Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Osteoarthritis Cartilage ; 31(7): 934-943, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36898656

RESUMEN

INTRODUCTION: The association between metabolic syndrome (MetS) and osteoarthritis (OA) development has become increasingly recognized. In this context, the exact role of cholesterol and cholesterol-lowering therapies in OA development has remained elusive. Recently, we did not observe beneficial effects of intensive cholesterol-lowering treatments on spontaneous OA development in E3L.CETP mice. We postulated that in the presence of local inflammation caused by a joint lesion, cholesterol-lowering therapies may ameliorate OA pathology. MATERIALS AND METHODS: Female ApoE3∗Leiden.CETP mice were fed a cholesterol-supplemented Western type diet. After 3 weeks, half of the mice received intensive cholesterol-lowering treatment consisting of atorvastatin and the anti-PCSK9 antibody alirocumab. Three weeks after the start of the treatment, OA was induced via intra-articular injections of collagenase. Serum levels of cholesterol and triglycerides were monitored throughout the study. Knee joints were analyzed for synovial inflammation, cartilage degeneration, subchondral bone sclerosis and ectopic bone formation using histology. Inflammatory cytokines were determined in serum and synovial washouts. RESULTS: Cholesterol-lowering treatment strongly reduced serum cholesterol and triglyceride levels. Mice receiving cholesterol-lowering treatment showed a significant reduction in synovial inflammation (P = 0.008, WTD: 95% CI: 1.4- 2.3; WTD + AA: 95% CI: 0.8- 1.5) and synovial lining thickness (WTD: 95% CI: 3.0-4.6, WTD + AA: 95% CI: 2.1-3.2) during early-stage collagenase-induced OA. Serum levels of S100A8/A9, MCP-1 and KC were significantly reduced after cholesterol-lowering treatment (P = 0.0005, 95% CI: -46.0 to -12.0; P = 2.8 × 10-10, 95% CI: -398.3 to -152.1; P = 2.1 × 10-9, -66.8 to -30.4, respectively). However, this reduction did not reduce OA pathology, determined by ectopic bone formation, subchondral bone sclerosis and cartilage damage at end-stage disease. CONCLUSION: This study shows that intensive cholesterol-lowering treatment reduces joint inflammation after induction of collagenase-induced OA, but this did not reduce end stage pathology in female mice.


Asunto(s)
Cartílago Articular , Osteoartritis , Ratones , Femenino , Animales , Esclerosis/patología , Membrana Sinovial/metabolismo , Osteoartritis/inducido químicamente , Osteoartritis/tratamiento farmacológico , Osteoartritis/complicaciones , Inflamación/metabolismo , Colagenasas/toxicidad , Colagenasas/metabolismo , Colesterol/metabolismo , Modelos Animales de Enfermedad , Cartílago Articular/patología
2.
PLoS One ; 18(1): e0281135, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36719872

RESUMEN

OBJECTIVE: To compare two agents that can induce a rat model of temporomandibular joint osteoarthritis (TMJOA) by chemical induction: monosodium iodoacetate (MIA) and collagenase type 2 (Col-2). We wished to ascertain the best agent for assessing drug-delivery systems (DDSs). METHOD: Male Wistar rats underwent intra-articular injection with MIA or Col-2. They were manipulated for 30 days. The head withdrawal threshold (HWT), immunohistological assessment, and positron emission tomography (PET) were used to evaluate the relevance of our models. RESULTS: For both the MIA and Col-2 groups, pain persisted for 30 days after injection. Change in the HWT showed that Col-2 elicited a strong action initially that decreased progressively. MIA had a constant action upon pain behavior. Histology of TMJ tissue from both groups showed progressive degradation of TMJ components. CONCLUSIONS: MIA and Col-2 induced orofacial pain by their local chemical action on TMJs. However, based on a prolonged and greater sustained effect on the pain threshold, persistent histological changes, and imaging results, MIA appeared to be more suitable for creation of a rat model of TMJOA for the study of DDSs.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ácido Yodoacético , Metaloproteinasa 8 de la Matriz , Osteoartritis , Trastornos de la Articulación Temporomandibular , Animales , Masculino , Ratas , Colagenasas/administración & dosificación , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Inyecciones Intraarticulares , Ácido Yodoacético/administración & dosificación , Ácido Yodoacético/toxicidad , Osteoartritis/diagnóstico por imagen , Osteoartritis/tratamiento farmacológico , Osteoartritis/etiología , Osteoartritis/patología , Dolor/inducido químicamente , Dolor/etiología , Ratas Wistar , Tomografía Computarizada por Rayos X , Metaloproteinasa 8 de la Matriz/administración & dosificación , Metaloproteinasa 8 de la Matriz/toxicidad , Artralgia/inducido químicamente , Artralgia/etiología , Trastornos de la Articulación Temporomandibular/diagnóstico por imagen , Trastornos de la Articulación Temporomandibular/tratamiento farmacológico , Trastornos de la Articulación Temporomandibular/etiología , Trastornos de la Articulación Temporomandibular/patología
3.
Molecules ; 26(23)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34885759

RESUMEN

Osteoarthritis is a common multifactorial chronic disease that occurs in articular cartilage, subchondral bone, and periarticular tissue. The pathogenesis of OA is still unclear. To investigate the differences in serum metabolites between OA and the control group, liquid chromatography/mass spectrometry (LC/MS)-based metabolomics was used. To reveal the pathogenesis of OA, 12 SD male rats were randomly divided into control and OA groups using collagenase to induce OA for modeling, and serum was collected 7 days after modeling for testing. The OA group was distinguished from the control group by principal component analysis and orthogonal partial least squares-discriminant analysis, and six biomarkers were finally identified. These biomarkers were metabolized through tryptophan metabolism, glutamate metabolism, nitrogen metabolism, spermidine metabolism, and fatty acid metabolism pathways. The study identified metabolites that may be altered in OA, suggesting a role in OA through relevant metabolic pathways. Metabolomics, as an important tool for studying disease mechanisms, provides useful information for studying the metabolic mechanisms of OA.


