Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Exp Neurol ; 374: 114727, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38360257

RESUMEN

Previous studies have demonstrated that endogenous tissue-type plasminogen activator (tPA) is upregulated in the brain after an acute ischemic stroke (AIS). While mixed results were observed in genetic models, the pharmacological inhibition of endogenous tPA showed beneficial effects. Treatment with exogenous recombinant tPA exacerbated brain damage in rodent models of stroke. Despite the detrimental effects of tPA in ischemic stroke, recombinant tPA is administered to AIS patients to recanalize the occluded blood vessels because the benefits of its administration outweigh the risks associated with tPA upregulation and increased activity. We hypothesized that tPA knockdown following recanalization would ameliorate sensorimotor deficits and reduce brain injury. Young male and female rats (2-3 months old) were subjected to transient focal cerebral ischemia by occlusion of the right middle cerebral artery. Shortly after reperfusion, rats from appropriate cohorts were administered a nanoparticle formulation containing tPA shRNA or control shRNA plasmids (1 mg/kg) intravenously via the tail vein. Infarct volume during acute and chronic phases, expression of matrix metalloproteinases (MMPs) 1, 3, and 9, enlargement of cerebral ventricle volume, and white matter damage were all reduced by shRNA-mediated gene silencing of tPA following reperfusion. Additionally, recovery of somatosensory and motor functions was improved. In conclusion, our results provide evidence that reducing endogenous tPA following recanalization improves functional outcomes and reduces post-stroke brain damage.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Ratas , Masculino , Femenino , Animales , Lactante , Activador de Tejido Plasminógeno , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Fibrinolíticos/uso terapéutico , Fibrinolíticos/farmacología , Modelos Animales de Enfermedad
2.
Front Neurosci ; 16: 1012812, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267234

RESUMEN

We recently showed that the post-ischemic induction of matrix metalloproteinase-12 (MMP-12) in the brain degrades tight junction proteins, increases MMP-9 and TNFα expression, and contributes to the blood-brain barrier (BBB) disruption, apoptosis, demyelination, and infarct volume development. The objectives of this study were to (1) determine the effect of MMP-12 suppression by shRNA-mediated gene silencing on neurological/functional recovery, (2) establish the optimal timing of MMP-12shRNA treatment that provides maximum therapeutic benefit, (3) compare the effectiveness of acute versus chronic MMP-12 suppression, and (4) evaluate potential sex-related differences in treatment outcomes. Young male and female Sprague-Dawley rats were subjected to transient middle cerebral artery occlusion and reperfusion. Cohorts of rats were administered either MMP-12shRNA or scrambled shRNA sequence (control) expressing plasmids (1 mg/kg; i.v.) formulated as nanoparticles. At designated time points after reperfusion, rats from various groups were subjected to a battery of neurological tests to assess their reflex, balance, sensory, and motor functions. Suppression of MMP-12 promoted the neurological recovery of stroke-induced male and female rats, although the effect was less apparent in females. Immediate treatment after reperfusion resulted in a better recovery of sensory and motor function than delayed treatments. Chronic MMP-12 suppression neither enhanced nor diminished the therapeutic effects of acute MMP-12 suppression, indicating that a single dose of plasmid may be sufficient. We conclude that suppressing MMP-12 after an ischemic stroke is a promising therapeutic strategy for promoting the recovery of neurological function.

