Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
eNeuro ; 9(4)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35840315

RESUMEN

Altered lipoprotein metabolism is considered a pathogenic component of amyotrophic lateral sclerosis (ALS). Apolipoprotein A1 (ApoA1), a major high-density lipoprotein (HDL) protein, is associated with prevention of vascular damage. However, ApoA1's effects on damaged endothelium in ALS are unknown. This study aimed to determine therapeutic potential of ApoA1 for endothelial cell (EC) repair under a pathologic condition reminiscent of ALS. We performed in vitro studies using mouse brain ECs (mBECs) exposed to plasma from symptomatic G93A SOD1 mice. Dosage effects of ApoA1, including inhibition of the phosphoinoside 3-kinase (PI3K)/Akt signaling pathway and integration of ApoA1 into mBECs were examined. Also, human bone marrow-derived endothelial progenitor cells (hBM-EPCs) and mBECs were co-cultured without cell contact to establish therapeutic mechanism of hBM-EPC transplantation. Results showed that ApoA1 significantly reduced mBEC death via the PI3K/Akt downstream signaling pathway. Also, ApoA1 was incorporated into mBECs as confirmed by blocked ApoA1 cellular integration. Co-culture system provided evidence that ApoA1 was secreted by hBM-EPCs and incorporated into injured mBECs. Thus, our study findings provide important evidence for ApoA1 as a potential novel therapeutic for endothelium protection in ALS. This in vitro study lays the groundwork for further in vivo research to fully determine therapeutic effects of ApoA1 in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Animales , Apolipoproteína A-I/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Endotelio/metabolismo , Humanos , Ratones , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Stem Cell Rev Rep ; 17(1): 9-32, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32789802

RESUMEN

The human population is in the midst of battling a rapidly-spreading virus- Severe Acute Respiratory Syndrome Coronavirus 2, responsible for Coronavirus disease 2019 or COVID-19. Despite the resurgences in positive cases after reopening businesses in May, the country is seeing a shift in mindset surrounding the pandemic as people have been eagerly trickling out from federally-mandated quarantine into restaurants, bars, and gyms across America. History can teach us about the past, and today's pandemic is no exception. Without a vaccine available, three lessons from the 1918 Spanish flu pandemic may arm us in our fight against COVID-19. First, those who survived the first wave developed immunity to the second wave, highlighting the potential of passive immunity-based treatments like convalescent plasma and cell-based therapy. Second, the long-term consequences of COVID-19 are unknown. Slow-progressive cases of the Spanish flu have been linked to bacterial pneumonia and neurological disorders later in life, emphasizing the need to reduce COVID-19 transmission. Third, the Spanish flu killed approximately 17 to 50 million people, and the lack of human response, overcrowding, and poor hygiene were key in promoting the spread and high mortality. Human behavior is the most important strategy for preventing the virus spread and we must adhere to proper precautions. This review will cover our current understanding of the pathology and treatment for COVID-19 and highlight similarities between past pandemics. By revisiting history, we hope to emphasize the importance of human behavior and innovative therapies as we wait for the development of a vaccine. Graphical Abstract.


Asunto(s)
COVID-19/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , COVID-19/patología , COVID-19/prevención & control , COVID-19/virología , Historia del Siglo XX , Humanos , Inmunización Pasiva , Influenza Pandémica, 1918-1919/historia , Pandemias/historia , Medicina Regenerativa/historia , SARS-CoV-2/patogenicidad , Sueroterapia para COVID-19
3.
Neuromolecular Med ; 22(4): 503-516, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32820422