Asunto(s)
Biomarcadores/sangre , Cartílago Articular/metabolismo , Metabolómica , Osteoartritis/sangre , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Cromatografía Liquida , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Ácidos Grasos/sangre , Ácido Glutámico/sangre , Humanos , Espectrometría de Masas , Redes y Vías Metabólicas , Metaboloma/genética , Nitrógeno/sangre , Osteoartritis/inducido químicamente , Osteoartritis/genética , Osteoartritis/metabolismo , Ratas , Espermidina/sangre , Triptófano/sangre
4.
Eur J Pharmacol ; 910: 174507, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34536364

RESUMEN

Intracerebral hemorrhage (ICH) is a devastating disease, and there is currently no specific pharmacological treatment that can improve clinical outcomes. Y-2 sublingual tablets, each containing 30 mg edaravone and 6 mg (+)-borneol, is undergoing a phase III clinical trial for treatment of ischemic stroke in China. The purpose of the present study is to investigate the efficacy and potential mechanism of Y-2 in a rat model of collagenase IV injection induced ICH. Sublingual administration of Y-2 at the dose of 1, 3 and 6 mg/kg improved ICH-induced sensorimotor dysfunction, alleviated cell death and histopathological change, restored the hippocampal long-term potentiation (LTP), reduced brain edema and maintained blood-brain barrier (BBB) integrality in ICH rats. Further study demonstrated that Y-2 could reduce inflammatory response and oxidative stress by decreasing the levels of myeloperoxidase (MPO), ionized calcium-binding adaptor protein-1 (Iba-1), inflammatory cytokines and oxidative products, inhibit transcription factor nuclear factor-κB (NF-κB) activation, cyclooxygenase-2 (COX-2) and matrix metallopeptidase 9 (MMP-9) expression in brain tissue around in the core regions of hematoma. Importantly, the protective efficacy of Y-2 from ICH-induced injury was superior to edaravone. In conclusion, Y-2 sublingual tablets might be a promising therapeutic agent for the treatment of ICH.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Canfanos/farmacología , Hemorragia Cerebral/tratamiento farmacológico , Edaravona/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Edema Encefálico/inmunología , Edema Encefálico/patología , Canfanos/uso terapéutico , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Colagenasas/administración & dosificación , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Combinación de Medicamentos , Edaravona/uso terapéutico , Humanos , Masculino , Fármacos Neuroprotectores/uso terapéutico , Ratas
5.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34576050

RESUMEN

Intracerebral hemorrhage (ICH) occurs when brain blood vessels rupture, causing inflammation and cell death. 2-Fucosyllactose (2FL), a human milk oligosaccharide, has potent antiapoptotic and anti-inflammatory effects. The purpose of this study was to examine the protective effect of 2FL in cellular and rodent models of ICH. Hemin was added to a primary rat cortical neuronal and BV2 microglia coculture to simulate ICH in vitro. IBA1 and MAP2 immunoreactivities were used to determine inflammation and neuronal survival. Hemin significantly increased IBA1, while it reduced MAP2 immunoreactivity. 2FL significantly antagonized both responses. The protective effect of 2FL was next examined in a rat ICH model. Intracerebral administration of type VII collagenase reduced open-field locomotor activity. Early post-treatment with 2FL significantly improved locomotor activity. Brain tissues were collected for immunohistochemistry and qRT-PCR analysis. 2FL reduced IBA1 and CD4 immunoreactivity in the lesioned striatum. 2FL downregulated the expression of ER stress markers (PERK and CHOP), while it upregulated M2 macrophage markers (CD206 and TGFß) in the lesioned brain. Taken together, our data support that 2FL has a neuroprotective effect against ICH through the inhibition of neuroinflammation and ER stress. 2FL may have clinical implications for the treatment of ICH.


Asunto(s)
Proteínas de Unión al Calcio/genética , Accidente Cerebrovascular Hemorrágico/tratamiento farmacológico , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Trisacáridos/farmacología , Animales , Línea Celular , Técnicas de Cocultivo , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hemina/toxicidad , Accidente Cerebrovascular Hemorrágico/inducido químicamente , Accidente Cerebrovascular Hemorrágico/genética , Accidente Cerebrovascular Hemorrágico/patología , Humanos , Locomoción/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/patología , Leche Humana/química , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Oligosacáridos/química , Oligosacáridos/farmacología , Ratas , Trisacáridos/química
6.
Inflammopharmacology ; 29(3): 695-704, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34085175

