SCI causes a significant imbalance in circulating lymphocytes and immunoglobulin levels at 2 and 10 weeks. As we discuss in this article, these findings are largely in line with clinical observations, and we anticipate that this study will fuel more research on the effect of adaptive immunity on SCI recovery.


Asunto(s)
Inmunidad Adaptativa/fisiología , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/patología , Animales , Linfocitos B/metabolismo , Médula Cervical/inmunología , Médula Cervical/patología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Ganglios Linfáticos/inmunología , Ratones , Ratas , Ratas Wistar , Linfocitos T/metabolismo
11.
Expert Opin Biol Ther ; 17(5): 529-541, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28306359

RESUMEN

INTRODUCTION: Spinal cord injury (SCI) is a devastating condition, where regenerative failure and cell loss lead to paralysis. The heterogeneous and time-sensitive pathophysiology has made it difficult to target tissue repair. Despite many medical advances, there are no effective regenerative therapies. As stem cells offer multi-targeted and environmentally responsive benefits, cell therapy is a promising treatment approach. Areas covered: This review highlights the cell therapies being investigated for SCI, including Schwann cells, olfactory ensheathing cells, mensenchymal stem/stromal cells, neural precursors, oligodendrocyte progenitors, embryonic stem cells, and induced pluripotent stem cells. Through mechanisms of cell replacement, scaffolding, trophic support and immune modulation, each approach targets unique features of SCI pathology. However, as the injury is multifaceted, it is increasingly recognized that a combinatorial approach will be necessary to treat SCI. Expert opinion: Most preclinical studies, and an increasing number of clinical trials, are finding that single cell therapies have only modest benefits after SCI. These considerations, alongside issues of therapy cost-effectiveness, need to be addressed at the bench. In addition to exploring combinatorial strategies, researchers should consider cell reproducibility and storage parameters when designing animal experiments. Equally important, clinical trials must follow strict regulatory guidelines that will enable transparency of results.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Traumatismos de la Médula Espinal/diagnóstico , Traumatismos de la Médula Espinal/terapia , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/trasplante , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Regeneración/fisiología , Reproducibilidad de los Resultados , Médula Espinal/patología
12.
J Neurotrauma ; 34(6): 1209-1226, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27775474

RESUMEN

The immune system plays a critical and complex role in the pathobiology of spinal cord injury (SCI), exerting both beneficial and detrimental effects. Increasing evidence suggests that there are injury level-dependent differences in the immune response to SCI. Patients with traumatic SCI have elevated levels of circulating autoantibodies against components of the central nervous system, but the role of these antibodies in SCI outcomes remains unknown. In rodent models of mid-thoracic SCI, antibody-mediated autoimmunity appears to be detrimental to recovery. However, whether autoantibodies against the spinal cord are generated following cervical SCI (cSCI), the most common level of injury in humans, remains undetermined. To address this knowledge gap, we investigated the antibody responses following cSCI in a rat model of injury. We found increased immunoglobulin G (IgG) and IgM antibodies in the spinal cord in the subacute phase of injury (2 weeks), but not in more chronic phases (10 and 20 weeks). At 2 weeks post-cSCI, antibodies were detected at the injury epicenter and co-localized with the astroglial scar and neurons of the ventral horn. These increased levels of antibodies corresponded with enhanced activation of immune responses in the spleen. Higher counts of antibody-secreting cells were observed in the spleen of injured rats. Further, increased levels of secreted IgG antibodies and enhanced proliferation of T-cells in splenocyte cultures from injured rats were found. These findings suggest the potential development of autoantibody responses following cSCI in the rat. The impact of the post-traumatic antibody responses on functional outcomes of cSCI is a critical topic that requires further investigation.


Asunto(s)
Autoanticuerpos/inmunología , Médula Cervical/lesiones , Traumatismos de la Médula Espinal/inmunología , Animales , Células Productoras de Anticuerpos/inmunología , Astrocitos/inmunología , Modelos Animales de Enfermedad , Femenino , Ratas , Ratas Wistar , Bazo/inmunología
13.
Exp Eye Res ; 145: 363-372, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26601926

RESUMEN

Parvalbumin (PARV) is a Ca(2+)-binding protein that may offer resistance to cell death as it primarily functions to maintain Ca(2+) homeostasis. The purpose of this study was to investigate whether PARV expressing retinal ganglion cells (RGCs) would be more resistant to cell death than RGCs that do not express PARV. RGCs in Sprague-Dawley rats were retrogradely labeled with Fluorogold (FG). After 2-28 days following an optic nerve crush (ONC) injury immunohistochemistry was performed on the sections using antibodies against PARV and markers of RGCs. The proportion of retinal ganglion cell layer cells labeled with PARV colocalized with FG or Brn3a and labeled only with PARV (displaced amacrine cells; dACs) were analyzed. PARV staining intensity was measured in ACs, dACs, and RGCs. Double labeling studies revealed that 49% of RGCs and 22% of dACs expressed PARV. There was an immediate reduction in RGC PARV staining after ONC but the overall rate of cell death after 28 days was similar in PARV and non-PARV expressing RGCs. There was no change in PARV AC or dAC number or staining intensity. Although this study suggests that there is no selective survival of the subpopulation of RGCs that contain PARV, there is down-regulation of PARV expression by these RGCs. This suggests that down-regulation of PARV may contribute to RGC death due to a compromised Ca(2+) buffering capacity. Maintaining PARV expression after injury could be an important neuroprotective strategy to improve RGC survival after injury.


