Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Heliyon ; 10(9): e30419, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38765173

RESUMEN

Objective: To develop a novel strategy for identifying acquired demyelination in diabetic distal symmetrical polyneuropathy (DSP). Background: Motor nerve conduction velocity (CV) slowing in diabetic DSP exceeds expectations for pure axonal loss thus implicating superimposed acquired demyelination. Methods: After establishing demyelination confidence intervals by regression analysis of nerve conduction data from chronic inflammatory demyelinating polyneuropathy (CIDP), we prospectively studied CV slowing in 90 diabetic DSP patients with and without at least one motor nerve exhibiting CV slowing (groups A and B) into the demyelination range by American Academy of Neurology (AAN) criteria respectively and 95 amyotrophic lateral sclerosis (ALS) patients. Simultaneously, secretory phospholipase A2 (sPLA2) activity was assessed in both diabetic groups and 46 healthy controls. Results: No ALS patient exhibited CV slowing in more than two motor nerves based on AAN criteria or the confidence intervals. Group A demonstrated a significantly higher percentage of patients as compared to group B fulfilling the above criteria, with an additional criterion of at least one motor nerve exhibiting CV slowing in the demyelinating range and a corresponding F response in the demyelinating range by AAN criteria (70.3 % vs. 1.9 %; p < 0.0001). Urine sPLA2 activity was increased significantly in diabetic groups as compared to healthy controls (942.9 ± 978.0 vs. 591.6 ± 390.2 pmol/min/ml, p < 0.05), and in group A compared to Group B (1328.3 ± 1274.2 vs. 673.8 ± 576.9 pmol/min/ml, p < 0.01). More patients with elevated sPLA2 activity and more than 2 motor nerves with CV slowing in the AAN or the confidence intervals were identified in group A as compared to group B (35.1 % vs. 5.7 %, p < 0.001). Furthermore, 13.5 % of patients in diabetic DSP Group A, and no patients in diabetic DSP Group B, fulfilled an additional criterion of more than one motor nerve with CV slowing into the demyelinating range with its corresponding F response into the demyelinating range by AAN criteria. Conclusion: A combination of regression analysis of electrodiagnostic data and a urine biological marker of systemic inflammation identifies a subgroup of diabetic DSP with superimposed acquired demyelination that may respond favorably to immunomodulatory therapy.

2.
Rev Neurosci ; 35(1): 85-97, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-37525497

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder. Mutations in C9orf72 and the resulting hexanucleotide repeat (GGGGCC) expansion (HRE) has been identified as a major cause of familial ALS, accounting for about 40 % of familial and 6 % of sporadic cases of ALS in Western patients. The pathological outcomes of HRE expansion in ALS have been recognized as the results of two mechanisms that include both the toxic gain-of-function and loss-of-function of C9ORF72. The gain of toxicity results from RNA and dipeptide repeats (DPRs). The HRE can be bidirectionally transcribed into RNA foci, which can bind to and disrupt RNA splicing, transport, and translation. The DPRs that include poly-glycine-alanine, poly-glycine-proline, poly-glycine- arginine, poly-proline-alanine, and poly-proline-arginine can induce toxicity by direct binding and sequestrating other proteins to interfere rRNA synthesis, ribosome biogenesis, translation, and nucleocytoplasmic transport. The C9ORF72 functions through binding to its partners-Smith-Magenis chromosome regions 8 (SMCR8) and WD repeat-containing protein (WDR41). Loss of C9ORF72 function results in impairment of autophagy, deregulation of autoimmunity, increased stress, and disruption of nucleocytoplasmic transport. Further insight into the mechanism in C9ORF72 HRE pathogenesis will facilitate identifying novel and effective therapeutic targets for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Humanos , Esclerosis Amiotrófica Lateral/patología , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Proteínas/genética , Proteínas/metabolismo , Dipéptidos/genética , Dipéptidos/metabolismo , ARN , Arginina , Alanina , Glicina , Prolina
3.
Heliyon ; 9(8): e18400, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37520962