RESUMEN

OBJECTIVE: NLRP3 inflammasome may play a key role in OA pathogenesis. Stromal cell-derived factor-1 (SDF-1) is a homeostatic CXC chemokine. Since the role of SDF-1 in OA has not been explored, this study aimed to examine the effect of SDF-1 on NLRP3 inflammasome and pyroptosis in synoviocytes from OA joints. MATERIALS AND METHODS: Human synovium was obtained from OA patients for isolation of primary synoviocytes and a murine model of collagenase-induced OA was established for testing intra-articular injections of SDF-1. Immunoblotting assays were used to examine the effects and underlying mechanism of action of SDF-1 on NLRP3 inflammasome and synoviocyte pyroptosis in synoviocytes. Inhibitors of AMPK and PI3K-mTOR were utilized to investigate the key signaling pathways involved in SDF-1-mediated OA inflammasome formation and pyroptosis. RESULTS: Synoviocytes from OA joints exhibited significantly higher expression of NLRP3 inflammasome and biomarkers of synoviocyte pyroptosis relative to healthy individuals. This was confirmed in the collagenase-induced OA model, where OA synoviocytes had a significantly lower SDF-1 expression than healthy ones. SDF-1 treatment in synoviocytes of OA patients and collagenase-induced OA led to significant downregulation in the expression of NLRP3 inflammasome and synoviocyte pyroptosis biomarkers. Inhibition of the AMPK signaling pathway significantly suppressed the inhibitory effect of SDF-1 on NLRP3 inflammasome expression of OA synoviocytes. However, blocking the SDF-1-activated PI3K-mTOR signaling pathway could still suppress the expression of NLRP3 inflammasome and synoviocyte pyroptosis biomarkers. CONCLUSIONS: SDF-1 ameliorates NLRP3 inflammasome and pyroptosis in OA synoviocytes through activation of the AMPK signaling pathway. Therefore, SDF-1 may be a novel therapeutic target for OA.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quimiocina CXCL12/administración & dosificación , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Osteoartritis/metabolismo , Piroptosis/efectos de los fármacos , Sinoviocitos/metabolismo , Animales , Células Cultivadas , Colagenasas/toxicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Osteoartritis/inducido químicamente , Osteoartritis/tratamiento farmacológico , Piroptosis/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sinoviocitos/efectos de los fármacos
7.
J Ethnopharmacol ; 274: 114028, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-33775807

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Osteoarthritis (OA), a degenerative joint disease, is characterized by cartilage erosion and matrix degradation. Solanum xanthocarpum Schrad. & Wendl. fruits (SXF) and leaves have long been used as folk remedy in the treatment of pain in rheumatism. AIM OF THE STUDY: This study was aimed to investigate the phytochemical components and protective benefits of SXF on in vitro chondrocytes proliferation, and in vivo suppression of collagenase-induced OA. MATERIALS AND METHODS: Phytochemical components in ethanolic SXF extract were evaluated using gas chromatography-mass spectrometry (GC-MS). Effect of SXF on in vitro cell proliferation of primary chondrocytes was determined by cell proliferation assay and cell cycle analysis by flow cytometry. OA was induced in the right knees of rats through intra-articular injection of collagenase type-II. To evaluate in vivo preventive function of SXF, body weight, blood ALP, histopathological changes in the knee joint, proteoglycan, and collagen content were determined. The mRNA expression of COL-2, MMP-3 and COX-2 genes through qRT-PCR was studied. Antioxidant activities, total phenolics and flavonoid contents of SXF were also examined. RESULTS: GC-MS analysis revealed that SXF constitutes 28 phytochemicals including flavonoids (3-methoxy apigenin, quercetin, luteolin), tannin (quinic acid), terpenes (oleanolic acid, lupeol, psi.psi carotene), phytosterols (campesterol, stigmasterol, ß-sitosterol), and ascorbic acid. In vitro studies demonstrated that SXF enhanced the cell proliferation in a dose-dependent manner and has no cytotoxic effect on primary chondrocytes. In vivo study suggests that SXF protects the cartilage destruction induced by collagenase. The histological study revealed that SXF restored the synthesis of collagen and proteoglycan, vital factors for cartilage restoration, and reduced the arthritic score. An up-regulation in COL-2 expression and suppression of MMP-3 and COX-2 were detected by qRT-PCR analysis. Thus, in vivo study suggests the protective effects of SXF on cartilage destruction induced by collagenase. CONCLUSIONS: Our results imply that SXF benefits and ameliorates OA by enhancing the chondrocytes proliferation and preventing the articular cartilage damage through the restoration of their structural molecules, arthritic score reduction, suppression of MMP-3 and COX-2 expression level and up regulation of COL-2 genes expression. These results suggest that SXF could be a promising alternative treatment candidate for osteoarthritis.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Osteoartritis/tratamiento farmacológico , Extractos Vegetales/farmacología , Sustancias Protectoras/farmacología , Solanum/química , Administración Oral , Fosfatasa Alcalina/sangre , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Peso Corporal/efectos de los fármacos , Cartílago Articular/lesiones , Proliferación Celular/efectos de los fármacos , Colágeno Tipo II/metabolismo , Colagenasas/toxicidad , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Flavonoides/análisis , Depuradores de Radicales Libres/administración & dosificación , Depuradores de Radicales Libres/farmacología , Frutas/química , Indometacina/farmacología , Metaloproteinasa 3 de la Matriz/metabolismo , Osteoartritis/inducido químicamente , Fenoles/análisis , Extractos Vegetales/administración & dosificación , Cultivo Primario de Células , Sustancias Protectoras/administración & dosificación , Proteoglicanos/metabolismo , Ratas Sprague-Dawley
8.
Neuroreport ; 32(3): 228-237, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33470757