3.
Neurochem Int ; 161: 105436, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36283468

RESUMEN

Tissue-type plasminogen activator (t-PA) expression is known to increase following transient focal cerebral ischemia and reperfusion. Previously, we reported downregulation of t-PA upon suppression of matrix metalloproteinase-12 (MMP-12), following transient focal cerebral ischemia and reperfusion. We now present data on the temporal expression of t-PA in the brain after transient ischemia, as well as the interaction between MMP-12 and t-PA, two proteases associated with the breakdown of the blood-brain barrier (BBB) and ischemic brain damage. We hypothesized that there might be reciprocal interactions between MMP-12 and t-PA in the brain after ischemic stroke. This hypothesis was tested using shRNA-mediated gene silencing and computational modeling. Suppression of t-PA following transient ischemia and reperfusion in rats attenuated MMP-12 expression in the brain. The overall effect of t-PA shRNA administration was to attenuate the degradation of BBB tight junction protein claudin-5, diminish BBB disruption, and reduce neuroinflammation by decreasing the expression of the microglia/macrophage pro-inflammatory M1 phenotype (CD68, iNOS, IL-1ß, and TNFα). Reduced BBB disruption and subsequent lack of infiltration of macrophages (the main source of MMP-12 in the ischemic brain) could account for the decrease in MMP-12 expression after t-PA suppression. Computational modeling of in silico protein-protein interactions indicated that MMP-12 and t-PA may interact physically. Overall, our findings demonstrate that MMP-12 and t-PA interact directly or indirectly at multiple levels in the brain following an ischemic stroke. The present findings could be useful in the development of new pharmacotherapies for the treatment of stroke.


Asunto(s)
Isquemia Encefálica , Ataque Isquémico Transitorio , Accidente Cerebrovascular Isquémico , Metaloproteinasa 12 de la Matriz , Activador de Tejido Plasminógeno , Animales , Ratas , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Ataque Isquémico Transitorio/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , ARN Interferente Pequeño/genética , Activador de Tejido Plasminógeno/metabolismo
4.
Stroke Vasc Neurol ; 6(4): 519-527, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33741744

RESUMEN

BACKGROUND AND PURPOSE: The therapeutic potential of different stem cells for ischaemic stroke treatment is intriguing and somewhat controversial. Recent results from our laboratory have demonstrated the potential benefits of human umbilical cord blood-derived mesenchymal stem cells (MSC) in a rodent stroke model. We hypothesised that MSC treatment would effectively promote the recovery of sensory and motor function in both males and females, despite any apparent sex differences in post stroke brain injury. METHODS: Transient focal cerebral ischaemia was induced in adult Sprague-Dawley rats by occlusion of the middle cerebral artery. Following the procedure, male and female rats of the untreated group were euthanised 1 day after reperfusion and their brains were used to estimate the resulting infarct volume and tissue swelling. Additional groups of stroke-induced male and female rats were treated with MSC or vehicle and were subsequently subjected to a battery of standard neurological/neurobehavioral tests (Modified Neurological Severity Score assessment, adhesive tape removal, beam walk and rotarod). The tests were administered at regular intervals (at days 1, 3, 5, 7 and 14) after reperfusion to determine the time course of neurological and functional recovery after stroke. RESULTS: The infarct volume and extent of swelling of the ischaemic brain were similar in males and females. Despite similar pathological stroke lesions, the clinical manifestations of stroke were more pronounced in males than females, as indicated by the neurological scores and other tests. MSC treatment significantly improved the recovery of sensory and motor function in both sexes, and it demonstrated efficacy in both moderate stroke (females) and severe stroke (males). CONCLUSIONS: Despite sex differences in the severity of post stroke outcomes, MSC treatment promoted the recovery of sensory and motor function in male and female rats, suggesting that it may be a promising treatment for stroke.


Asunto(s)
Isquemia Encefálica , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Accidente Cerebrovascular , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/patología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/terapia
5.
Transl Stroke Res ; 12(5): 923-936, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33426628