RESUMEN

Objective: Since motor nerve conduction slowing can occur due to loss of large axons, we investigate the conduction slowing profile in amyotrophic lateral sclerosis (ALS) and identify the limits beyond which the diagnosis of exclusive axonal loss is unlikely. Methods: First, using linear regression analysis, we established the range of motor conduction slowing in 76 chronic inflammatory demyelinating polyneuropathy (CIDP) patients. Demyelinating range confidence intervals were defined by assessing conduction velocity (CV), distal latency (DML), and F-wave latency (F) in relation to distal compound muscle action potential (CMAP) amplitude of median, ulnar, fibular, and tibial nerves. Results were subsequently validated in 38 additional CIDP patients. Then, the newly established demyelination confidence intervals were used to investigate the profile of conduction slowing in 95 ALS patients. Results: CV slowing, prolonged DML, and abnormal F were observed in 22.2%, 19.6%, and 47.1% of the studied nerves respectively in ALS patients. When slowing occurred, it affected more than one segment of the motor nerve, suggesting that CMAP amplitude dependent conduction slowing caused by an exclusive loss of large axons is the main mechanism of slowing. No ALS patient had more than 2 nerves with CV slowing in the confidence interval defined by the regression equations or the American Academy of Neurology (AAN) research criteria for CIDP diagnosis. Conclusions: The presence of more than two motor nerves with CV slowing in the demyelinating range defined by the regression analysis or AAN criteria in ALS patients suggests the contribution of acquired demyelination or other additional mechanisms exist in the electrodiagnostic profile of ALS.

4.
Curr Med Chem ; 29(12): 2051-2074, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33970839

RESUMEN

The new coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can present neurological symptoms and induce neurological complications. The involvement in both the central and peripheral nervous systems in COVID-19 patients has been associated with direct invasion of the virus and the induction of cytokine storm. This review discussed the pathways for the virus invasion into the nervous system and characterized the SARS-CoV-2 induced cytokine storm. In addition, the mechanisms underlying the immune responses and cytokine storm induction after SARS-CoV-2 infection were also discussed. Although some neurological symptoms are mild and disappear after recovery from infection, some severe neurological complications contribute to the mortality of COVID-19 patients. Therefore, the insight into the cause of SARS-CoV-2 induced cytokine storm in context with neurological complications will formulate the novel management of the disease and also further identify new therapeutic targets for COVID-19.


Asunto(s)
COVID-19 , Enfermedades del Sistema Nervioso , COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Humanos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , SARS-CoV-2
5.
Cytokine ; 138: 155404, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33360025

RESUMEN

The new coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can trigger a hyperinflammatory state characterized by elevated cytokine levels known as hypercytokinemia or cytokine storm, observed most often in severe patients. Though COVID-19 is known to be a primarily respiratory disease, neurological complications affecting both the central and peripheral nervous systems have also been reported. This review discusses potential routes of SARS-CoV-2 neuroinvasion and pathogenesis, summarizes reported neurological sequelae of COVID-19, and examines how aberrant cytokine levels may precipitate these complications. Clarification of the pathogenic mechanisms of SARS-CoV-2 is needed to encourage prompt diagnosis and optimized care. In particular, identifying the presence of cytokine storm in patients with neurological COVID-19 manifestations will facilitate avenues for treatment. Future investigations into aberrant cytokine levels in COVID-19 patients with neurological symptoms as well as the efficacy of cytokine storm-targeting treatments will be critical in elucidating the pathogenic mechanisms and effective treatments of COVID-19.


Asunto(s)
COVID-19/patología , Trastornos Cerebrovasculares/patología , Síndrome de Liberación de Citoquinas/patología , Citocinas/sangre , Enfermedades del Sistema Nervioso/patología , COVID-19/terapia , Sistema Nervioso Central/patología , Trastornos Cerebrovasculares/virología , Síndrome de Liberación de Citoquinas/terapia , Humanos , Enfermedades del Sistema Nervioso/virología , Sistema Nervioso Periférico/patología , SARS-CoV-2
6.
J Neuroinflammation ; 14(1): 137, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28720149

RESUMEN

BACKGROUND: Heparanase, a mammalian endo-ß-D-glucoronidase that specifically degrades heparan sulfate, has been implicated in inflammation and ischemic stroke. However, the role of heparanase in neuroinflammatory response in subarachnoid hemorrhage (SAH) has not yet been investigated. This study was designed to examine the association between heparanase expression and neuroinflammation during subarachnoid hemorrhage. METHODS: Rats were subjected to SAH by endovascular perforation, and the expression of heparanase was determined by Western blot analysis and immunofluorescence in the ipsilateral brain cortex at 24 h post-SAH. Pial venule leukocyte trafficking was monitored by using intravital microscopy through cranial window. RESULTS: Our results indicated that, compared to their sham-surgical controls, the rats subjected to SAH showed marked elevation of heparanase expression in the ipsilateral brain cortex. The SAH-induced elevation of heparanase was accompanied by increased leukocyte trafficking in pial venules and significant neurological deficiency. Intracerebroventricular application of a selective heparanase inhibitor, OGT2115, which was initiated at 3 h after SAH, significantly suppressed the leukocyte trafficking and improved the neurological function. CONCLUSIONS: Our findings indicate that heparanase plays an important role in mediating the neuroinflammatory response after SAH and contributes to SAH-related neurological deficits and early brain injury following SAH.


Asunto(s)
Glucuronidasa/biosíntesis , Hemorragia Subaracnoidea/enzimología , Hemorragia Subaracnoidea/patología , Animales , Inflamación/enzimología , Inflamación/patología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
7.
J Vasc Res ; 53(5-6): 349-357, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997923

RESUMEN

High-mobility group box 1 protein (HMGB1) has been implicated in inflammatory responses, and is also associated with cerebral vasospasm after subarachnoid hemorrhage (SAH). However, there are no direct evident links between HMGB1 and cerebral vasospasm. We therefore investigated the effects of HMGB1 on pial arteriole reactivity following SAH in rats. We initially found that SAH induced a significant decrease in pial arteriole dilating responses to sciatic nerve stimulation (SNS), hypercapnia (CO2), and the topical suffusion of acetylcholine (ACh), adenosine (ADO), and s-nitroso-N-acetylpenicillamine (SNAP) over a 7-day period after SAH. The percent change of arteriolar diameter was decreased to the lowest point at 48 h after SAH, in response to dilating stimuli (i.e., it decreased from 41.0 ± 19.0% in the sham group to 11.00 ± 0.70% after SNS) (n = 5, p < 0.01). HMGB1 infusion in the lateral ventricle in normal rats for 48 h did not change the pial arteriole dilating response. In addition, inhibitors of HMGB1-receptor for advanced glycation end-product or HMGB1-toll-like receptor 2/4 interaction, or the HMBG1 antagonist did not improve pial arteriole reactivity 48 h after SAH. These findings suggest that HMGB1 may not be a major player in cerebral vascular dilating dysfunction after SAH.


Asunto(s)
Arteriolas/metabolismo , Proteína HMGB1/metabolismo , Piamadre/irrigación sanguínea , Hemorragia Subaracnoidea/metabolismo , Vasodilatación , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/farmacología , Hipercapnia/metabolismo , Hipercapnia/fisiopatología , Masculino , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Nervio Ciático/fisiopatología , Transducción de Señal , Hemorragia Subaracnoidea/fisiopatología , Factores de Tiempo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
8.
Rev Neurosci ; 27(7): 745-759, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27442365

RESUMEN

Subarachnoid hemorrhage (SAH) is a devastating disease with high mortality and mobility, the novel therapeutic strategies of which are essentially required. The calcium binding protein S100B has emerged as a brain injury biomarker that is implicated in pathogenic process of SAH. S100B is mainly expressed in astrocytes of the central nervous system and functions through initiating intracellular signaling or via interacting with cell surface receptor, such as the receptor of advanced glycation end products. The biological roles of S100B in neurons have been closely associated with its concentrations, resulting in either neuroprotection or neurotoxicity. The levels of S100B in the blood have been suggested as a biomarker to predict the progress or the prognosis of SAH. The role of S100B in the development of cerebral vasospasm and brain damage may result from the induction of oxidative stress and neuroinflammation after SAH. To get further insight into mechanisms underlying the role of S100B in SAH based on this review might help us to find novel therapeutic targets for SAH.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Productos Finales de Glicación Avanzada/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Hemorragia Subaracnoidea/fisiopatología , Vasoespasmo Intracraneal/fisiopatología , Animales , Lesiones Encefálicas/complicaciones , Humanos , Estrés Oxidativo/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100/líquido cefalorraquídeo
9.
Brain Res ; 1646: 193-198, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27265419

RESUMEN

Recent studies have shown that microglia affects the fate of neural stem cells in response to ionizing radiation, which suggests a role for microglia in radiation-induced degenerative outcomes. We therefore investigated the effects of γ-irradiation on cell survival, proliferation, and activation of microglia and explored associated mechanisms. Specifically, we evaluated cellular and molecular changes associated with exposure of human microglial cells (CHME5) to low and high doses of acute cesium-137 γ rays. Twenty-four hours after irradiation, cell cycle analyses revealed dose-dependent decreases in the fraction of cells in S and G2/M phase, which correlated with significant oxidative stress. By one week after irradiation, 20-30% of the cells exposed to high doses of γ rays underwent apoptosis, which correlated with significant concomitant decrease in metabolic activity as assessed by the MTT assay, and microglial activation as judged by both morphological changes and increased expression of Glut-5 and CR43. These changes were associated with increases in the mRNA levels for IL-1α, IL-10 and TNFα. Together, the results show that human CHME5 microglia are relatively resistant to low and moderate doses of γ rays, but are sensitive to acute high doses, and that CHME5 cells are a useful tool for in vitro study of human microglia.


Asunto(s)
Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Rayos gamma , Microglía/efectos de la radiación , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de la radiación , Células Cultivadas , Citocinas/metabolismo , Citocinas/efectos de la radiación , Humanos , Microglía/metabolismo , Estrés Oxidativo/efectos de la radiación
10.
Curr Med Chem ; 23(15): 1571-96, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27048377

RESUMEN

The calcium binding protein S100B has attracted great attention as a biomarker for a variety of diseases. S100B is mainly expressed in glial cells and functions through intracellular and extracellular signaling pathways. The biological roles of S100B have been closely associated with its concentrations and its physiological states. The released S100B can bind to the receptor of advanced glycation end products and induce the initiation of multiple cell signaling transductions. The regulation of S100B bioactivities has been suggested through phosphoinositide 3 kinase/Akt, p53, mitogen-activated protein kinases, transcriptional factors including nuclear factor-kappaB, and cyclic adenosine monophosphate. The levels of S100B in the blood may function to predict the progress or the prognosis of many kinds of diseases, such as cerebrovascular diseases, neurodegenerative diseases, motor neuron diseases, traumatic brain injury, schizophrenia, depression, diabetes mellitus, myocardial infarction, cancer, and infectious diseases. Given that the activity of S100B has been implicated in the pathological process of these diseases, S100B should not be simply regarded as a biomarker, it may also function as therapeutic target for these diseases. Further elucidation of the roles of S100B may formulate innovative therapeutic strategies for multiple diseases.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Enfermedad , Subunidad beta de la Proteína de Unión al Calcio S100/antagonistas & inhibidores , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Humanos , Subunidad beta de la Proteína de Unión al Calcio S100/análisis , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo
11.
Drug Discov Today ; 21(8): 1222-31, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27086010

RESUMEN

Proline-rich Akt substrate 40kDa (PRAS40) bridges cell signaling between protein kinase B (Akt) and the mammalian target of rapamycin complex 1 (mTORC1). Both Akt and mTORC1 can phosphorylate PRAS40. As a negative regulator of mTORC1, PRAS40 prevents the binding of mTOR to its substrates. The phosphorylation of PRAS40 results in its dissociation from mTORC1 and enhanced mTOR activation. PRAS40 in conjunction with mTORC1 has been closely associated with programmed cell death and is implicated in diabetes mellitus (DM), cardiovascular diseases, cancer, and neurological diseases. Thus, targeting PRAS40 might hold great promise for innovative therapeutic strategies for these diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus/metabolismo , Neoplasias/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Animales , Apoptosis , Autofagia , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Estrés Oxidativo
12.
Brain Res ; 1634: 171-178, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26773687

RESUMEN

S100B is an astrocyte-derived protein that can act through the receptor for advanced glycation endproducts (RAGE) to mediate either "trophic" or "toxic" responses. Its levels increase in many neurological conditions with associated microvascular dysregulation, such as subarachnoid hemorrhage (SAH) and traumatic brain injury. The role of S100B in the pathogenesis of microvasculopathy has not been addressed. This study was designed to examine whether S100B alters pial arteriolar vasodilating function. Rats were randomized to receive (1) artificial cerebrospinal fluid (aCSF), (2) exogenous S100B, and (3) exogenous S100B+the decoy soluble RAGE (sRAGE). S100B was infused intracerebroventricularly (icv) using an osmotic pump and its levels in the CSF were adjusted to achieve a concentration similar to what we observed in SAH. After 48 h of continuous icv infusion, a cranial window/intravital microscopy was applied to animals for evaluation of pial arteriolar dilating responses to sciatic nerve stimulation (SNS), hypercapnia, and topical suffusion of vasodilators including acetylcholine (ACh), s-nitroso-N-acetyl penicillamine (SNAP), or adenosine (ADO). Pial arteriolar dilating responses were calculated as the percentage change of arteriolar diameter in relation to baseline. The continuous S100B infusion for 48 h was associated with reduced responses to the neuronal-dependent vasodilator SNS (p<0.05) and the endothelial-dependent vasodilator ACh (p<0.05), compared to controls. The inhibitory effects of S100B were prevented by sRAGE. On the other hand, S100B did not alter the responses elicited by vascular smooth muscle cell-dependent vasodilators, namely hypercapnia, SNAP, or ADO. These findings indicate that S100B regulates neuronal and endothelial dependent cerebral arteriolar dilation and suggest that this phenomenon is mediated through RAGE-associated pathways.


Asunto(s)
Piamadre/irrigación sanguínea , Piamadre/fisiología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100/administración & dosificación , Subunidad beta de la Proteína de Unión al Calcio S100/fisiología , Acetilcolina/administración & dosificación , Adenosina/administración & dosificación , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Estimulación Eléctrica , Hipercapnia/metabolismo , Infusiones Intraventriculares , Masculino , Piamadre/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina/administración & dosificación , Subunidad beta de la Proteína de Unión al Calcio S100/líquido cefalorraquídeo , Nervio Ciático/fisiología , Transducción de Señal/efectos de los fármacos , Vasodilatadores/administración & dosificación
13.
Curr Drug Targets ; 16(10): 1107-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25850623

RESUMEN

Significant progress in the research of mammalian target of rapamycin (mTOR) in recent years, has greatly enhanced our understanding of the role and cellular pathways through which mTOR control cellular processes, such as translational initiation, actin organization, cell proliferation, and cell survival. mTOR is activated by phosphorylation and functions mainly through mTOR complex 1 or mTOR complex 2. mTORC1 is activated through tuberous sclerosis complex 1/2 dependent and independent mechanisms following the stimulation by growth factors, nutrient, amino acids, and other signaling pathways. The activity of mTOR is closely associated with cell proliferation and differentiation, apoptosis, and autophagy. Activation of mTOR prevents the induction of both apoptosis and autophagy through regulating its multiple targets. Given that the activity of mTOR has been involved in the pathogenesis of neurodegenerative disorders, cardiovascular abnormalities, metabolic diseases, renal transplantation, autoimmune abnormalities, and cancer, manipulating mTOR activation may represent as an innovative therapeutic strategy for these diseases. Yet, the role of mTOR in the body is complicated and therefore, its activity needs to be tightly regulated to achieve beneficial outcome in a specific pathological condition.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/efectos de los fármacos , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/terapia , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus/patología , Diabetes Mellitus/terapia , Humanos , Neoplasias/patología , Neoplasias/terapia , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Fosforilación , Serina-Treonina Quinasas TOR/genética
14.
Cardiovasc Pathol ; 24(3): 179-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25586361

RESUMEN

BACKGROUND: As Wnt/ß-catenin/glycogen synthase kinase 3ß (GSK3ß) signaling has been implicated in myocardial injury and diabetic cardiomyopathy (DCM) is a major part of diabetic cardiovascular complications, we therefore investigated the alterations of Wnt/ß-catenin/GSK3ß signaling during the development of DCM. METHODS: The rat model of diabetes mellitus (DM) was established using a single intraperitoneal injection of streptozotocin (STZ, 60 mg/kg). The alterations of Wnt/ß-catenin/GSK3ß signaling were determined 4, 8, and 12 weeks following DM using Western blotting, immunohistochemistry, and quantitative real-time reverse transcriptase polymerase chain reaction. Cardiac pathology changes were evaluated using hematoxylin and eosin, Masson trichromatic, and terminal dUTP nick-end labeling staining. RESULTS: Histological analyses revealed that DM induced significant myocardial injury and progressive cardiomyocyte apoptosis. The protein and mRNA levels of Wnt2, ß-catenin, and c-Myc were progressively increased 4, 8, and 12 weeks following DM. The expression of T-cell factor 4 and phosphorylated of GSK3ß on Ser9 were progressively increased. However, the expression of the endogenous Wnt inhibitor Dickkopf-1 was increased after STZ injection and then decreased as DCM developed. CONCLUSION: Wnt/ß-catenin/GSK3ß signaling pathway is activated in the development of DCM. Further investigation into the role of Wnt signaling during DCM will functionally find novel therapeutic target for DCM.


Asunto(s)
Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Apoptosis/fisiología , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Oncol Res ; 20(8): 369-76, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23924857

RESUMEN

Oldhamianoside II is a new triterpenoid saponin that was isolated from the roots of Gypsophila oldhamiana. The present study aims to investigate the potential inhibitory activity of oldhamianoside II on tumor growth using an S180 tumor implantation mouse model. Oldhamianoside II at doses of 5.0 and 10.0 mg/kg was given with intraperitoneal injection for 10 days following subcutaneous inoculation of S180 tumor cells in anterior flank of mice. The tumor growth, the cell apoptosis, the microvessel density (MVD) in S180 tumors, the tumor cell viability, the tubular formation in vitro, and migration of tumor cells were examined. The expression of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and cyclooxygenase-2 (COX-2) was determined to analyze the associated mechanisms. The results showed that oldhamianoside II potently inhibited tumor cell viability in vitro. In addition, oldhamianoside II delayed tumor growth in anterior flank, induced S180 cell apoptosis, and reduced the MVD. Oldhamianoside II was also demonstrated to decrease the number of tubular structure and vessel formation in HUVEC cultures and chick embryo chorioallantoic membrane (CAM) model, respectively. Further study indicated that oldhamianoside II reduced the expression of VEGF, bFGF, and COX-2 in tumor sections. Moreover, oldhamianoside II inhibited the activity of migration and penetration to Matrigel of SGC7901 tumor cells in scratch wound and transwell chamber. In conclusion, our work defines oldhamianoside II, a new triterpenoid saponin, as a novel compound that can effectively inhibit S180 tumor growth, induce tumor cell apoptosis, prevent tumor angiogenesis, and inhibit cancer cell migration, suggesting that oldhamianoside II is a potential drug candidate for the treatment of cancer and for the prevention of metastasis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Saponinas/farmacología , Triterpenos/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ratones , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
17.
Cell Signal ; 25(7): 1598-607, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23563259

RESUMEN

Given the current limitation of therapeutic approach for ischemic stroke, a leading cause of disability and mortality in the developed countries, to develop new therapeutic strategies for this devastating disease is urgently necessary. As a serine/threonine kinase, mammalian target of rapamycin (mTOR) activation can mediate broad biological activities that include protein synthesis, cytoskeleton organization, and cell survival. mTOR functions through mTORC1 and mTORC2 complexes and their multiple downstream substrates, such as eukaryotic initiation factor 4E-binding protein 1, p70 ribosomal S6 kinase, sterol regulatory element-binding protein 1, hypoxia inducible factor-1, and signal transducer and activator transcription 3, Yin Ying 1, Akt, protein kinase c-alpha, Rho GTPase, serum-and gucocorticoid-induced protein kinase 1, etc. Specially, the role of mTOR in the central nervous system has been attracting considerable attention. Based on the ability of mTOR to prevent neuronal apoptosis, inhibit autophagic cell death, promote neurogenesis, and improve angiogenesis, mTOR may acquire the capability of limiting the ischemic neuronal death and promoting the neurological recovery. Consequently, to regulate the activity of mTOR holds a potential as a novel therapeutic strategy for ischemic stroke.


Asunto(s)
Infarto Encefálico/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/fisiopatología , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Neurogénesis , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
18.
Expert Opin Ther Targets ; 17(6): 707-20, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23510463

RESUMEN

INTRODUCTION: Since erythropoietin (EPO) and EPO receptor (EPOR) are expressed in the central nervous system (CNS) beyond hematopoietic system, EPO illustrates a robust biological function in maintaining neuronal survival and regulating neurogenesis and may play a crucial role in neurodegenerative diseases. AREAS COVERED: EPO is capable of modulating multiple cellular signal transduction pathways to promote neuronal survival and enhance the proliferation and differentiation of neuronal progenitor cells. Initially, EPO binds to EPOR to activate the Janus-tyrosine kinase 2 (Jak2) protein followed by modulation of protein kinase B (Akt), mammalian target of rapamycin, signal transducer and activators of transcription 5, mitogen-activated protein kinases, protein tyrosine phosphatases, Wnt1 and nuclear factor κB. As a result, EPO may actively prevent the progression of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis and motor neuron diseases. EXPERT OPINION: Novel knowledge of the cell signaling pathways regulated by EPO in the CNS will allow us to establish the foundation for the development of therapeutic strategies against neurodegenerative diseases. Further investigation of the role of EPO in neurodegenerative diseases can not only formulate EPO as a therapeutic candidate, but also further identify novel therapeutic targets for these disorders.


Asunto(s)
Diseño de Fármacos , Eritropoyetina/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Receptores de Eritropoyetina/metabolismo , Animales , Enfermedad Crónica , Humanos , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/fisiopatología , Neurogénesis/fisiología , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Curr Neurovasc Res ; 10(1): 29-38, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23244622

RESUMEN

More than 110 million individuals will suffer from cognitive loss worldwide by the year 2050 with a majority of individuals presenting with Alzheimer's disease (AD). Yet, successful treatments for etiologies that involve ß.-amyloid (Aß.) toxicity in AD remain elusive and await novel avenues for drug development. Here we show that Wnt1 inducible signaling pathway protein 1 (WISP1/CCN4) controls the post-translational phosphorylation of Akt1, p70S6K, and AMP activated protein kinase (AMPK) to the extent that tuberous sclerosis complex 2 (TSC2) (Ser1387) phosphorylation, a target of AMPK, is decreased and TSC2 (Thr1462) phosphorylation, a target of Akt1, is increased. The ability of WISP1 to limit TSC2 activity allows WISP1 to increase the activity of p70S6K, since gene silencing of TSC2 further enhances WISP1 phosphorylation of p70S6K. However, a minimal level of TSC2 activity is necessary to modulate WISP1 cytoprotection that may require modulation of mTOR activity, since gene knockdown of TSC2 impairs the ability of WISP1 to protect microglia against apoptotic membrane phosphatidylserine (PS) exposure, nuclear DNA degradation, mitochondrial membrane depolarization, and cytochrome c release during Aß. exposure.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/fisiología , Proteínas CCN de Señalización Intercelular/metabolismo , Citoprotección/fisiología , Microglía/metabolismo , Fragmentos de Péptidos/toxicidad , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Citocromos c/metabolismo , Citoprotección/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Microglía/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína 2 del Complejo de la Esclerosis Tuberosa
20.
Expert Opin Drug Discov ; 8(1): 35-48, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23092114

RESUMEN

INTRODUCTION: Diabetes mellitus impacts almost 200 million individuals worldwide and leads to debilitating complications. New avenues of drug discovery must target the underlying cellular processes of oxidative stress, apoptosis, autophagy, and inflammation that can mediate multi-system pathology during diabetes mellitus. AREAS COVERED: The authors examine the novel directions for drug discovery that involve: the ß-nicotinamide adenine dinucleotide (NAD(+)) precursor nicotinamide, the cytokine erythropoietin, the NAD(+)-dependent protein histone deacetylase SIRT1, the serine/threonine-protein kinase mammalian target of rapamycin (mTOR), and the wingless pathway. Furthermore, the authors present the implications for the targeting of these pathways that oversee gluconeogenic genes, insulin signaling and resistance, fatty acid beta-oxidation, inflammation, and cellular survival. EXPERT OPINION: Nicotinamide, erythropoietin, and the downstream pathways of SIRT1, mTOR, forkhead transcription factors, and wingless signaling offer exciting prospects for novel directions of drug discovery for the treatment of metabolic disorders. Future investigations must dissect the complex relationship and fine modulation of these pathways for the successful translation of robust reparative and regenerative strategies against diabetes mellitus and the complications of this disorder.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/tendencias , Animales , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/tendencias , Eritropoyetina/administración & dosificación , Eritropoyetina/metabolismo , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Niacinamida/administración & dosificación , Niacinamida/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...