RESUMEN

Memantine has demonstrated beneficial effects on several types of brain insults via therapeutic mechanisms mainly related to its activity as a receptor antagonist of N-methyl-d-aspartate. However, the influences of memantine on intracerebral hemorrhage (ICH) remain obscure. This research probed into the neurovascular protective mechanisms of memantine after ICH and its impacts on neuronal nitric oxide synthase (nNOS) ser1412 phosphorylation. ICH model was established by employing intrastriatal collagenase injection in rats. After modeling, rats were then allocated randomly into sham-operated (sham), vehicle-treated (ICH+V), and memantine-administrated (ICH+M) groups. Memantine (20 mg/kg/day) was intraperitoneally administered 30 min after ICH and thenceforth once daily. Rats were dedicated at 0.25, 6, 12, 24 h, 3 and 7 d post-ICH for measurement of corresponding indexes. Behavioral changes, brain edema, levels of nNOS ser1412 phosphorylation, peroxynitrite, matrix metalloproteinase (MMP)-9, NLRP3, IL-1ß and numbers of dying neurons, as well as the cellular localization of gelatinolytic activity, were detected among the groups. Memantine improved the neurologic deficits and mitigated brain water content, levels of MMP-9, NLRP3, IL-1ß and dying neurons. Additionally, treatment with memantine also reduced nNOS ser1412 phosphorylation and peroxynitrite formation compared with the ICH+V group at 24 h after ICH. In situ zymography simultaneously revealed that gelatinase activity was primarily colocalized with vessel walls and neurons. We concluded that memantine ameliorated blood-brain barrier disruption and neurologic dysfunction in an ICH rat model. The underlying mechanism might involve repression of nNOS ser1412 phosphorylation, as well as peroxynitrite-related MMP-9 and NLRP3 inflammasome activation.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Hemorragia Cerebral/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Memantina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa de Tipo I/efectos de los fármacos , Ácido Peroxinitroso/metabolismo , Animales , Edema Encefálico , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Gelatinasas/metabolismo , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosforilación/efectos de los fármacos , Ratas
9.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33055425

RESUMEN

Thalamic pain, a type of central poststroke pain, frequently occurs following ischemia/hemorrhage in the thalamus. Current treatment of this disorder is often ineffective, at least in part due to largely unknown mechanisms that underlie thalamic pain genesis. Here, we report that hemorrhage caused by microinjection of type IV collagenase or autologous whole blood into unilateral ventral posterior lateral nucleus and ventral posterior medial nucleus of the thalamus increased the expression of Fgr, a member of the Src family nonreceptor tyrosine kinases, at both mRNA and protein levels in thalamic microglia. Pharmacological inhibition or genetic knockdown of thalamic Fgr attenuated the hemorrhage-induced thalamic injury on the ipsilateral side and the development and maintenance of mechanical, heat, and cold pain hypersensitivities on the contralateral side. Mechanistically, the increased Fgr participated in hemorrhage-induced microglial activation and subsequent production of TNF-α likely through activation of both NF-κB and ERK1/2 pathways in thalamic microglia. Our findings suggest that Fgr is a key player in thalamic pain and a potential target for the therapeutic management of this disorder.


Asunto(s)
Accidente Cerebrovascular Hemorrágico/genética , Hiperalgesia/genética , Neuralgia/genética , Dimensión del Dolor/métodos , Proteínas Proto-Oncogénicas/genética , Familia-src Quinasas/genética , Animales , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Accidente Cerebrovascular Hemorrágico/inducido químicamente , Accidente Cerebrovascular Hemorrágico/patología , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , FN-kappa B/genética , Neuralgia/inducido químicamente , Neuralgia/patología , Tálamo/efectos de los fármacos , Tálamo/metabolismo , Tálamo/patología
10.
Osteoarthritis Cartilage ; 28(4): 486-491, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32028021

RESUMEN

OBJECTIVES: Pharmacological options for treating osteoarthritis (OA) are limited and alternative treatments are required. Given the clinical data indicating that granulocyte macrophage-colony stimulating factor (GM-CSF) may be a therapeutic target in human OA, we evaluated different treatment regimens with a neutralizing anti-GM-CSF monoclonal antibody (mAb) in an experimental OA model to determine their effectiveness on amelioration of pain and disease. METHODS: The collagenase-induced osteoarthritis (CiOA) model was induced in C57BL/6 mice, followed by different treatment regimens of anti-GM-CSF mAb or isotype control. Anti-CCL17 mAb treatment was also administered continually during the late stage of CiOA. Pain-related behavior (change in weight distribution of hind limbs), and disease (cartilage damage and osteophyte size) were assessed. RESULTS: Blocking GM-CSF only during early synovitis in CiOA prevented pain and disease development. Once OA pain was established, regardless of the treatment regimen, anti-GM-CSF mAb treatment rapidly and efficiently ameliorated it; however, unless the treatment was continued, pain returned and disease progressed. Continual late stage blockade of GM-CSF was able to ameliorate pain (between-group difference: -6.567; 95% confidence interval (CI): -10.12, -3.011) and suppress cartilage damage (P = 0.0317, 95% CI: -1.75, -0.0556). Continual late stage blockade of CCL17 showed similar effects on pain and disease development. CONCLUSIONS: Early and short-term GM-CSF neutralization is effective at preventing CiOA pain and disease development but, once pain is evident, continual GM-CSF blockade is required to prevent pain from returning and to suppress disease progression in mice. These data reinforce the potential benefits of anti-GM-CSF (and anti-CCL17) mAb therapy in OA and should inform further clinical trials.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Cartílago Articular/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Osteoartritis de la Rodilla/patología , Rodilla de Cuadrúpedos/efectos de los fármacos , Membrana Sinovial/efectos de los fármacos , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/patología , Cartílago Articular/patología , Quimiocina CCL17/antagonistas & inhibidores , Colagenasas/toxicidad , Progresión de la Enfermedad , Intervención Médica Temprana , Inyecciones Intraarticulares , Ratones , Osteoartritis de la Rodilla/inducido químicamente , Osteofito/patología , Dimensión del Dolor , Rodilla de Cuadrúpedos/patología , Membrana Sinovial/patología , Sinovitis/patología
11.
Pol J Vet Sci ; 22(4): 743-751, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31867928