RESUMEN

The intense inflammatory response triggered in the brain after focal cerebral ischemia is detrimental. Recently, we showed that the suppression of toll-like receptors (TLRs) 2 and 4 attenuates infarct size and reduces the expression of pro-inflammatory cytokines in the ischemic brain. In this study, we further examined the effect of unsuppressed induction of TLRs 2 and 4 on the expression of its downstream signaling molecules and pro-inflammatory cytokines 1 week after reperfusion. The primary purpose of this study was to investigate the effect of simultaneous knockdown of TLRs 2 and 4 on M1/M2 microglial polarization dynamics and post-stroke neurological deficits and the recovery. Transient focal cerebral ischemia was induced in young adult male Sprague-Dawley rats by the middle cerebral artery occlusion (MCAO) procedure using a monofilament suture. Appropriate cohorts of rats were treated with a nanoparticle formulation of TLR2shRNA and TLR4shRNA (T2sh+T4sh) expressing plasmids (1 mg/kg each of T2sh and T4sh) or scrambled sequence inserted vector (vehicle control) expressing plasmids (2 mg/kg) intravenously via tail vein immediately after reperfusion. Animals from various cohorts were euthanized during reperfusion, and the ischemic brain tissue was isolated and utilized for PCR followed by agarose gel electrophoresis, real-time PCR, immunoblot, and immunofluorescence analysis. Appropriate groups were subjected to a battery of standard neurological tests at regular intervals until 14 days after reperfusion. The increased expression of both TLRs 2 and 4 and their downstream signaling molecules including the pro-inflammatory cytokines was observed even at 1-week after reperfusion. T2sh+T4sh treatment immediately after reperfusion attenuated the post-ischemic inflammation, preserved the motor function, and promoted recovery of the sensory and motor functions. We conclude that the post-ischemic induction of TLRs 2 and 4 persists for at least 7 days after reperfusion, contributes to the severity of acute inflammation, and impedes neurological recovery. Unlike previous studies in TLRs 2 or 4 knockout models, results of this study in a pharmacologically relevant preclinical rodent stroke model have translational significance.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Accidente Cerebrovascular , Animales , Infarto de la Arteria Cerebral Media , Inflamación/etiología , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico
6.
Plast Reconstr Surg ; 145(1): 85e-93e, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31881615

RESUMEN

BACKGROUND: Objective assessment of tissue viability is critical to improve outcomes of cosmetic and reconstructive procedures. A widely used method to predict tissue viability is indocyanine green angiography. The authors present an alternative method that determines the relative proportions of oxyhemoglobin to deoxyhemoglobin through multispectral reflectance imaging. This affordable, hand-held device is noninvasive and may be used in clinic settings. The authors hypothesize that multispectral reflectance imaging is not inferior to indocyanine green angiography in predicting flap necrosis in the murine model. METHODS: Reverse McFarlane skin flaps measuring 10 × 3 cm were raised on 300- to 400-g male Sprague-Dawley rats. Indocyanine green angiography and multispectral reflectance imaging was performed before surgery, immediately after surgery, and 30 minutes after surgery. Clinical outcome images acquired 72 hours after surgery were evaluated by three independent plastic surgeons. Objective data obtained immediately after surgery were compared to postsurgical clinical outcomes to determine which method more accurately predicted flap necrosis. RESULTS: Nine reverse McFarlane skin flaps were evaluated 72 hours after flap elevation. Data analysis demonstrated that the 95 percent confidence intervals for the sensitivity of postoperative multispectral reflectance imaging and indocyanine green angiography imaging to predict 72-hour tissue viability at a fixed specificity of 90 percent for predicting tissue necrosis were 86.3 to 91.0 and 79.1 to 86.9, respectively. CONCLUSIONS: In this experimental animal model, multispectral reflectance imaging does not appear to be inferior to indocyanine green angiography in detecting compromised tissue viability. With the advantages of noninvasiveness, portability, affordability, and lack of disposables, multispectral reflectance imaging has an exciting potential for widespread use in cosmetic and reconstructive procedures.