RESUMEN

Repairing the damaged blood-CNS-barrier in amyotrophic lateral sclerosis (ALS) is necessary to prevent entry of detrimental blood-borne factors contributing to motor neuron dysfunction. Recently, we showed benefits of human bone marrow endothelial progenitor cell (hBM-EPC) transplantation into symptomatic ALS mice on barrier restoration by replacing damaged endothelial cells (ECs). Additionally, transplanted cells may endogenously repair ECs by secreting angiogenic factors as our subsequent in vitro study demonstrated. Based on these study results, hBM-EPCs may secrete extracellular vesicles, which may contain and transfer diverse vesicular biomolecules towards maintenance of EC functionality. The study aimed to characterize extracellular vesicles (EVs) derived from hBM-EPCs as potential cell-free therapeutics for endothelium repair in ALS. EVs were isolated from hBM-EPC media at different culture times and vesicle properties were evaluated. The protective effects of EVs on mouse brain endothelial cells (mBECs) exposed to ALS mouse plasma were investigated. Uptake and blockage of EVs from GFP-transfected hBM-EPCs in ECs were determined in vitro. Results showed that EVs isolated from hBM-EPCs as nanosized vesicles significantly reduced mBEC damage from the pathological environment and these EVs were taken up by cells. Blockage of ß1 integrin on EVs prevented internalization of vesicles in mBECs. Together, these results provide evidence for potential of hBM-EPC-derived EVs as novel cell-free therapeutics for repair of endothelium in ALS. Although determining translational potential of hBM-EPC-derived EVs will require evaluation in vivo, this in vitro study represents a step towards an extracellular vesicle-based approach for repair of the damaged microvascular endothelium in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Células Progenitoras Endoteliales/ultraestructura , Vesículas Extracelulares/trasplante , Esclerosis Amiotrófica Lateral/sangre , Animales , Barrera Hematoencefálica , Células de la Médula Ósea , Células Cultivadas , Medios de Cultivo Condicionados/química , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Vesículas Extracelulares/ultraestructura , Genes Reporteros , Humanos , Masculino , Ratones , Superóxido Dismutasa-1/genética
4.
Nanomedicine ; 29: 102247, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32599163

RESUMEN

Traumatic brain injury (TBI) is a devastating neurological disorder, although the underlying pathophysiology is poorly understood. TBI causes blood-brain barrier (BBB) disruption, immune cell trafficking, neuroinflammation and neurodegeneration. CCL20 is an important chemokine mediating neuroinflammation. Human mesenchymal stem cell (hMSC) therapy is a promising regenerative approach but the inflammatory microenvironment in the brain tends to decrease the efficacy of the hMSC transplantation. Reducing the inflammation prior to hMSC therapy improves the outcome. We developed a combined nano-cell therapy by using dendrimers complexed with plasmids (dendriplexes) targeting CCL20 and its sole receptor CCR6 to reduce inflammation followed by hMSC transplantation. Treatment of TBI mice with shRNA conjugated dendriplexes followed by hMSC administration downregulated the inflammatory markers and significantly increased brain-derived neurotrophic factor (BDNF) expression in the cerebral cortex indicating future possible neurogenesis and improved behavioral deficits. Taken together, this nano-cell therapy ameliorates neuroinflammation and promotes brain tissue repair after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Factor Neurotrófico Derivado del Encéfalo/genética , Quimiocina CCL20/genética , Inflamación/terapia , Receptores CCR6/genética , Animales , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Quimiocina CCL20/antagonistas & inhibidores , Dendrímeros/química , Dendrímeros/farmacología , Humanos , Inflamación/genética , Inflamación/patología , Trasplante de Células Madre Mesenquimatosas , Ratones , Plásmidos/química , Plásmidos/genética , Plásmidos/farmacología , ARN Interferente Pequeño/farmacología , Receptores CCR6/antagonistas & inhibidores
5.
Cell Transplant ; 29: 963689720913494, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32207340

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal disease of motor neuron degeneration in the brain and spinal cord. Progressive paralysis of the diaphragm and other respiratory muscles leading to respiratory dysfunction and failure is the most common cause of death in ALS patients. Respiratory impairment has also been shown in animal models of ALS. Vascular pathology is another recently recognized hallmark of ALS pathogenesis. Central nervous system (CNS) capillary damage is a shared disease element in ALS rodent models and ALS patients. Microvascular impairment outside of the CNS, such as in the lungs, may occur in ALS, triggering lung damage and affecting breathing function. Stem cell therapy is a promising treatment for ALS. However, this therapeutic strategy has primarily targeted rescue of degenerated motor neurons. We showed functional benefits from intravenous delivery of human bone marrow (hBM) stem cells on restoration of capillary integrity in the CNS of an superoxide dismutase 1 (SOD1) mouse model of ALS. Due to the widespread distribution of transplanted cells via this route, administered cells may enter the lungs and effectively restore microvasculature in this respiratory organ. Here, we provided preliminary evidence of the potential role of microvasculature dysfunction in prompting lung damage and treatment approaches for repair of respiratory function in ALS. Our initial studies showed proof-of-principle that microvascular damage in ALS mice results in lung petechiae at the late stage of disease and that systemic transplantation of mainly hBM-derived endothelial progenitor cells shows potential to promote lung restoration via re-established vascular integrity. Our new understanding of previously underexplored lung competence in this disease may facilitate therapy targeting restoration of respiratory function in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Microvasos/patología , Neuronas Motoras/metabolismo , Trasplante de Células Madre , Animales , Humanos , Pulmón/patología , Médula Espinal/patología
6.
CNS Neurosci Ther ; 26(6): 616-627, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32157822