RESUMEN

Tendinopathy treatment poses a current challenge for sport medicine due to unique physiology and biomechanics of tendons. The goal of this work was to compare the efficacy of the addition of the radial pressure wave therapy (RPWT) treatment to injection of autologous Adipose Derived Stem Cells (ADSCs) or Platelet Rich Plasma (PRP) in the therapeutic procedure for collagenase induced Achilles tendinopathy in sheep. 14 sheep (aged 5 and 6 years, Polish Mountain Sheep breed, weight 60-70 kg) were injected bacterial collagenase type 1A-S (Clostridium histolyticum, C-5894, Sigma Aldrich, Poznan, Poland) bilaterally to Achilles tendons. Subsequently, the animals were injected with PRP (7 sheep) or ADSCs (7 sheep) to previously induced tendinopathy foci. Left limbs of all the animals were additionally treated with RPWT focused above the tendinopathy origins. Treatment progress was controlled by ultrasound scans, and tendon samples were taken on the 126th day of the experiment. Tendon samples taken from the sheep treated with RPWT+ADSCs showed lower cellularity and the highest number of thick collage fibers. Samples taken from the sheep treated with RPWT+PRP showed an elevated rate of neovascularization. Addition of the RPWT to ADSCs injections in the treatment of induced Achilles tendinopathy in sheep resulted in good quality of the tissue regeneration. Dual therapy with RPWT+PRP injection can lead to neovascularization in the tendon tissue.


Asunto(s)
Células Madre Mesenquimatosas , Plasma Rico en Plaquetas , Enfermedades de las Ovejas/etiología , Tendinopatía/veterinaria , Terapia por Ultrasonido/veterinaria , Animales , Colagenasas/toxicidad , Ovinos , Enfermedades de las Ovejas/terapia , Tendinopatía/inducido químicamente , Tendinopatía/terapia , Terapia por Ultrasonido/efectos adversos
12.
Pol J Vet Sci ; 22(4): 735-742, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31867936

RESUMEN

Novel tendinopathy treatment protocols should be assessed for safety. The goal of this work was to compare differences in selected systemic inflammatory marker concentrations after two treatment protocols for collagenase induced Achilles tendinopathy in sheep. 14 sheep (aged 5 and 6 years, Polish Mountain Sheep breed, weight 60-70kg) were injected with bacterial collagenase type 1A-S (Clostridium histolyticum, C-5894, Sigma Aldrich, Poznan, Poland) bilaterally to Achilles tendons. Subsequently, the animals were injected with Platelet Rich Plasma (7 sheep) or Adipose Derived Stem Cells (7 sheep) to induced tendinopathy foci. Left limbs of all sheep were additionally treated with Radial Pressure Wave Therapy (RPWT) focused above the tendinopathy origins. Treatment progress was controlled by ultrasound scans, and tendon samples were taken on the 126th day of the experiment. Serum Amyloid A (SAA) concentration showed mild elevation before the experiment (2 sheep from group I, 4 sheep from group II) and two days after the intratendinous growth factors injection ( 4 sheep from group I, 3 sheep from group II) combined with RPWT (mean 22,63 mg/L and 53, 6 mg/L respectively). Haptoglobine (Hp) concentration increased from 0 to 0,01 g/L in 2 animals from group I two days after injection. These values declined to 0 during the course of the treatment. Fibrinogen (Fb) concentrations were within reference levels throughout the research, although mild elevation was observed before the treatment course in 6 sheep from group I and 1 sheep from group II. In conclusion, addition of RPWT to growth factors injections in the treatment of yatrogenic Achilles tendinopathy in sheep did not induce systemic inflammatory response.


Asunto(s)
Células Madre Mesenquimatosas , Plasma Rico en Plaquetas , Enfermedades de las Ovejas/etiología , Síndrome de Respuesta Inflamatoria Sistémica/veterinaria , Tendinopatía/veterinaria , Terapia por Ultrasonido/veterinaria , Animales , Colagenasas/toxicidad , Ovinos , Enfermedades de las Ovejas/terapia , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Tendinopatía/inducido químicamente , Tendinopatía/terapia , Terapia por Ultrasonido/efectos adversos
13.
Aging (Albany NY) ; 11(20): 9128-9146, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31644429