Asunto(s)
Angiografía/métodos , Procedimientos de Cirugía Plástica/efectos adversos , Complicaciones Posoperatorias/diagnóstico por imagen , Piel/patología , Colgajos Quirúrgicos , Animales , Colorantes/administración & dosificación , Modelos Animales de Enfermedad , Fluorescencia , Humanos , Verde de Indocianina/administración & dosificación , Masculino , Necrosis/diagnóstico por imagen , Necrosis/etiología , Necrosis/patología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/patología , Ratas , Ratas Sprague-Dawley , Procedimientos de Cirugía Plástica/métodos , Piel/irrigación sanguínea , Supervivencia Tisular
7.
Neuromolecular Med ; 21(4): 529-539, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31077035

RESUMEN

Emerging stroke literature suggests that treatment of experimentally induced stroke with stem cells offered post-stroke neuroprotection via exosomes produced by these cells. Treatment with exosomes has great potential to overcome the limitations associated with cell-based therapies. However, in our preliminary studies, we noticed that the exosomes released from human umbilical cord blood-derived mesenchymal stem cells (MSCs) under standard culture conditions did not improve the post-stroke neurological outcome. Because of this apparent discrepancy, we hypothesized that exosome characteristics vary with the conditions of their production. Specifically, we suggest that the exosomes produced from the cocultures of regular and oxygen-glucose-deprived (OGD) MSCs in vitro would represent the exosomes produced from MSCs that are exposed to ischemic brain cells in vivo, and offer similar therapeutic benefits that the cell treatment would provide. We tested the efficacy of therapy with exosomes secreted from human umbilical cord blood (HUCB)-derived MSCs under in vitro hypoxic conditions on post-stroke brain damage and neurological outcome in a rat model of transient focal cerebral ischemia. We performed the TTC staining procedure as well as the neurological tests including the modified neurological severity scores (mNSS), the modified adhesive removal (sticky-tape), and the beam walking tests before ischemia and at regular intervals until 7 days reperfusion. Treatment with exosomes obtained from the cocultures of normal and OGD-induced MSCs reduced the infarct size and ipsilateral hemisphere swelling, preserved the neurological function, and facilitated the recovery of stroke-induced rats. Based on the results, we conclude that the treatment with exosomes secreted from MSCs at appropriate experimental conditions attenuates the post-stroke brain damage and improves the neurological outcome.


Asunto(s)
Daño Encefálico Crónico/prevención & control , Isquemia Encefálica/terapia , Exosomas , Células Madre Mesenquimatosas/metabolismo , Daño por Reperfusión/prevención & control , Animales , Peso Corporal , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/patología , Isquemia Encefálica/complicaciones , Hipoxia de la Célula , Técnicas de Cocultivo , Sangre Fetal/citología , Glucosa/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Masculino , Oxígeno/farmacología , Equilibrio Postural , Desempeño Psicomotor , Ratas , Daño por Reperfusión/etiología , Regulación hacia Arriba
8.
Cell Physiol Biochem ; 52(6): 1280-1291, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31026391

RESUMEN

BACKGROUND/AIMS: Recent studies demonstrated that the treatment with mesenchymal stem cells (MSCs) obtained from the human umbilical cord blood improved survival, reduced brain damage, prevented apoptosis, suppressed inflammatory responses, downregulated the DNA damage-inducing genes, upregulated the DNA repair genes, and facilitated neurological recovery in stroke-induced animals. Emerging stroke literature supports the concept that the exosomes released from MSCs are the primary biological principles underlying the post-stroke neuroprotection offered by MSCs treatment. METHODS: Because the treatment with exosomes has a great potential to overcome the limitations associated with cell-based therapies, we tested the efficacy of exosomes secreted from HUCB-MSCs under standard culture conditions on post-stroke brain damage and neurological outcome in a rat model of ischemic stroke by performing TTC staining as well as the modified neurological severity scores, modified adhesive removal, beam-walking, and accelerating Rotarod performance tests before ischemia and at regular intervals until seven days reperfusion. RESULTS: Exosomes treatment attenuated the infarct size. Treatment with exosomes did not affect the post-stroke survival rate and body weight changes, but exacerbated the somatosensory and motor dysfunction and adversely affected the natural recovery that occurs without any treatment. CONCLUSION: Treatment with exosomes secreted from HUCB-MSCs under standard culture conditions attenuates the ischemic brain damage but does not improve the post-stroke neurological outcome.