RESUMEN

Traumatic brain injury (TBI) causes death and disability in the United States and around the world. The traumatic insult causes the mechanical injury of the brain and primary cellular death. While a comprehensive pathological mechanism of TBI is still lacking, the focus of the TBI research is concentrated on understanding the pathophysiology and developing suitable therapeutic approaches. Given the complexities in pathophysiology involving interconnected immunologic, inflammatory, and neurological cascades occurring after TBI, the therapies directed to a single mechanism fail in the clinical trials. This has led to the development of the paradigm of a combination therapeutic approach against TBI. While there are no drugs available for the treatment of TBI, stem cell therapy has shown promising results in preclinical studies. But, the success of the therapy depends on the survival of the stem cells, which are limited by several factors including route of administration, health of the administered cells, and inflammatory microenvironment of the injured brain. Reducing the inflammation prior to cell administration may provide a better outcome of cell therapy following TBI. This review is focused on different therapeutic approaches of TBI and the present status of the clinical trials.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Terapia Combinada/métodos , Terapia Combinada/tendencias , Humanos , Trasplante de Células Madre Mesenquimatosas/tendencias
7.
Sci Rep ; 9(1): 13646, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541141

RESUMEN

Traumatic brain injury is a leading cause of death and disability around the world. So far, drugs are not available to repair brain damage. Human mesenchymal stem cell (hMSC) transplantation therapy is a promising approach, although the inflammatory microenvironment of the injured brain affects the efficacy of transplanted hMSCs. We hypothesize that reducing the inflammation in the cerebral microenvironment by reducing pro-inflammatory chemokines prior to hMSC administration will improve the efficacy of hMSC therapy. In a rat model of lateral fluid percussion injury, combined pioglitazone (PG) and hMSC (combination) treatment showed less anxiety-like behavior and improved sensorimotor responses to a noxious cold stimulus. Significant reduction in brain lesion volume, neurodegeneration, microgliosis and astrogliosis were observed after combination treatment. TBI induced expression of inflammatory chemokine CCL20 and IL1-ß were significantly decreased in the combination treatment group. Combination treatment significantly increased brain-derived neurotrophic factor (BDNF) level and subventricular zone (SVZ) neurogenesis. Taken together, reducing proinflammatory cytokine expression in the cerebral tissues after TBI by PG administration and prior to hMSC therapy improves the outcome of the therapy in which BDNF could have a role.


Asunto(s)
Antiinflamatorios/administración & dosificación , Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Pioglitazona/administración & dosificación , Administración Intranasal , Animales , Antiinflamatorios/farmacología , Lesiones Traumáticas del Encéfalo/etiología , Lesiones Traumáticas del Encéfalo/inmunología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Quimiocina CCL20/metabolismo , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-1beta/metabolismo , Ventrículos Laterales/metabolismo , Masculino , Pioglitazona/farmacología , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Regulación hacia Arriba
8.
Mil Med ; 184(11-12): e626-e631, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31004163