RESUMEN

Tissue accumulation of p16INK4a-positive senescent cells is associated with age-related disorders, such as osteoarthritis (OA). These cell-cycle arrested cells affect tissue function through a specific secretory phenotype. The links between OA onset and senescence remain poorly described. Using experimental OA protocol and transgenic Cdkn2a+/luc and Cdkn2aluc/luc mice, we found that the senescence-driving p16INK4a is a marker of the disease, expressed by the synovial tissue, but is also an actor: its somatic deletion partially protects against cartilage degeneration. We test whether by becoming senescent, the mesenchymal stromal/stem cells (MSCs), found in the synovial tissue and sub-chondral bone marrow, can contribute to OA development. We established an in vitro p16INK4a-positive senescence model on human MSCs. Upon senescence induction, their intrinsic stem cell properties are altered. When co-cultured with OA chondrocytes, senescent MSC show also a seno-suppressive properties impairment favoring tissue degeneration. To evaluate in vivo the effects of p16INK4a-senescent MSC on healthy cartilage, we rely on the SAMP8 mouse model of accelerated senescence that develops spontaneous OA. MSCs isolated from these mice expressed p16INK4a. Intra-articular injection in 2-month-old C57BL/6JRj male mice of SAMP8-derived MSCs was sufficient to induce articular cartilage breakdown. Our findings reveal that senescent p16INK4a-positive MSCs contribute to joint alteration.


Asunto(s)
Senescencia Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Osteoartritis/inducido químicamente , Comunicación Paracrina/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Condrocitos/fisiología , Técnicas de Cocultivo , Colagenasas/toxicidad , Etopósido/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/metabolismo , Luciferasas/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos , Ratones Transgénicos
14.
J Biomed Sci ; 26(1): 53, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31307481

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) induces a complex sequence of apoptotic cascades that contribute to secondary neuronal damage. Tropomyosin-related kinase receptor B (TrkB) signaling plays a crucial role in promoting neuronal survival following brain damage. METHODS: The present study investigated the protective effects and underlying mechanisms of TrkB activation by the specific TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF), in a model of collagenase-induced ICH and in neuronal cultures. Mice subjected to collagenase-induced ICH were intraperitoneally injected with either 7,8-DHF or vehicle 10 min after ICH and, subsequently, daily for 3 days. Behavioral studies, brain edema measurement, and histological analysis were conducted. Levels of TrkB signaling-related molecules and apoptosis-related proteins were analyzed by western blots. RESULTS: Treatment with 20 mg/kg 7,8-DHF significantly improved functional recovery and reduced brain damage up to 28 days post-ICH. Reduction in neuronal death, apoptosis, and brain edema were also observed in response to 7,8-DHF treatment at 3 days post-ICH. These changes were accompanied by a significant increase in the phosphorylation of TrkB and Akt (Ser473/Thr308) at 1 and 3 days, but had no effect on Erk 44/42 phosphorylation. 7,8-DHF also enhanced the phosphorylation of Ask-1 Ser967 and FOXO-1, downstream targets of Akt at 1 and 3 days. Moreover, 7,8-DHF increased brain-derived neurotrophic factor levels at 1 day. In primary cultured neurons stimulated with hemin, 7,8-DHF promoted survival and reduced apoptosis. Furthermore, delaying the administration of 7,8-DHF to 3 h post-ICH reduced brain tissue damage and neuronal death. CONCLUSIONS: Our findings demonstrate that the activation of TrkB signaling by 7,8-DHF protects against ICH via the Akt, but not the Erk, pathway. These data provide new insights into the role of TrkB signaling deficit in the pathophysiology of ICH and highlight TrkB/Akt as possible therapeutic targets in this disease.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Flavonas/farmacología , Glicoproteínas de Membrana/agonistas , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Hemorragia Cerebral/inducido químicamente , Colagenasas/toxicidad , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
15.
PLoS One ; 14(5): e0215952, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31042750

RESUMEN

The sulfonylurea 1 transient receptor potential melastatin 4 (Sur1-Trpm4) receptor is selectively expressed after intracerebral hemorrhage (ICH). This upregulation contributes to increases in intracellular sodium. Water follows sodium through aquaporin channels, leading to cytotoxic edema. Even after edema is thought to have resolved, ionic dyshomeostasis persists, as does blood-brain barrier (BBB) damage. Glibenclamide, a hypoglycemic agent that inhibits Sur1-Trpm4, has been shown to reduce BBB damage and edema following infusion of autologous blood into the brain (ICH) as well as after other brain injuries. In order to further assess efficacy, we used the collagenase ICH model in rats to test whether glibenclamide reduces edema, attenuates ion dyshomeostasis, improves BBB damage, and reduces lesion volume. We tested a widely-used glibenclamide dose shown effective in other studies (10 µg/kg loading dose followed by 200 ng/hr for up to 7 days). Early initiation of glibenclamide did not significantly impact edema (72 hours), BBB permeability (72 hours), or lesion volume after ICH (28 days). Recovery from neurological impairments was also not improved by glibenclamide. These results suggest that glibenclamide will not improve outcome in ICH. However, the treatment appeared to be safe as there was no effect on bleeding or other physiological variables.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Colagenasas/metabolismo , Gliburida/uso terapéutico , Hipoglucemiantes/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Glucemia/análisis , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Edema Encefálico/patología , Hemorragia Cerebral/etiología , Hemorragia Cerebral/patología , Colagenasas/toxicidad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Gliburida/farmacología , Hipoglucemiantes/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo , Receptores de Sulfonilureas/antagonistas & inhibidores , Receptores de Sulfonilureas/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/metabolismo , Temperatura
16.
BMC Musculoskelet Disord ; 20(1): 120, 2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30902076