Asunto(s)
Encéfalo/patología , Exosomas/trasplante , Células Madre Mesenquimatosas/citología , Accidente Cerebrovascular/terapia , Animales , Encéfalo/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Masculino , Actividad Motora , Ratas Sprague-Dawley , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Resultado del Tratamiento
9.
Stroke Vasc Neurol ; 3(3): 153-159, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30294471

RESUMEN

Background and purpose: Recent reports from our laboratory demonstrated the post-ischaemic expression profile of various matrix metalloproteinases (MMPs) in rats and the detrimental role of MMP-12 in post-stroke brain damage. We hypothesise that the post-stroke dysregulation of MMPs is similar across species and that genetic deletion of MMP-12 would not affect the post-stroke expression of other MMPs. We tested our hypothesis by determining the pre-ischaemic and post-ischaemic expression profile of MMPs in wild-type and MMP-12 knockout mice. Methods: Focal cerebral ischaemia was induced in wild-type and MMP-12 knockout mice by middle cerebral artery occlusion procedure by insertion of a monofilament suture. One hour after ischaemia, reperfusion was initiated by removing the monofilament. One day after reperfusion, ischaemic brain tissues from various groups of mice were collected, and total RNA was isolated and subjected to cDNA synthesis followed by PCR analysis. Results: Although the post-stroke expression profile of MMPs in the ischaemic brain of mice is different from rats, there is a clear species similarity in the expression of MMP-12, which was found to be predominantly upregulated in both species. Further, the post-stroke induction or inhibition of various MMPs in MMP-12 knockout mice is different from their respective expression profile in wild-type mice. Moreover, the brain mRNA expression profile of various MMPs in MMP-12 knockout mice under normal conditions is also different to their expression in wild-type mice. Conclusions: In the ischaemic brain, MMP-12 upregulates several fold higher than any other MMP. Mice derived with the genetic deletion of MMP-12 are constitutive and have altered MMP expression profile both under normal and ischaemic conditions.


Asunto(s)
Eliminación de Gen , Infarto de la Arteria Cerebral Media/enzimología , Metaloproteinasa 12 de la Matriz/deficiencia , ARN Mensajero/metabolismo , Transcriptoma , Animales , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Infarto de la Arteria Cerebral Media/genética , Masculino , Metaloproteinasa 12 de la Matriz/genética , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Ratas , Especificidad de la Especie , Factores de Tiempo
11.
Neuroscience ; 373: 82-91, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337240

RESUMEN

Toll-like receptor 2 (TLR2) and TLR4 belong to a family of highly conserved pattern recognition receptors and are well-known upstream sensors of signaling pathways of innate immunity. TLR2 and TLR4 upregulation is thought to be associated with poor outcome in stroke patients. We currently show that transient focal ischemia in adult rats induces TLR2 and TLR4 expression within hours and shRNA-mediated knockdown of TLR2 and TLR4 alone and in combination decreases the infarct size and swelling. We further show that TLR2 and TLR4 knockdown also prevented the induction of their downstream signaling molecules MyD88, IRAK1, and NFκB p65 as well as the pro-inflammatory cytokines IL-1ß, IL-6, and TNFα. This study thus shows that attenuation of the severity of TLR2- and TLR4-mediated post-stroke inflammation ameliorates ischemic brain damage.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Inflamación/metabolismo , Inflamación/prevención & control , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Animales , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/prevención & control , Isquemia Encefálica/complicaciones , Modelos Animales de Enfermedad , Escherichia coli , Técnicas de Silenciamiento del Gen , Inflamación/etiología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas de Neoplasias/metabolismo , Neuroprotección/fisiología , Proteínas de Transporte Nucleocitoplasmático/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , Distribución Aleatoria , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
12.
Mol Neurobiol ; 55(2): 1405-1409, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28155200

RESUMEN

The role of matrix metalloproteinase-12 (MMP-12) in the pathogenesis of several inflammatory diseases such as chronic obstructive pulmonary disease, emphysema, and asthma is well established. Several new studies and recent reports from our laboratory and others highlighted the detrimental role of MMP-12 in the pathogenesis of several neurological diseases. In this review, we discuss in detail the pathological role of MMP-12 and the possible underlying molecular mechanisms that contribute to disease pathogenesis in the context of central nervous system diseases such as stroke, spinal cord injury, and multiple sclerosis. The available information on the specific MMP-12 inhibitors used in several preclinical and clinical studies is also reviewed. Based on the reported studies to date, MMP-12 suppression could emerge as a promising therapeutic target for several CNS diseases that were discussed in this review.


