Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 35(5): 1416-1430, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28233425

RESUMEN

Traumatic brain injury (TBI) is soon predicted to become the third leading cause of death and disability worldwide. After the primary injury, a complex set of secondary injuries develops hours and days later with prolonged neuroinflammation playing a key role. TBI and other inflammatory conditions are currently being treated in preclinical and clinical trials by a number of cellular therapies. Mesenchymal stem cells (MSC) are of great interest due to their widespread usage, safety, and relative ease to isolate and culture. However, there has been a wide range in efficacy reported using MSC clinically and in preclinical models, likely due to differences in cell preparations and a significant amount of donor variability. In this study, we seek to find a correlation between in vitro activity and in vivo efficacy. We designed assays to explore the responsiveness of MSC to immunological cues to address the immunomodulatory properties of MSC, one of their primary modes of therapeutic activity in TBI. Our results showed intrinsic differences in the immunomodulatory capacity of MSC preparations from different bone marrow and amniotic fluid donors. This difference mirrored the therapeutic capacity of the MSC in an experimental model of TBI, an effect confirmed using siRNA knockdown of COX2 followed by overexpressing COX2. Among the immunomodulatory factors assessed, the therapeutic benefit correlated with the secretion of prostaglandin E2 (PGE2 ) by MSC prior to treatment, suggesting that measurement of PGE2 could be a very useful potency marker to create an index of predicted efficacy for preparations of MSC to treat TBI. Stem Cells 2017;35:1416-1430.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Dinoprostona/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Líquido Amniótico/citología , Animales , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Recuento de Células , Enfermedad Crónica , Constricción Patológica , Ciclooxigenasa 2/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inmunomodulación , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/patología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Permeabilidad , Ratas Sprague-Dawley
2.
Stem Cells Transl Med ; 5(1): 33-44, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26586775

RESUMEN

UNLABELLED: More than 6.5 million patients are burdened by the physical, cognitive, and psychosocial deficits associated with traumatic brain injury (TBI) in the U.S. Despite extensive efforts to develop neuroprotective therapies for this devastating disorder, there have been no successful outcomes in human clinical trials to date. Retrospective studies have shown that ß-adrenergic receptor blockers, specifically propranolol, significantly decrease mortality of TBI through mechanisms not yet fully elucidated but are thought to counterbalance a hyperadrenergic state resulting from a TBI. Conversely, cellular therapies have been shown to improve long-term behavior following TBI, likely by reducing inflammation. Given the nonredundancy in their therapeutic mechanisms, we hypothesized that a combination of acute propranolol followed by mesenchymal stem cells (MSCs) isolated from human bone marrow would have additive effects in treating a rodent model of TBI. We have found that the treatments are well-tolerated individually and in combination with no adverse events. MSCs decrease BBB permeability at 96 hours after injury, inhibit a significant accumulation of activated microglia/macrophage in the thalamic region of the brain both short and long term, and enhance neurogenesis short term. Propranolol decreases edema and reduces the number of fully activated microglia at 7 days and the number of semiactivated microglia at 120 days. Combinatory treatment improved cognitive and memory functions 120 days following TBI. Therefore, the results here suggest a new, efficacious sequential treatment for TBI may be achieved using the ß-blocker propranolol followed by MSC treatment. SIGNIFICANCE: Despite continuous efforts, traumatic brain injury (TBI) remains the leading cause of death and disability worldwide in patients under the age of 44. In this study, an animal model of moderate-severe TBI was treated with an acute dose of propranolol followed by a delayed dose of human mesenchymal stem cells (MSCs), resulting in improved short- and long-term measurements. These results have direct translational application. They reinforce the inevitable clinical trial of MSCs to treat TBI by demonstrating, among other benefits, a notable decrease in chronic neuroinflammation. More importantly, these results demonstrate that MSCs and propranolol, which is increasingly being used clinically for TBI, are compatible treatments that improve overall outcome.


Asunto(s)
Lesiones Encefálicas/terapia , Células Madre Mesenquimatosas/metabolismo , Microglía/metabolismo , Neurogénesis/efectos de los fármacos , Propranolol/farmacología , Adulto , Aloinjertos , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/patología , Microglía/patología , Ratas , Ratas Sprague-Dawley
3.
Sci Rep ; 4: 4565, 2014 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-24691433

RESUMEN

Advances in the field of Multipotent Mesenchymal Stromal cell (MSC) biology have demonstrated that MSCs can improve disease outcome when 'activated' to exert immunomodulatory effects. However, the precise mechanisms modulating MSC-immune cells interactions remain largely elusive. In here, we activated MSC based on a recent polarization paradigm, in which MSCs can be polarized towards a pro- or anti-inflammatory phenotype depending on the Toll-like receptor stimulated, to dissect the mechanisms through which MSCs physically interact with and modulate leukocytes in this context. Our data show that MSCs activated through the Toll-like receptor (TLR) 4 pathway increased VCAM-1 and ICAM-1 dependent binding of leukocytes. On the other hand, TLR3 stimulation strongly increases leukocytes affinity to MSC comparatively, through the formation of cable-like hyaluronic acid structures. In addition, TLR4 activation elicited secretion of pro-inflammatory mediators by MSCs, whereas TLR3-activated MSCs displayed a milder pro-inflammatory phenotype, similar to inactivated MSCs. However, the differently activated MSCs maintained their ability to suppress leukocyte activation at similar levels in our in vitro model, and this immunomodulatory property was shown here to be partially mediated by prostaglandin. These results reinforce the concept that alternate activation profiles control MSC responses and may impact the therapeutic use of MSCs.


Asunto(s)
Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Leucocitos Mononucleares/fisiología , Células Madre Mesenquimatosas/fisiología , Células Cultivadas , Humanos , Inflamación/metabolismo , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos Mononucleares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...