RESUMEN

BACKGROUND: Aging impairs tendon healing and is a potential risk factor for chronic tendinitis. During normal aging, tendons undergo structural and biomechanical degenerative changes, accompanied by a reduction in the number of tenocytes and changes to their properties. However, molecular changes in aged tendons under inflammatory conditions are not well understood. The present study analyzed the molecular changes in collagenase induced acute tendon injury using a senescence-accelerated mouse (SAM) model. METHODS: SAMP6 mice were used as an aging animal model and SAMR1 mice were used as a control to represent a senescence-resistant inbred strain. All the mice used in the study were 40 weeks old. Collagenase I from Clostridium histolyticum (20 µL) was injected percutaneously to the tendon-bone junction of the Achilles tendon. Two weeks after treatment, the Achilles tendons were harvested and stained using Picrosirius Red to determine collagen expression. Real-time PCR was performed to analyze gene expression of IL-6, tenomodulin, type I and type II collagen, MMP-9, TIMP-1, and TIMP-2. RESULTS: Collagenase injection resulted in significantly higher gene expression of IL-6 but significantly lower tenomodulin expression compared with the control in SAMP6 and SAMR1 mice. In SAMP6 mice, gene expression of type III collagen and MMP-9 was significantly higher in the collagenase-injected group compared with the control group. SAMP6 mice also showed lower expression of type I collagen, TIMP-1, and TIMP-2 in the collagenase-injected group compared with the control group. Picrosirius Red staining showed the highest expression of type III collagen in the collagenase-injected SAMP6 group compared with the other groups. CONCLUSIONS: The collagenase-injected SAMP6 group showed higher expression of IL-6, MMP-9, and type III collagen and lower expression of type I collagen, TIMP-1, and TIMP-2, which are known to suppress metalloproteinases. The results indicate that aging may lead to dysfunction of the tendon healing process after acute tendon injury.


Asunto(s)
Envejecimiento/metabolismo , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Traumatismos de los Tendones/inducido químicamente , Traumatismos de los Tendones/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Envejecimiento/patología , Animales , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Traumatismos de los Tendones/genética , Traumatismos de los Tendones/patología
17.
Foot Ankle Surg ; 25(2): 186-192, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29409286

RESUMEN

BACKGROUND: This study investigated the effects of percutaneous soft tissue release (PSTR) performed using a blunt cannula on (1) the inflammatory cells-count, (2) expressions of calcitonin gene-related peptide (CGRP) and (3) substance P (SP) in rabbits with chronic phase of collagenase-induced Achilles tendinopathy. METHODS: Thirty-two adult male New Zealand rabbits were randomly divided into four groups: (1) collagenase and PSTR treatment; (2) collagenase and sham-operated PSTR treatment; (3) vehicle-only injection and PSTR treatment; and (4) vehicle-only injection and sham-operated PSTR treatment. Achilles tendon of adult male rabbits was injected with 10µl of collagenase under ultrasonography localization. After 30 days, PSTR was performed using an 18G beauty cosmetic blunt tip micro cannula needle to release the soft tissue and paratenon above the inflamed Achilles tendon. The treated tendons and spinal cords of L5-S2 were harvested 5days after treatment for histological assessment and immunohistochemical analysis. RESULTS: Histopathological examination revealed that PSTR achieved significant reduction in hypercellularity with pronounced infiltration of immune cells at the site of paratenon in tendons injected with collagenase compared with sham operation (p<0.05). Immunohistochemical analysis also showed marked decrease in expression of CGRP in tendon and SP in dorsal horns after PSTR (p<0.05). CONCLUSIONS: This study showed positive effects in an animal model of chronic tendinopathy, and can be considered a treatment option, but that further research is necessary to determine its role in clinical practice.


Asunto(s)
Tendón Calcáneo , Cánula , Procedimientos Quirúrgicos Mínimamente Invasivos , Procedimientos Ortopédicos , Tendinopatía , Animales , Masculino , Conejos , Tendón Calcáneo/patología , Tendón Calcáneo/cirugía , Enfermedad Crónica , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Nocicepción , Procedimientos Ortopédicos/métodos , Distribución Aleatoria , Tendinopatía/inducido químicamente , Tendinopatía/fisiopatología , Tendinopatía/cirugía , Resultado del Tratamiento , Ultrasonografía
18.
Ann Neurol ; 84(6): 854-872, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30294906

RESUMEN

OBJECTIVES: N-acetylcysteine (NAC) is a clinically approved thiol-containing redox modulatory compound currently in trials for many neurological and psychiatric disorders. Although generically labeled as an "antioxidant," poor understanding of its site(s) of action is a barrier to its use in neurological practice. Here, we examined the efficacy and mechanism of action of NAC in rodent models of hemorrhagic stroke. METHODS: Hemin was used to model ferroptosis and hemorrhagic stroke in cultured neurons. Striatal infusion of collagenase was used to model intracerebral hemorrhage (ICH) in mice and rats. Chemical biology, targeted lipidomics, arachidonate 5-lipoxygenase (ALOX5) knockout mice, and viral-gene transfer were used to gain insight into the pharmacological targets and mechanism of action of NAC. RESULTS: NAC prevented hemin-induced ferroptosis by neutralizing toxic lipids generated by arachidonate-dependent ALOX5 activity. NAC efficacy required increases in glutathione and is correlated with suppression of reactive lipids by glutathione-dependent enzymes such as glutathione S-transferase. Accordingly, its protective effects were mimicked by chemical or molecular lipid peroxidation inhibitors. NAC delivered postinjury reduced neuronal death and improved functional recovery at least 7 days following ICH in mice and can synergize with clinically approved prostaglandin E2 (PGE2 ). INTERPRETATION: NAC is a promising, protective therapy for ICH, which acted to inhibit toxic arachidonic acid products of nuclear ALOX5 that synergized with exogenously delivered protective PGE2 in vitro and in vivo. The findings provide novel insight into a target for NAC, beyond the generic characterization as an antioxidant, resulting in neuroprotection and offer a feasible combinatorial strategy to optimize efficacy and safety in dosing of NAC for treatment of neurological disorders involving ferroptosis such as ICH. Ann Neurol 2018;84:854-872.