RESUMEN

INTRODUCTION: The United States has been actively involved in major armed conflicts over the last 15 years. As a result, a significant proportion of active duty service personnel and returning veterans have endured combat, putting them at risk for developing post-traumatic stress disorder (PTSD), a disabling disorder that may occur after exposure to a traumatic event. Current therapies often require long-term, time-intensive and costly commitment from the patient and have variable degrees of success. There remains an ongoing need for better therapies, including complementary medicine approaches that can effectively reduce PTSD symptoms. While anecdotal evidence suggests that routine practice of Brazilian Jiu Jitsu (BJJ) can reduce symptoms of PTSD, there have been no formal studies to address this. MATERIALS AND METHODS: This study was approved by the University of South Florida Institutional Review Board (#PRO00019430). Male US active duty service members and veterans from the Tampa area participated in a 5-month (40 sessions) BJJ training program. Before beginning and again midway through and upon completion of training the participants completed several validated self-report measures that addressed symptoms of PTSD and other co-morbid conditions. Effect size and 95% confidence intervals were determined using a within-person single-group pretest-posttest design. RESULTS: Study participants demonstrated clinically meaningful improvements in their PTSD symptoms as well as decreased symptoms of major depressive disorder, generalized anxiety and decreased alcohol use; effect sizes varied from 0.80 to 1.85. CONCLUSIONS: The results from this first-of-kind pilot study suggest that including BJJ as a complementary treatment to standard therapy for PTSD may be of value. It will be necessary to validate these promising results with a larger subject cohort and a more rigorous experimental design before routinely recommending this complementary therapy.


Asunto(s)
Artes Marciales/psicología , Personal Militar/psicología , Trastornos por Estrés Postraumático/terapia , Veteranos/psicología , Adulto , Femenino , Florida , Humanos , Masculino , Artes Marciales/educación , Artes Marciales/estadística & datos numéricos , Personal Militar/estadística & datos numéricos , Proyectos Piloto , Psicometría/instrumentación , Psicometría/métodos , Trastornos por Estrés Postraumático/psicología , Encuestas y Cuestionarios , Estados Unidos , Veteranos/estadística & datos numéricos
9.
J Neurosci Res ; 96(8): 1353-1366, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29732581

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an adult onset neurodegenerative disease characterized by progressive motor neuron degeneration in the brain and spinal cord leading to muscle atrophy, paralysis, and death. Mitochondrial dysfunction is a major contributor to motor neuron degeneration associated with ALS progression. Mitochondrial abnormalities have been determined in spinal cords of animal disease models and ALS patients. However, molecular mechanisms leading to mitochondrial dysfunction in sporadic ALS (sALS) patients remain unclear. Also, segmental or regional variation in mitochondrial activity in the spinal cord has not been extensively examined in ALS. In our study, the activity of mitochondrial electron transport chain complex IV was examined in post-mortem gray and white matter of the cervical and lumbar spinal cords from male and female sALS patients and controls. Mitochondrial distribution and density in spinal cord motor neurons, lateral funiculus, and capillaries in gray and white matter were analyzed by immunohistochemistry. Results showed that complex IV activity was significantly decreased only in gray matter in both cervical and lumbar spinal cords from ALS patients. In ALS cervical and lumbar spinal cords, significantly increased mitochondrial density and altered distribution were observed in motor neurons, lateral funiculus, and cervical white matter capillaries. Discrete decreased complex IV activity in addition to changes in mitochondria distribution and density determined in the spinal cord in sALS patients are novel findings. These explicit mitochondrial defects in the spinal cord may contribute to ALS pathogenesis and should be considered in development of therapeutic approaches for this disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Médula Espinal/metabolismo , Médula Espinal/patología , Adulto , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Sustancia Gris/patología , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Sustancia Blanca/patología
10.
Cell Transplant ; 24(4): 721-35, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25413246

RESUMEN

Human umbilical cord blood (HUCB) cell therapies have shown promising results in reducing brain infarct volume and most importantly in improving neurobehavioral function in rat permanent middle cerebral artery occlusion, a model of stroke. In this study, we examined the gene expression profile in neurons subjected to oxygen-glucose deprivation (OGD) with or without HUCB treatment and identified signaling pathways (Akt/MAPK) important in eliciting HUCB-mediated neuroprotective responses. Gene chip microarray analysis was performed using RNA samples extracted from the neuronal cell cultures from four experimental groups: normoxia, normoxia+HUCB, OGD, and OGD+HUCB. Both quantitative RT-PCR and immunohistochemistry were carried out to verify the microarray results. Using the Genomatix software program, promoter regions of selected genes were compared to reveal common transcription factor-binding sites and, subsequently, signal transduction pathways. Under OGD condition, HUCB cells significantly reduced neuronal loss from 68% to 44% [one-way ANOVA, F(3, 16)=11, p=0.0003]. Microarray analysis identified mRNA expression of Prdx5, Vcam1, CCL20, Alcam, and Pax6 as being significantly altered by HUCB cell treatment. Inhibition of the Akt pathway significantly abolished the neuroprotective effect of HUCB cells [one-way ANOVA, F(3, 11)=8.663, p=0.0031]. Our observations show that HUCB neuroprotection is dependent on the activation of the Akt signaling pathway that increases transcription of the Prdx5 gene. We concluded that HUCB cell therapy would be a promising treatment for stroke and other forms of brain injury by modifying acute gene expression to promote neural cell protection.