Asunto(s)
Células Amacrinas/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Parvalbúminas/metabolismo , Células Ganglionares de la Retina/metabolismo , Análisis de Varianza , Animales , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Inmunohistoquímica , Compresión Nerviosa , Ratas , Ratas Sprague-Dawley
14.
Prog Brain Res ; 218: 15-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25890131

RESUMEN

One of the big challenges in neuroscience that remains to be understood is why the central nervous system is not able to regenerate to the extent that the peripheral nervous system does. This is especially problematic after traumatic injuries, like spinal cord injury (SCI), since the lack of regeneration leads to lifelong deficits and paralysis. Treatment of SCI has improved during the last several decades due to standardized protocols for emergency medical response teams and improved medical, surgical, and rehabilitative treatments. However, SCI continues to result in profound impairments for the individual. There are many processes that lead to the pathophysiology of SCI, such as ischemia, vascular disruption, neuroinflammation, oxidative stress, excitotoxicity, demyelination, and cell death. Current treatments include surgical decompression, hemodynamic control, and methylprednisolone. However, these early treatments are associated with modest functional recovery. Some treatments currently being investigated for use in SCI target neuroprotective (riluzole, minocycline, G-CSF, FGF-2, and polyethylene glycol) or neuroregenerative (chondroitinase ABC, self-assembling peptides, and rho inhibition) strategies, while many cell therapies (embryonic stem cells, neural stem cells, induced pluripotent stem cells, mesenchymal stromal cells, Schwann cells, olfactory ensheathing cells, and macrophages) have also shown promise. However, since SCI has multiple factors that determine the progress of the injury, a combinatorial therapeutic approach will most likely be required for the most effective treatment of SCI.


Asunto(s)
Neuroprotección , Regeneración/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Investigación Biomédica Traslacional , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/epidemiología
15.
Invest Ophthalmol Vis Sci ; 55(3): 1919-29, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24526440

RESUMEN

PURPOSE: BM88 is a cell-cycle exit and neuronal differentiation protein that has been used as a marker of surviving retinal ganglion cells (RGCs) after optic nerve injury. Thy1.1 has also been used as a marker for RGC loss, but after optic nerve crush (ONC) a decrease in Thy1.1 expression precedes the loss of RGCs. The purpose of this study was to determine if BM88 expression was correlated with RGC loss after ONC and optic nerve transection (ONT) injuries. METHODS: Rats were injected with Fluorogold (FG) into the superior colliculus to label RGCs and received ONC or ONT 7 days later. Eyes were collected 2 to 28 days after injury. Retinas were labeled with BM88 and intensity of the BM88 cell labeling was measured. RESULTS: In control retinas, 98.9% of RGCs were immunoreactive (-IR) for BM88. There was a significant downregulation of BM88 by 52% to 80% of RGCs 7 days after ONC or ONT. The staining intensity of the remaining labeled cells was reduced to 41% to 51% of the control after 28 days of optic nerve injury. However, early in the injury there was a significant increase in the staining intensity of BM88. CONCLUSIONS: Nearly all BM88-IR RGCs colocalized with FG-labeled RGCs in control retinas. However, both the number of BM88-IR RGCs and their intensity decreased gradually between 4 and 28 days, preceding the loss of FG-labeled cells. These findings indicate that BM88 is not a good marker of surviving RGCs but may indicate abnormal RGC functioning, which precedes cell death.


Asunto(s)
Regulación hacia Abajo , Proteínas de la Membrana/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Traumatismos del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/patología , Animales , Biomarcadores , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Traumatismos del Nervio Óptico/patología , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/metabolismo
16.
J Clin Med ; 4(1): 37-65, 2014 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26237017

RESUMEN

Spinal cord injury (SCI) is a devastating trauma causing long-lasting disability. Although advances have occurred in the last decade in the medical, surgical and rehabilitative treatments of SCI, the therapeutic approaches are still not ideal. The use of cell transplantation as a therapeutic strategy for the treatment of SCI is promising, particularly since it can target cell replacement, neuroprotection and regeneration. Cell therapies for treating SCI are limited due to several translational roadblocks, including ethical and practical concerns regarding cell sources. The use of iPSCs has been particularly attractive, since they avoid the ethical and moral concerns that surround other stem cells. Furthermore, various cell types with potential for application in the treatment of SCI can be created from autologous sources using iPSCs. For applications in SCI, the iPSCs can be differentiated into neural precursor cells, neurons, oligodendrocytes, astrocytes, neural crest cells and mesenchymal stromal cells that can act by replacing lost cells or providing environmental support. Some methods, such as direct reprogramming, are being investigated to reduce tumorigenicity and improve reprogramming efficiencies, which have been some of the issues surrounding the use of iPSCs clinically to date. Recently, iPSCs have entered clinical trials for use in age-related macular degeneration, further supporting their promise for translation in other conditions, including SCI.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...