Asunto(s)
Acetilcisteína/uso terapéutico , Araquidonato 5-Lipooxigenasa/metabolismo , Proteínas de Transporte de Catión/metabolismo , Dinoprostona/metabolismo , Depuradores de Radicales Libres/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Acetilcisteína/farmacología , Animales , Araquidonato 5-Lipooxigenasa/genética , Proteínas de Transporte de Catión/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Células Cultivadas , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/complicaciones , Colagenasas/toxicidad , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Eicosanoides/metabolismo , Femenino , Depuradores de Radicales Libres/farmacología , Glutatión/metabolismo , Hemina/toxicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Accidente Cerebrovascular/etiología , Resultado del Tratamiento
19.
Int J Mol Sci ; 19(3)2018 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-29534523

RESUMEN

Tendon rupture induces an inflammatory response characterized by release of pro-inflammatory cytokines and impaired tendon performance. This study sought to investigate the therapeutic effects of simvastatin-loaded porous microspheres (SIM/PMSs) on inflamed tenocytes in vitro and collagenase-induced Achilles tendinitis in vivo. The treatment of SIM/PMSs in lipopolysaccharide (LPS)-treated tenocytes reduced the mRNA expressions of pro-inflammatory cytokines (Matrix metalloproteinase-3 (MMP-3), cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)). In addition, the local injection of SIM/PMSs into the tendons of collagenase-induced Achilles tendinitis rat models suppressed pro-inflammatory cytokines (MMP-3, COX-2, IL-6, TNF-α, and MMP-13). This local treatment also upregulated anti-inflammatory cytokines (IL-4, IL-10, and IL-13). Furthermore, treatment with SIM/PMSs also improved the alignment of collagen fibrils and effectively prevented collagen disruption in a dose-dependent manner. Therefore, SIM/PMSs treatment resulted in an incremental increase in the collagen content, stiffness, and tensile strength in tendons. This study suggests that SIM/PMSs have great potential for tendon healing and restoration in Achilles tendinitis.


Asunto(s)
Antiinflamatorios/farmacología , Microesferas , Simvastatina/farmacología , Tendinopatía/tratamiento farmacológico , Tenocitos/efectos de los fármacos , Tendón Calcáneo/patología , Animales , Antiinflamatorios/administración & dosificación , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Colagenasas/toxicidad , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Lipopolisacáridos/toxicidad , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley , Simvastatina/administración & dosificación , Tendinopatía/etiología , Tenocitos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
20.
Amino Acids ; 50(3-4): 439-451, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29256178

RESUMEN

Intracerebral hemorrhage (ICH) initiates a neuroinflammatory cascade that contributes to substantial neuronal damage and neurological deterioration. Taurine, an abundant amino acid in the nervous system, is reported to reduce inflammatory injury in various central nervous system diseases, but its role and the possible underlying mechanisms in the pathology following ICH remains unclear. This study was designed to evaluate the effect of taurine supplementation on neurological deficits, acute inflammatory responses and white matter injury in a model of ICH in rats. Adult male Sprague-Dawley (SD) rats subjected to collagenase-induced ICH injury were injected intravenously with different concentrations of taurine or vehicle 10 min after ICH and subsequently daily for 3 days. Behavioral studies, brain water content, and assessments of hemorrhagic lesion volume were quantified at day 1 and day 3 post-ICH. Neuronal damage, peri-hematomal inflammatory responses, and white matter injury were determined at 24 h, meanwhile, the content of hydrogen sulfide (H2S) along with the expression of cystathionine-ß-synthase (CBS) and P2X7 receptor (P2X7R) in peri-hematomal tissues was analyzed to investigate the possible anti-inflammatory mechanism of taurine. Treatment with a high dosage of taurine (50 mg/kg) significantly attenuated functional deficits and reduced brain edema and hemorrhagic lesion volume after ICH. Taurine administration also resulted in significant amelioration of neuronal damage and white matter injury. These changes were associated with marked reductions in neutrophil infiltration, glial activation, and expression levels of inflammatory mediators. Moreover, the anti-inflammatory effect of taurine was accompanied by increased H2S content, enhanced CBS expression, and less expression of P2X7R. Our study demonstrated that the high dosage of taurine supplementation effectively mitigated the severity of pathological inflammation and white matter injury after ICH, and the mechanism may be related to upregulation of H2S content and reduced P2X7R expression.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Taurina/administración & dosificación , Sustancia Blanca/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/metabolismo , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Taurina/metabolismo , Sustancia Blanca/lesiones , Sustancia Blanca/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...