Asunto(s)
Sangre Fetal/metabolismo , Regulación de la Expresión Génica , Infarto de la Arteria Cerebral Media/patología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transcriptoma , Animales , Sitios de Unión , Células Cultivadas , Técnicas de Cocultivo , Sangre Fetal/citología , Sangre Fetal/trasplante , Humanos , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/terapia , Neuronas/citología , Fármacos Neuroprotectores , Análisis de Secuencia por Matrices de Oligonucleótidos , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factores de Transcripción/química , Factores de Transcripción/metabolismo
11.
J Neuroimmune Pharmacol ; 9(5): 679-89, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25104571

RESUMEN

The delayed immune response to stroke is responsible for the increased neural injury that continues to occur after the initial ischemic event. This delayed immune response has been linked to the spleen, as splenectomy prior to middle cerebral artery occlusion (MCAO) is neuroprotective. Interferon gamma (IFNγ) is linked to the splenic response, which enhances neural injury following MCAO. IFNγ activates the expression of the inflammatory chemokine interferon-inducible protein 10 (IP-10). This study was designed to determine the role of IFNγ signaling in the inflammatory response following MCAO. Expression of IP-10 increased in the brain and the spleen following MCAO. Splenectomy inhibited the increase of IP-10 in the brain post-MCAO, while recombinant IFNγ administration to splenectomized rats returned IP-10 levels in the brain to levels found in rats after MCAO only. Systemic administration of an IFNγ neutralizing antibody to MCAO-treated rats reduced infarct volume and IP-10 levels in the brain. T cell infiltration was reduced in the MCAO-damaged brains of IFNγ antibody-treated animals relative to those that received isotype control antibodies. Additionally, inhibiting IFNγ signaling with splenectomy or an IFNγ neutralizing antibody blocked the induction of IP-10 expression and decreased neurodegeneration following MCAO. Targeting this pro-inflammatory pathway following stroke could be a promising stroke therapeutic.


Asunto(s)
Quimiocina CXCL10/biosíntesis , Interferón gamma/uso terapéutico , Enfermedades Neurodegenerativas/metabolismo , Transducción de Señal/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Mediadores de Inflamación/metabolismo , Interferón gamma/farmacología , Masculino , Enfermedades Neurodegenerativas/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/patología
12.
Eur J Neurosci ; 40(7): 3111-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25041106

RESUMEN

Human umbilical cord blood (HUCB) cells have shown efficacy in rodent models of focal ischemia and in vitro systems that recapitulate stroke conditions. One potential mechanism of protection is through secretion of soluble factors that protect neurons and oligodendrocytes (OLs) from oxidative stress. To overcome practical issues with cellular therapies, identification of soluble factors released by HUCB and other stem cells may pave the way for treatment modalities that are safer for a larger percentage of stroke patients. Among these soluble factors is leukemia inhibitory factor (LIF), a cytokine that exerts pleiotropic effects on cell survival. Here, data show that LIF effectively reduced infarct volume, reduced white matter injury and improved functional outcomes when administered to rats following permanent middle cerebral artery occlusion. To further explore downstream signaling, primary oligodendrocyte cultures were exposed to oxygen-glucose deprivation to mimic stroke conditions. LIF significantly reduced lactate dehydrogenase release from OLs, reduced superoxide dismutase activity and induced peroxiredoxin 4 (Prdx4) transcript. Additionally, the protective and antioxidant capacity of LIF was negated by both Akt inhibition and co-incubation with Prdx4-neutralising antibodies, establishing a role for the Akt signaling pathway and Prdx4-mediated antioxidation in LIF protection.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Factor Inhibidor de Leucemia/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Oligodendroglía/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Factor Inhibidor de Leucemia/farmacología , Fármacos Neuroprotectores/farmacología , Proteína Oncogénica v-akt/metabolismo , Peroxirredoxinas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Sustancia Blanca/efectos de los fármacos
13.
Cell Transplant ; 23(12): 1613-30, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25565636