Asunto(s)
Metaloproteinasa 12 de la Matriz/metabolismo , Esclerosis Múltiple/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Accidente Cerebrovascular/metabolismo , Humanos
13.
Cell Physiol Biochem ; 44(4): 1360-1369, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29186705

RESUMEN

BACKGROUND/AIMS: Stem cell treatment is one of the potential treatment options for ischemic stroke. We recently demonstrated a protective effect of human umbilical cord blood-derived mesenchymal stem cells (HUCB-MSCs) in a rat model of ischemic stroke. The treatment attenuated apoptosis and prevented DNA damage. A collection of published studies, including several from our laboratory, indicated the induction and detrimental role for several matrix metalloproteinases (MMPs) in post-stroke brain injury. We hypothesized that the HUCB-MSCs treatment after focal cerebral ischemia prevents the dysregulation of MMPs and induces the expression of endogenous tissue inhibitors of metalloproteinases (TIMPs) to neutralize the elevated activity of MMPs. METHODS: To test our hypothesis, we administered HUCB-MSCs (0.25 million cells/animal and 1 million cells/animal) intravenously via tail vein to male Sprague-Dawley rats that were subjected to a transient (two-hour) right middle cerebral artery occlusion (MCAO) and one-day reperfusion. Ischemic brain tissues obtained from various groups of rats seven days after reperfusion were subjected to real-time PCR, immunoblot, and immunofluorescence analysis. RESULTS: HUCB-MSCs treatment prevented the induction of MMPs, which were upregulated in ischemia-induced rats that received no treatment. HUCB-MSCs treatment also prevented the induction of TIMPs expression. The extent of prevention of MMPs and TIMPs induction by HUCB-MSCs treatment is similar at both the doses tested. CONCLUSION: Prevention of stroke-induced MMPs upregulation after HUCB-MSCs treatment is not mediated through TIMPs upregulation.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Trasplante de Células Madre Mesenquimatosas , Accidente Cerebrovascular/terapia , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Animales , Modelos Animales de Enfermedad , Sangre Fetal/citología , Masculino , Metaloproteinasas de la Matriz/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Microscopía Fluorescente , Arteria Cerebral Media/lesiones , Ratas , Ratas Sprague-Dawley , Inhibidores Tisulares de Metaloproteinasas/genética , Regulación hacia Arriba/efectos de los fármacos
15.
Stroke ; 46(12): 3523-31, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26534974

RESUMEN

BACKGROUND AND PURPOSE: Matrix metalloproteinases (MMPs) have a central role in compromising the integrity of the blood-brain barrier (BBB). The role of MMP-12 in brain damage after ischemic stroke remains unknown. The main objective of the current study is to investigate the effect of MMP-12 suppression at an early time point before reperfusion on the BBB damage in rats. METHODS: Sprague-Dawley rats were subjected to middle cerebral artery occlusion and reperfusion. MMP-12 shRNA-expressing plasmids formulated as nanoparticles were administered at a dose of 1 mg/kg body weight. The involvement of MMP-12 on BBB damage was assessed by performing various techniques, including Evans blue dye extravasation, 2,3,5-triphenyltetrazolium chloride staining, immunoblot, gelatin zymography, and immunofluorescence analysis. RESULTS: MMP-12 is upregulated ≈31-, 47-, and 66-fold in rats subjected 1-, 2-, or 4-hour ischemia, respectively, followed by 1-day reperfusion. MMP-12 suppression protected the BBB integrity by inhibiting the degradation of tight-junction proteins. Either intravenous or intra-arterial delivery of MMP-12 shRNA-expressing plasmid significantly reduced the percent Evans blue dye extravasation and infarct size. Furthermore, MMP-12 suppression reduced the endogenous levels of other proteases, such as tissue-type plasminogen activator and MMP-9, which are also known to be the key players involved in BBB damage. CONCLUSIONS: These results demonstrate the adverse role of MMP-12 in acute brain damage that occurs after ischemic stroke and, thereby, suggesting that MMP-12 suppression could be a promising therapeutic target for cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica/enzimología , Barrera Hematoencefálica/patología , Isquemia Encefálica/enzimología , Isquemia Encefálica/patología , Metaloproteinasa 12 de la Matriz/biosíntesis , Animales , Encéfalo/enzimología , Encéfalo/patología , Ratas , Ratas Sprague-Dawley
16.
Sci Rep ; 5: 9504, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25955565

RESUMEN

This study highlights the possible pathological role of MMP-12 in the context of ischemic stroke. Male rats were subjected to a two-hour middle cerebral artery occlusion (MCAO) procedure. MMP-12 shRNA expressing plasmid formulation was administered to these rats twenty-four hours after reperfusion. The results showed a predominant upregulation of MMP-12 (approximately 47, 58, 143, and 265 folds on days 1, 3, 5, 7 post-ischemia, respectively) in MCAO subjected rats. MMP-12 expression was localized to neurons, oligodendrocytes and microglia, but not astrocytes. Transcriptional inactivation of MMP-12 significantly reduced the infarct size. The percent infarct size was reduced from 62.87±4.13 to 34.67±5.39 after MMP-12 knockdown compared to untreated MCAO subjected rats. Expression of myelin basic protein was increased, and activity of MMP-9 was reduced in ischemic rat brains after MMP-12 knockdown. Furthermore, a significant reduction in the extent of apoptosis was noticed after MMP-12 knockdown. TNFα expression in the ipsilateral regions of MCAO-subjected rats was reduced after MMP-12 knockdown in addition to the reduced protein expression of apoptotic molecules that are downstream to TNFα signaling. Specific knockdown of MMP-12 after focal cerebral ischemia offers neuroprotection that could be mediated via reduced MMP-9 activation and myelin degradation as well as inhibition of apoptosis.


Asunto(s)
Lesiones Encefálicas/enzimología , Lesiones Encefálicas/genética , Isquemia Encefálica/enzimología , Isquemia Encefálica/genética , Silenciador del Gen , Metaloproteinasa 12 de la Matriz/genética , Transcripción Genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Masculino , Metaloproteinasa 12 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Básica de Mielina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reperfusión , Especificidad por Sustrato/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
17.
Neurochem Res ; 39(8): 1511-21, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24879430

RESUMEN

Evidence suggests that apoptosis contributes significantly to cell death after cerebral ischemia. Our recent studies that utilized human umbilical cord blood-derived mesenchymal stem cells (hUCBSCs) demonstrated the potential of hUCBSCs to inhibit neuronal apoptosis in a rat model of CNS injury. Therefore, we hypothesize that intravenous administration of hUCBSCs after focal cerebral ischemia would reduce brain damage by inhibiting apoptosis and downregulating the upregulated apoptotic pathway molecules. Male Sprague-Dawley rats were obtained and randomly assigned to various groups. After the animals reached a desired weight, they were subjected to a 2 h middle cerebral artery occlusion (MCAO) procedure followed by 7 days of reperfusion. The hUCBSCs were obtained, cultured, and intravenously injected (0.25 × 10(6) cells or 1 × 10(6) cells) via the tail vein to separate groups of animals 24 h post-MCAO procedure. We performed various techniques including PCR microarray, hematoxylin and eosin, and TUNEL staining in addition to immunoblot and immunofluorescence analysis in order to investigate the effect of our treatment on regulation of apoptosis after focal cerebral ischemia. Most of the apoptotic pathway molecules which were upregulated after focal cerebral ischemia were downregulated after hUCBSCs treatment. Further, the staining techniques revealed a prominent reduction in brain damage and the extent of apoptosis at even the lowest dose of hUCBSCs tested in the present study. In conclusion, our treatment with hUCBSCs after cerebral ischemia in the rodent reduces brain damage by inhibiting apoptosis and downregulating the apoptotic pathway molecules.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Apoptosis/fisiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Trasplante de Células Madre/tendencias , Animales , Isquemia Encefálica/patología , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
19.
Lab Anim (NY) ; 42(9): 325-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23965565

RESUMEN

Provision of adequate veterinary care is a required component of animal care and use programs in the United States. Program participants other than veterinarians, including non-medically trained research personnel and technicians, also provide veterinary care to animals, and administrators are responsible for assuring compliance with federal mandates regarding adequate veterinary care. All program participants therefore should understand the regulatory requirements for providing such care. The author provides a training primer on the US regulatory requirements for the provision of veterinary care to research animals. Understanding the legal basis and conditions of a program of veterinary care will help program participants to meet the requirements advanced in the laws and policies.


Asunto(s)
Crianza de Animales Domésticos/legislación & jurisprudencia , Animales de Laboratorio , Regulación Gubernamental , Legislación Veterinaria , Crianza de Animales Domésticos/métodos , Bienestar del Animal , Animales , Estados Unidos , Medicina Veterinaria/métodos
20.
PLoS One ; 8(8): e71822, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23951251

RESUMEN

BACKGROUND: Protein-tyrosine sulfation is a post-translational modification of an unknown number of secreted and membrane proteins mediated by two known Golgi tyrosylprotein sulfotransferases (TPST-1 and TPST-2). We reported that Tpst2-/- mice have mild-moderate primary hypothyroidism, whereas Tpst1-/- mice are euthyroid. While using magnetic resonance imaging (MRI) to look at the thyroid gland we noticed that the salivary glands in Tpst2-/- mice appeared smaller than in wild type mice. This prompted a detailed analysis to compare salivary gland structure and function in wild type, Tpst1-/-, and Tpst2 -/- mice. METHODOLOGY/PRINCIPAL FINDINGS: Quantitative MRI imaging documented that salivary glands in Tpst2-/- females were (≈) 30% smaller than wild type or Tpst1-/- mice and that the granular convoluted tubules in Tpst2-/- submandibular glands were less prominent and were almost completely devoid of exocrine secretory granules compared to glands from wild type or Tpst1-/- mice. In addition, pilocarpine-induced salivary flow and salivary α-amylase activity in Tpst2-/- mice of both sexes was substantially lower than in wild type and Tpst1-/- mice. Anti-sulfotyrosine Western blots of salivary gland extracts and saliva showed no differences between wild type, Tpst1-/-, and Tpst2-/- mice, suggesting that the salivary gland hypofunction is due to factor(s) extrinsic to the salivary glands. Finally, we found that all indicators of hypothyroidism (serum T4, body weight) and salivary gland hypofunction (salivary flow, salivary α-amylase activity, histological changes) were restored to normal or near normal by thyroid hormone supplementation. CONCLUSIONS/SIGNIFICANCE: Our findings conclusively demonstrate that low body weight and salivary gland hypofunction in Tpst2-/- mice is due solely to primary hypothyroidism.


Asunto(s)
Hipotiroidismo/metabolismo , Glándulas Salivales/metabolismo , Sulfotransferasas/metabolismo , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Peso Corporal/fisiología , Suplementos Dietéticos , Femenino , Expresión Génica , Hipotiroidismo/sangre , Hipotiroidismo/genética , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándulas Salivales/patología , Glándulas Salivales/fisiopatología , alfa-Amilasas Salivales/metabolismo , Glándula Submandibular/metabolismo , Glándula Submandibular/patología , Glándula Submandibular/fisiopatología , Sulfotransferasas/genética , Tiroides (USP)/administración & dosificación , Tiroides (USP)/farmacología , Tiroxina/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...