RESUMEN

Sanfilippo syndrome type III B (MPS III B) is an inherited disorder characterized by a deficiency of α-N-acetylglucosaminidase (Naglu) enzyme leading to accumulation of heparan sulfate in lysosomes and severe neurological deficits. We have previously shown that a single administration of human umbilical cord mononuclear cells (hUCB MNCs) into Naglu knockout mice decreased behavioral abnormalities and tissue pathology. In this study, we tested whether repeated doses of hUCB MNCs would be more beneficial than a single dose of cells. Naglu mice at 3 months of age were randomly assigned to either a Media-only group or one of three hUCB MNC treatment groups--single low dose (3 × 10(6) cells), single high dose (1.8 × 10(7) cells), or multiple doses (3 × 10(6) cells monthly for 6 months) delivered intravenously; cyclosporine was injected intraperitoneally to immune suppress the mice for the duration of the study. An additional control group of wild-type mice was also used. We measured anxiety in an open field test and cognition in an active avoidance test prior to treatment and then at monthly intervals for 6 months. hUCB MNCs restored normal anxiety-like behavior in these mice (p < 0.001). The repeated cell administrations also restored hippocampal cytoarchitecture, protected the dendritic tree, decreased GM3 ganglioside accumulation, and decreased microglial activation, particularly in the hippocampus and cortex. These data suggest that the neuroprotective effect of hUCB MNCs can be enhanced by repeated cell administrations.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Mucopolisacaridosis III/terapia , Cordón Umbilical/citología , Acetilglucosaminidasa/deficiencia , Acetilglucosaminidasa/metabolismo , Animales , Ansiedad/complicaciones , Ansiedad/fisiopatología , Reacción de Prevención , Conducta Animal , Encéfalo/patología , Recuento de Células , Cognición , Dendritas/patología , Modelos Animales de Enfermedad , Femenino , Gangliósido G(M3)/metabolismo , Humanos , Masculino , Ratones Noqueados , Microglía/patología , Mucopolisacaridosis III/complicaciones , Mucopolisacaridosis III/fisiopatología , Fenotipo , Resultado del Tratamiento , Orina
14.
Age (Dordr) ; 35(6): 2071-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23263793

RESUMEN

Neurogenesis occurs throughout life but significantly decreases with age. Human umbilical cord blood mononuclear cells (HUCB MNCs) have been shown to increase the proliferation of neural stem cells (NSCs) in the dentate gyrus (DG) of the hippocampus and the subgranular zone of aging rats (Bachstetter et al., BMC Neurosci 9:22, 2008), but it is unclear which fraction or combination of the HUCB MNCs are responsible for neurogenesis. To address this issue, we examined the ability of HUCB MNCs, CD4+, CD8+, CD3+, CD14+, and CD133+ subpopulations to increase proliferation of NSCs both in vitro and in vivo. NSCs were first grown in conditioned media generated from HUCB cultures, and survival and proliferation of NSC were determined with the fluorescein diacetate/propidium iodide and 5-bromo-2'-deoxyuridine incorporation assays, respectively. In a second study, we injected HUCB cells intravenously in young and aged Fisher 344 rats and examined proliferation in the DG at 1 week (study 2.1) and 2 weeks (study 2.2) postinjection. The effects of the HUCB MNC fractions on dendritic spine density and microglial activation were also assessed. HUCB T cells (CD3+, CD4+, and CD8+ cells) induced proliferation of NSCs (p < 0.001) and increased cell survival. In vivo, HUCB-derived CD4+ cells increased NSC proliferation at both 1 and 2 weeks while also enhancing the density of dendritic spines at 1 week and decreasing inflammation at 2 weeks postinjection. Collectively, these data indicate that a single injection of HUCB-derived T cells induces long-lasting effects and may therefore have tremendous potential to improve aging neurogenesis.


Asunto(s)
Envejecimiento/fisiología , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Sangre Fetal/citología , Hipocampo/crecimiento & desarrollo , Linfocitos T/trasplante , Animales , Supervivencia Celular , Células Cultivadas , Sangre Fetal/inmunología , Hipocampo/citología , Hipocampo/inmunología , Humanos , Masculino , Ratas , Ratas Endogámicas F344 , Linfocitos T/inmunología
15.
J Neuroimmune Pharmacol ; 7(4): 1017-24, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23054371

RESUMEN

The splenic response to stroke is a proinflammatory reaction to ischemic injury resulting in expanded neurodegeneration. Splenectomy reduces neural injury in rodent models of hemorrhagic and ischemic stroke, however the exact nature of this response has yet to be fully understood. This study examines the migration of splenocytes after brain ischemia utilizing carboxyfluorescein diacetate succinimidyl ester (CFSE) to label them in vivo. The spleen was found to significantly decrease in size from 24 to 48 h following middle cerebral artery occlusion (MCAO) in rats compared to sham operated controls. By 96 h post-MCAO the spleen size returned to levels not different from sham operated rats. To track splenocyte migration following MCAO, spleens were injected with CFSE to label cells. CFSE positive cell numbers were significantly reduced in the 48 h MCAO group versus 48 h sham and CFSE labeled cells were equivalent in 96 h MCAO and sham groups. A significant increase of labeled lymphocyte, monocytes, and neutrophils was detected in the blood at 48 h post-MCAO when compared to the other groups. CFSE labeled cells migrated to the brain following MCAO but appear to remain within the vasculature. These cells were identified as natural killer cells (NK) and monocytes at 48 h and at 96 h post-MCAO NK cells, T cells and monocytes. After ischemic injury, splenocytes enter into systemic circulation and migrate to the brain exacerbating neurodegeneration.


Asunto(s)
Bazo/patología , Accidente Cerebrovascular/patología , Animales , Recuento de Células , Movimiento Celular , Fluoresceínas , Colorantes Fluorescentes , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Células Asesinas Naturales/fisiología , Flujometría por Láser-Doppler , Masculino , Ratas , Ratas Sprague-Dawley , Bazo/citología , Accidente Cerebrovascular/sangre , Succinimidas
16.
Int Rev Neurobiol ; 102: 91-106, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22748827

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease with a complicated and poorly understood pathogenesis. Strong evidence indicates impairment of all neurovascular unit components including the blood-brain and blood-spinal cord barriers (BBB/BSCB) in both patients and animal models. The present review provides an updated analysis of the microvascular pathology and impaired BBB/BSCB in ALS. Based on experimental and clinical ALS studies, the roles of cellular components, cell interactions, tight junctions, transport systems, cytokines, matrix metalloproteinases, and free radicals in the BBB/BSCB disruption are discussed. The impact of BBB/BSCB damage in ALS pathogenesis is a novel research topic, and this review will reveal some aspects of microvascular pathology involved in the disease and hopefully engender new therapeutic approaches.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Barrera Hematoencefálica/fisiopatología , Permeabilidad Capilar/fisiología , Médula Espinal/fisiopatología , Animales , Transporte Biológico/fisiología , Modelos Animales de Enfermedad , Humanos , Modelos Neurológicos , Médula Espinal/irrigación sanguínea
17.
Metab Brain Dis ; 27(2): 131-41, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22354752

RESUMEN

Delayed neuronal death associated with stroke has been increasingly linked to the immune response to the injury. Splenectomy prior to middle cerebral artery occlusion (MCAO) is neuroprotective and significantly reduces neuroinflammation. The present study investigated whether splenic signaling occurs through interferon gamma (IFNγ). IFNγ was elevated early in spleens but later in the brains of rats following MCAO. Splenectomy decreased the amount of IFNγ in the infarct post-MCAO. Systemic administration of recombinant IFNγ abolished the protective effects of splenectomy with a concurrent increase in INFγ expression in the brain. These results suggest a role for spleen-derived IFNγ in stroke pathology.


Asunto(s)
Interferón gamma/fisiología , Degeneración Nerviosa/fisiopatología , Bazo/fisiopatología , Accidente Cerebrovascular/fisiopatología , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Hipoxia de la Célula , Células Cultivadas , Femenino , Fluoresceínas , Colorantes Fluorescentes , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Interferón gamma/farmacología , Flujometría por Láser-Doppler , Ligadura , Masculino , Arteria Cerebral Media/fisiología , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oligodendroglía/metabolismo , Compuestos Orgánicos , Embarazo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Transducción de Señal/fisiología , Bazo/metabolismo , Esplenectomía
18.
J Biol Chem ; 287(6): 4177-87, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22158864

RESUMEN

Human umbilical cord blood (HUCB) cells protect the brain against ischemic injury, yet the mechanism of protection remains unclear. Using both in vitro and in vivo paradigms, this study examined the role of Akt signaling and peroxiredoxin 4 expression in human umbilical cord blood cell-mediated protection of oligodendrocytes from ischemic conditions. As previously reported, the addition of HUCB cells to oligodendrocyte cultures prior to oxygen glucose deprivation significantly enhanced oligodendrocyte survival. The presence of human umbilical cord blood cells also increased Akt phosphorylation and elevated peroxiredoxin 4 expression in oligodendrocytes. Blocking either Akt or peroxiredoxin 4 activity with Akt Inhibitor IV or a peroxiredoxin 4-neutralizing antibody, respectively, negated the protective effects of human umbilical cord blood cells. In vivo, systemic administration of human umbilical cord blood cells 48 h after middle cerebral artery occlusion increased Akt phosphorylation and peroxiredoxin 4 protein expression while reducing proteolytic cleavage of caspase 3 in oligodendrocytes residing in the ipsilateral external capsule. Moreover, human umbilical cord blood cells protected striatal white matter bundles from degeneration following middle cerebral artery occlusion. These results suggest that the soluble factors released from human umbilical cord blood cells converge on Akt to elevate peroxiredoxin 4 levels, and these effects contribute to oligodendrocyte survival.


Asunto(s)
Isquemia Encefálica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Cordón Umbilical/citología , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Caspasa 3/biosíntesis , Supervivencia Celular , Humanos , Oligodendroglía/patología , Peroxirredoxinas/biosíntesis , Fosforilación , Ratas , Ratas Sprague-Dawley
19.
Brain Res ; 1398: 113-25, 2011 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-21632035

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease with a complicated pathogenesis. Compelling evidence indicates impairment of all neurovascular unit components including the blood-brain and blood-spinal cord barriers (BBB/BSCB) in both patients and animal models, leading to classification of ALS as a neurovascular disease. The present review provides an updated analysis of the normal and impaired BBB/BSCB, focusing on the ALS-altered barrier. Here we describe the roles of cellular components, tight junctions, transport systems, cell interactions, cytokines, matrix metalloproteinases, and free radicals in the BBB/BSCB disruption, along with recent evidence from experimental and clinical ALS studies. The BBB/BSCB is a promising research area in ALS and this review will reveal some aspects of microvascular pathology in ALS and hopefully provide ideas for the development of new therapeutic strategies.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/fisiopatología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Sistema Nervioso Central/metabolismo , Arterias Cerebrales/metabolismo , Arterias Cerebrales/fisiopatología , Humanos
20.
Cell Transplant ; 20(1): 85-94, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20887684

RESUMEN

Stem cell transplantation is a potentially important means of treatment for a number of disorders. Two different stem cell populations of interest are mononuclear umbilical cord blood cells and menstrual blood-derived stem cells. These cells are relatively easy to obtain, appear to be pluripotent, and are immunologically immature. These cells, particularly umbilical cord blood cells, have been studied as either single or multiple injections in a number of animal models of neurodegenerative disorders with some degree of success, including stroke, Alzheimer's disease, amyotrophic lateral sclerosis, and Sanfilippo syndrome type B. Evidence of anti-inflammatory effects and secretion of specific cytokines and growth factors that promote cell survival, rather than cell replacement, have been detected in both transplanted cells.


Asunto(s)
Células Madre Adultas/citología , Células Sanguíneas/citología , Sangre Fetal/citología , Enfermedades Neurodegenerativas/terapia , Trasplante de Células Madre , Células Madre Adultas/trasplante , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA