Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
World J Diabetes ; 15(9): 1979-2001, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39280179

RESUMO

BACKGROUND: Diabetic intracerebral hemorrhage (ICH) is a serious complication of diabetes. The role and mechanism of bone marrow mesenchymal stem cell (BMSC)-derived exosomes (BMSC-exo) in neuroinflammation post-ICH in patients with diabetes are unknown. In this study, we investigated the regulation of BMSC-exo on hyperglycemia-induced neuroinflammation. AIM: To study the mechanism of BMSC-exo on nerve function damage after diabetes complicated with cerebral hemorrhage. METHODS: BMSC-exo were isolated from mouse BMSC media. This was followed by transfection with microRNA-129-5p (miR-129-5p). BMSC-exo or miR-129-5p-overexpressing BMSC-exo were intravitreally injected into a diabetes mouse model with ICH for in vivo analyses and were cocultured with high glucose-affected BV2 cells for in vitro analyses. The dual luciferase test and RNA immunoprecipitation test verified the targeted binding relationship between miR-129-5p and high-mobility group box 1 (HMGB1). Quantitative polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay were conducted to assess the levels of some inflammation factors, such as HMGB1, interleukin 6, interleukin 1ß, toll-like receptor 4, and tumor necrosis factor α. Brain water content, neural function deficit score, and Evans blue were used to measure the neural function of mice. RESULTS: Our findings indicated that BMSC-exo can promote neuroinflammation and functional recovery. MicroRNA chip analysis of BMSC-exo identified miR-129-5p as the specific microRNA with a protective role in neuroinflammation. Overexpression of miR-129-5p in BMSC-exo reduced the inflammatory response and neurological impairment in comorbid diabetes and ICH cases. Furthermore, we found that miR-129-5p had a targeted binding relationship with HMGB1 mRNA. CONCLUSION: We demonstrated that BMSC-exo can reduce the inflammatory response after ICH with diabetes, thereby improving the neurological function of the brain.

2.
Eur J Pharmacol ; 968: 176381, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38341077

RESUMO

Diabetes mellitus causes brain microvascular endothelial cell (MEC) damage, inducing dysfunctional angiogenic response and disruption of the blood-brain barrier (BBB). Canagliflozin is a revolutionary hypoglycemic drug that exerts neurologic and/or vascular-protective effects beyond glycemic control; however, its underlying mechanism remains unclear. In the present study, we hypothesize that canagliflozin ameliorates BBB permeability by preventing diabetes-induced brain MEC damage. Mice with high-fat diet/streptozotocin-induced diabetes received canagliflozin for 8 weeks. We assessed vascular integrity by measuring cerebrovascular neovascularization indices. The expression of specificity protein 1 (Sp1), as well as tight junction proteins (TJs), phosphorylated AMP-activated protein kinase (p-AMPK), and adenosine A2A receptors was examined. Mouse brain MECs were grown in high glucose (30 mM) to mimic diabetic conditions. They were treated with/without canagliflozin and assessed for migration and angiogenic ability. We also performed validation studies using AMPK activator (AICAR), inhibitor (Compound C), Sp1 small interfering RNA (siRNA), and adenosine A2A receptor siRNA. We observed that cerebral pathological neovascularization indices were significantly normalized in mice treated with canagliflozin. Increased Sp1 and adenosine A2A receptor expression and decreased p-AMPK and TJ expression were observed under diabetic conditions. Canagliflozin or AICAR treatment alleviated these changes. However, this alleviation effect of canagliflozin was diminished again after Compound C treatment. Either Sp1 siRNA or adenosine A2A receptor siRNA could increase the expression of TJs. Luciferase reporter assay confirmed that Sp1 could bind to the adenosine A2A receptor gene promoter. Our study identifies the AMPK/Sp1/adenosine A2A receptor pathway as a treatment target for diabetes-induced cerebrovascular injury.


Assuntos
Diabetes Mellitus , Hiperglicemia , Camundongos , Animais , Barreira Hematoencefálica/metabolismo , Receptor A2A de Adenosina/metabolismo , Canagliflozina/farmacologia , Canagliflozina/uso terapêutico , Proteínas Quinases Ativadas por AMP/metabolismo , Hiperglicemia/complicações , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Diabetes Mellitus/metabolismo , RNA Interferente Pequeno/metabolismo
3.
Mol Neurobiol ; 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38055163

RESUMO

Circular RNAs (circRNAs) have been progressively recognized as critical regulators in the pathology and pathophysiology of central nervous system disease. However, the potential role of circRNAs in intracerebral hemorrhage (ICH) is still largely unclear. Here, we demonstrate that circTrim37 expression was significantly upregulated at 3 days after ICH by circular RNA microarray and qPCR assays. Overexpression of circTrim37 could significantly ameliorate brain injury volume, brain edema, neurologic deficits, and inflammation in vivo after ICH. CircTrim37 promotes M2 polarization while restrains M1 polarization in vitro. Furthermore, circTrim37 acts as an endogenous sponge for miR-30c-5p, thereby inhibiting miR-30c-5p activity, leading to the upregulation of SOCS3 and making the balance of microglial response towards an M2 phenotype. Taken together, our results indicate the participation of circTrim37 and its coupling mechanism in ICH and provide a novel therapeutic target for ICH.

4.
Mol Neurobiol ; 60(12): 7060-7079, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37525083

RESUMO

Intracerebral hemorrhage (ICH) is a lethal stroke with high mortality or disability. However, effective therapy for ICH damage is generally lacking. Previous investigations have suggested that lysosomal protein transmembrane 5 (LAPTM5) is involved in various pathological processes, including autophagy, apoptosis, and inflammation. In this study, we aimed to identify the expression and functions of LAPTM5 in collagenase-induced ICH mouse models and hemoglobin-induced cell models. We found that LAPTM5 was highly expressed in brain tissues around the hematoma, and double immunostaining studies showed that LAPTM5 was co-expressed with microglia cells, neurons, and astrocytes. Following ICH, the mice presented increased brain edema, blood-brain barrier permeability, and neurological deficits, while pathological symptoms were alleviated after the LAPTM5 knockdown. Adeno-associated virus 9-mediated downregulation of LAPTM5 also improves ICH-induced secondary cerebral damage, including neuronal degeneration, the polarization of M1-like microglia, and inflammatory cascades. Furthermore, LAPTM5 promoted activation of the nuclear factor kappa-B (NF-κB) pathway in response to neuroinflammation. Further investigations indicated that brain injury improved by LAPTM5 knockdown was further exacerbated after the overexpression of receptor-interacting protein kinase 1 (RIP1), which is revealed to trigger the NF-κB pathway. In vitro experiments demonstrated that LAPTM5 silencing inhibited hemoglobin-induced cell function and confirmed regulation between RIP1 and LAPTM5. In conclusion, the present study indicates that LAPTM5 may act as a positive regulator in the context of ICH by modulating the RIP1/NF-κB pathway. Thus, it may be a candidate gene for further study of molecular or therapeutic targets.


Assuntos
Lesões Encefálicas , Animais , Camundongos , Lesões Encefálicas/complicações , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Hemorragia Cerebral/patologia , Hemoglobinas , Lisossomos/metabolismo , NF-kappa B/metabolismo
5.
Mol Med ; 29(1): 44, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-37013491

RESUMO

BACKGROUND: Dysregulated long non-coding RNAs participate in the development of diabetic cerebral ischemia. This study aimed to investigate the underlying mechanism of lncRNA MALAT1 in diabetic cerebral ischemia. METHOD: Middle cerebral artery occlusion (MCAO) was performed to establish diabetic cerebral I/R in vivo. TTC and neurological deficits assessment were performed to assess cerebral ischemic injury. LDH was conducted to detect cytotoxicity. RT-qPCR and western blotting assays were applied to determine mRNA and protein expression. Flow cytometry was performed to detect the pyroptosis of BV2 cells. Immunofluorescence and FISH were conducted for subcellular localization of MALAT1 and STAT1. ELISA was performed to determine cytokine release. Dual luciferase reporter, RIP, and ChIP assays were used to validate the interaction between STAT1 and MALAT1/NLRP3. Diabetes aggravated cerebral injury in vivo and in vitro. Diabetic cerebral ischemia induced inflammatory response and inflammation-induced cell pyroptosis. RESULT: MALAT1 was overexpressed in diabetic cerebral ischemia models in vivo and in vitro. However, knockdown of MALAT1 suppressed inflammatory response and the pyroptosis of BV2 cells. Moreover, MALAT1 interacted with STAT1 to transcriptionally activate NLRP3. Knockdown of STAT1 significantly reversed the effects of MALAT1. Furthermore, STAT1 promotes the MALAT1 transcription. MALAT1 interacts with STAT1 to promote the pyroptosis of microglias induced by diabetic cerebral ischemia through activating NLRP3 transcription. CONCLUSION: Thus, knockdown of MALAT1 may be a potential promising therapy target for diabetic cerebral ischemia.


Assuntos
Isquemia Encefálica , Diabetes Mellitus , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Isquemia Encefálica/genética , Microglia/metabolismo , MicroRNAs/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Traumatismo por Reperfusão/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/farmacologia , Animais
6.
Curr Neurovasc Res ; 19(2): 181-187, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35702793

RESUMO

OBJECTIVE: The aim of this study was to investigate whether heme oxygenase-1 (HO-1) promotes an early neuroinflammatory response after intracerebral hemorrhage (ICH) by regulating the toll-like receptor 4 (TLR4) signaling pathway. METHODS: We used a stereotaxic instrument to induce a mouse model of ICH through collagenase. We divided the participants into a control group, an ICH group, and an ICH and zinc protoporphyrin IX (ZnPP) group. The temporal expression pattern and cell localization of HO-1 and TLR4 after the ICH were detected by immunofluorescence and western blot; after the expression of HO-1 was inhibited, the expression levels of the TLR4 protein, the downstream molecule myeloid differentiation factor 88 (MyD88), the Toll and interleukin-1 receptor (TIR) -domain-containing adapter-inducing interferon-ß (TRIF) and the inflammatory factors were measured by western blotting. RESULTS: Immunofluorescence showed that HO-1 and TLR4 had similar temporal expression patterns and cellular localization after ICH, and we found that inhibiting HO-1 reduces the expression of TLR4 protein pathways, including TLR4, MyD88, TRIF, and related inflammatory factors, by studying the inhibitor ZnPP. CONCLUSION: These results indicate that HO-1 may promote early neuroinflammation after ICH through the TLR4/MyD88/TRIF signaling pathway.


Assuntos
Hemorragia Cerebral , Heme Oxigenase-1 , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Camundongos , Hemorragia Cerebral/metabolismo , Heme Oxigenase-1/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo
7.
Curr Neurovasc Res ; 19(2): 196-202, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35657042

RESUMO

OBJECTIVE: This study's purpose is to investigate the neuroprotective role of ethyl pyruvate (EP) in the pathogenesis of diabetic intracerebral hemorrhage. METHODS: The present study used a mouse model of collagenase-induced intracerebral hemorrhage (ICH) and streptozotocin-induced diabetes. The C57BL/6 mice were randomly divided into 3 groups: sham operation, diabetic cerebral hemorrhage, and diabetic cerebral hemorrhage with EP. The EP (80 mg/kg) and EP (50 mg/kg) were injected intraperitoneally one day and one hour before modeling. The protein expression levels of high mobility group box 1 (HMGB1) and NOD-like receptors 3 (NLRP3) were detected with western blot. The mRNA levels of HMGB1 and toll-like receptor 4 (TLR4) were measured by quantitative real-time polymerase chain reaction (PCR). Immunofluorescence and ELISA were performed to confirm some inflammatory factors. RESULTS: Compared to the normal diabetic intracerebral hemorrhage group, the mRNA and protein expression levels of HMGB1 and TLR4 were downregulated in the EP-affected group with diabetic cerebral hemorrhage, together with the downregulation of the expression of inflammasomes, including NLRP3, apoptosis-associated speck-like protein containing CARD (ASC), and caspase 1. CONCLUSION: EP can reduce the inflammatory response after diabetic intracerebral hemorrhage and may inhibit the activation of inflammasomes by the HMGB1/TLR4 pathway.


Assuntos
Diabetes Mellitus Experimental , Transdução de Sinais , Animais , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Hemorragia Cerebral/complicações , Hemorragia Cerebral/tratamento farmacológico , Inflamassomos/metabolismo , RNA Mensageiro/metabolismo
8.
Pak J Pharm Sci ; 34(6): 2121-2129, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35034872

RESUMO

Our study aimed to explore the impacts of liraglutide on brain dysfunction of type 2 diabetes mellitus. Rats in liraglutide treatment group were diabetic rats further received daily intraperitoneal administration of liraglutide for continuous 6 weeks. Body weight and blood glucose were measured weekly. Vascular structure changes in brain tissues were evaluated by Periodic acid-Schiff (PAS) staining. Angiopoietin-2 (ANG-2), high-mobility group box 1 (HMGB-1), CD105, NeuN, Oligo-2 in brain tissues were measured by immunohistochemistry staining and ANG-2, HMGB-1, and matrix metalloproteinase-9 (MMP-9) were detected by western blotting. Blood glucose levels of rats in diabetic model group were significantly elevated and blood glucose levels of rats in liraglutide treatment group were reduced to comparable levels with control group. PAS staining showed vascular basement membrane of rats in the diabetic model group was thicker than that of the control group. ANG-2, HMGB1 and MMP-9 were up-regulated in the diabetic model group comparing the control group, while down-regulated after treated with liraglutide (p<0.05). NeuN expressions were significantly higher in liraglutide treatment group. Liraglutide may have protective roles against brain injury of streptozotocin induced diabetic rats by inhibiting HMGB1, which further suppressing the MMP-9 and ANG-2.


Assuntos
Lesões Encefálicas/prevenção & controle , Encéfalo/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Liraglutida/farmacologia , Angiopoietina-2/metabolismo , Animais , Antígenos Nucleares/metabolismo , Glicemia/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/induzido quimicamente , Endoglina/metabolismo , Proteína HMGB1/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ratos Sprague-Dawley , Estreptozocina
9.
Aging (Albany NY) ; 12(13): 12869-12895, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32602850

RESUMO

Differences in microRNA (miRNA) expression after intracerebral hemorrhage (ICH) have been reported in human and animal models, and miRNAs are being investigated as a new treatment for inflammation and oxidative stress after ICH. In this study, we found that microRNA-183-5p expression was decreased in the mouse brain after ICH. To investigate the effect of miRNA-183-5p on injury and repair of brain tissue after ICH, saline, miRNA-183-5p agomir, or miRNA-183-5p antagomir were injected into the lateral ventricles of 8-week-old mice with collagenase-induced ICH. Three days after ICH, mice treated with exogenous miRNA-183-5p showed less brain edema, neurobehavioral defects, inflammation, oxidative stress, and ferrous deposition than control mice. In addition, by alternately treating mice with a heme oxygenase-1 (HO-1) inducer, a HO-1 inhibitor, a nuclear factor erythroid 2-related factor (Nrf2) activator, and Nrf2 knockout, we demonstrated an indirect, HO-1-dependent regulatory relationship between miRNA-183-5p and Nrf2. Our results indicate that miRNA-183-5p and HO-1 are promising therapeutic targets for controlling inflammation and oxidative damage after hemorrhagic stroke.


Assuntos
Hemorragia Cerebral/metabolismo , Heme Oxigenase-1/metabolismo , Proteínas de Membrana/metabolismo , MicroRNAs , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Heme Oxigenase-1/análise , Heme Oxigenase-1/genética , Inflamação , Masculino , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL
10.
Oncol Lett ; 13(4): 2684-2690, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28454451

RESUMO

Recent reports have demonstrated that long-term and high dosage treatments with incretin-based medicine, such as hormone glucagon-like peptide-1 (GLP-1) may induce thyroid C-cell pathological changes in rodents, rather than in humans. Doubts regarding the tumorigenic potential of GLP-1 analogues in human thyroid C-cells remain. The present study aimed to determine the expression levels of GLP-1 receptor (GLP-1R) and cluster of differentiation 26 (CD26) in the C-cells of thyroid tissues from non-neoplastic, medullary carcinoma and hyperplasia subjects, and to explore the potential clinical significance. The following cases were analyzed: Medullary thyroid carcinoma (n=62, including 59 paraffin-embedded samples and 3 fresh frozen samples), C-cell hyperplasia (n=20, paraffin-embedded samples) and non-neoplastic thyroid tissue samples (n=7, paraffin-embedded samples). GLP-1R and CD26 expression was detected using immunohistochemical staining and western blotting. There were significant differences in the expression levels of the two markers between medullary thyroid carcinoma and C-cell hyperplasia, in addition to between medullary thyroid carcinoma and non-neoplastic thyroid tissue following immunohistochemical staining. Similar significant differences in the expression of GLP-1R and CD26 were detected using western blot analysis in the medullary thyroid carcinoma compared with non-neoplastic thyroid tissue sectioned from the aforementioned fresh frozen samples. There was a significant negative correlation between GLP-1R and CD26 expression. In addition, the present data indicated that GLP-1R expression was associated with the age of the patients with medullary thyroid carcinoma. These results suggested that GLP-1R and CD26 may be closely associated with the development of thyroid C-cell hyperplasia and medullary thyroid carcinoma, and indicated the importance of being aware of the side effects of incretin medicine.

11.
J Cereb Blood Flow Metab ; 37(1): 25-38, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27317654

RESUMO

Intracerebral hemorrhage (ICH) is a devastating form of stroke with high morbidity and mortality. Heme oxygenase-1 (HO-1), the key enzyme in heme degradation, is highly expressed after ICH, but its role is still unclear. In this study, we used an HO-1 inducer and inhibitor to test the role of HO-1 in different stages of ICH in vivo and in vitro. In the early stage of ICH, high HO-1 expression worsened the outcomes of mice subjected to the collagenase-induced ICH model. HO-1 increased brain edema, white matter damage, neuronal death, and neurobehavioral deficits. Furthermore, elevated HO-1 increased inflammation, oxidative stress, matrix metalloproteinase-9/2 activity, and iron deposition. In the later stage of ICH, long-term induction of HO-1 increased hematoma absorption, angiogenesis, and recovery of neurologic function. We conclude that HO-1 activation mediates early brain damage after ICH but promotes neurologic function recovery in the later stage of ICH.


Assuntos
Hemorragia Cerebral/enzimologia , Heme Oxigenase-1/sangue , Envelhecimento , Animais , Lesões Encefálicas/induzido quimicamente , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Colagenases/farmacologia , Heme Oxigenase-1/metabolismo , Camundongos , Recuperação de Função Fisiológica/efeitos dos fármacos , Fatores de Tempo
12.
Mol Med Rep ; 14(6): 5535-5543, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27840991

RESUMO

Stroke is a life-threatening disease that results in significant disability in the human population. Despite the advances in current stroke therapies, a host of patients do not benefit from the conventional treatments. Thus, more effective therapies are required. It has been previously reported that leucine­rich­α2­glycoprotein 1 (LRG1) is crucial during the formation of new blood vessels in retinal diseases. However, the function of LRG1 in the brain during the neovessel growth process following ischemic stroke has not been fully elucidated and the mechanism underlying its effect on angiogenesis remains unclear. The purpose of the current study was to demonstrate whether LRG1 may promote angiogenesis through the transforming growth factor (TGF)­ß1 signaling pathway in ischemic rat brain following middle cerebral artery occlusion (MCAO). In the present study, the spatial and temporal expression of LRG1, TGF­ß1, vascular endothelial growth factor (VEGF) and angiopoietin­2 (Ang­2) were detected in ischemic rat brain following MCAO using reverse transcription­quantitative polymerase chain reaction (RT­qPCR), western blot analysis and immunohistochemistry. CD34 immunohistochemistry staining was used as an indicator of microvessel density (MVD). The RT­qPCR and western blotting results revealed that the levels of LRG1 and TGF­ß1 mRNA and protein expression were significantly increased as early as 6 and 12 h after MCAO (P<0.05), respectively, peaked at 3 days and persisted at significantly higher level until 14 days, in comparison with the control group. Additionally, VEGF and Ang­2 were also increased following MCAO. Furthermore, the immunohistochemistry results suggested that the MVD was increased following MCAO. In addition, the results also revealed that the percentage of LRG1­positive cells was positively correlated with the percentage of TGF­ß1­positive cells, and the percentage of LRG1­positive and TGF­ß1­positive cells had a positively correlation with the MVD. Taken together, the present study indicated that LRG1 may promote angiogenesis through upregulating the TGF­ß1 signaling pathway in ischemic rat brain following MCAO. This may provide a potential therapeutic target for the treatment of ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Glicoproteínas/metabolismo , Neovascularização Patológica/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Biomarcadores , Isquemia Encefálica/genética , Modelos Animais de Doenças , Expressão Gênica , Glicoproteínas/genética , Masculino , Neovascularização Patológica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Can J Neurol Sci ; 40(6): 814-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24257222

RESUMO

BACKGROUND: Meningiomas account for approximately 24-30% of primary intracranial neoplasms. Histopathologic grade and degree of resection are two major prognostic factors. The aim of this study was to determine the factors associated with the prognosis of meningioma. METHODS: We used immunohistochemistry to analyze the expression levels of Vav3, SPARC, p-Akt, cyclin D1, and Ki-67 in 287 meningiomas of all grades. RESULTS: The expression of Vav3, SPARC, p-Akt, cyclin D1, and Ki- 67 significantly increased with meningioma grade (p<0.01), and was higher in brain-invasive meningiomas compared to non-invasive meningiomas (WHO grade I) (p<0.05). Furthermore, the expression of Vav3, p-Akt, and Ki-67 was higher in recurrent meningiomas compared to non-recurrent meningiomas (WHO grade I) (p<0.05). CONCLUSION: The expression of Vav3, SPARC, p-Akt, cyclin D1, and Ki-67 in meningiomas appears to correlate with meningioma invasiveness, aggressiveness, and recurrence.Expression de SPARC et de Vav3 dans le méningiome : facteurs reliés au pronostic.


Assuntos
Meningioma , Recidiva Local de Neoplasia , Humanos , Imuno-Histoquímica , Neoplasias Meníngeas , Prognóstico
14.
Breast Cancer (Auckl) ; 6: 1-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22259247

RESUMO

BACKGROUND: Recent studies have shown that androgen displays an inhibitory effect on breast cancer cell lines that express androgen receptor (AR) but not estrogen receptor (ER) and progesterone receptor (PR). We have previously reported that approximately 1/3 of ER negative high grade invasive ductal carcinomas express AR. Thus, AR can serve as a potential therapeutic target for this group of patients. AIM: Here we investigated AR expression patterns in 980 consecutive breast carcinomas. RESULTS: We found that (1) AR was expressed more frequently (77%) than ER (61%) and PR (60%) in breast carcinomas; (2) AR expression was associated with ER and PR expression (P < 0.0001), small tumor size (P = 0.0324) and lower Ki-67 expression (P = 0.0013); (3) AR expression was found in 65% of ER negative tumors; (4) AR expression was associated with PR and Ki-67 in ER negative tumors, but not in ER positive tumors; (5) AR expression was higher in ER positive subtypes (Luminal A, Luminal B and Luminal HER2 subtypes, 80%-86%) and lower in ER negative subtypes [HER2, triple negative (TN), and TN EFGR positive subtypes; 52%-66%], with over 50% of TN tumors expressing AR. CONCLUSION: More breast carcinomas express AR than ER and PR, including significant numbers of ER negative and TN tumors, for which AR could serve as a potential therapeutic target.

15.
Neurochem Int ; 58(8): 872-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21334414

RESUMO

A better understanding of the underlying mechanisms of angiogenesis and vascular permeability is necessary for the development of therapeutic strategies for ischemic injury. The purpose of this study was to examine the spatial and temporal expression of Src and Src-suppressed C kinase substrate (SSeCKS) in brain after middle cerebral artery occlusion (MCAO) and elucidate the relationships among Src, SSeCKS, and the key angiogenic factors present after stroke. Rats were subjected to either MCAO or sham operation. Reverse transcriptase-polymerase chain reaction and Western blotting results revealed that Src gradually increased starting as early as 2 h after MCAO and remained high for 1 day. In contrast, SSeCKS decreased after MCAO. Src expression correlated positively with that of vascular endothelial growth factor and angiopoietin-2, and negatively with that of SSeCKS, angiopoietin-1, and zonula occludens-1. However, SSeCKS had the reverse correlations. Changes in the expression of these factors correlated with the progress of angiogenesis and cerebral edema. Dynamic temporal changes in Src and SSeCKS expression may modulate angiogenesis and cerebral edema formation after focal cerebral ischemia.


Assuntos
Proteínas de Ancoragem à Quinase A/biossíntese , Indutores da Angiogênese/metabolismo , Edema Encefálico/enzimologia , Isquemia Encefálica/enzimologia , Proteínas de Ciclo Celular/biossíntese , Neovascularização Patológica/enzimologia , Quinases da Família src/biossíntese , Animais , Edema Encefálico/etiologia , Isquemia Encefálica/complicações , Masculino , Neovascularização Patológica/etiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
16.
J Ethnopharmacol ; 133(3): 1109-16, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21129477

RESUMO

Ginseng has long been used to alleviate many ailments, particularly those associated with aging and memory deterioration. In the present study we aimed to investigate the neuroprotective effects of ginsenoside Rb1, against Aß(1-42) toxicity in cultured cortical neurons and also the potential involvement of PI3K/Akt/GSK-3ß signal pathway. Cortical neurons were pre-treated with ginsenoside Rb1 (20, 40, 100 µM) or LiCl (1, 5, 10 mM) for 24 h, and then were co-treated with 20 µM Aß(1-42) for 12 h. In some experiments to evaluate the mechanism of Rb1 action, a PI3K inhibitor (LY294002 10 µM) was co-administered with Rb1 for the 24-h pretreatment. We revealed that Rb1 significantly attenuated Aß(1-42)-induced neurotoxicity and tau hyperphosphorylation at multiple AD-related sites in a dose-dependent manner. Simultaneously, it increased the levels of phospho-Ser(473)-Akt and down-regulated GSK-3ß activity by PI3K activation. The neuroprotective effects of Rb1 against Aß(1-42)-induced neurotoxicity and tau hyperphosphorylation were blocked by LY294002 (10 µM), a PI3K inhibitor. In addition, Rb1 reversed the Aß(1-42)-induced decrease in phosphorylation cyclic AMP response element binding (CREB) protein, which could also be blocked by the PI3K inhibitor. All these findings suggest that Rb1 may represent a potential treatment strategy for Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Ginsenosídeos/farmacologia , Quinase 3 da Glicogênio Sintase/metabolismo , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/prevenção & controle , Animais , Western Blotting , Glicogênio Sintase Quinase 3 beta , Microscopia de Fluorescência , Fosforilação , Ratos , Ratos Wistar
17.
Zhonghua Bing Li Xue Za Zhi ; 40(12): 834-9, 2011 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-22336210

RESUMO

OBJECTIVE: To examine the temporal and spatial expression of vascular endothelial growth factor (VEGF) and angiopoietins (Ang) in rat brain after cerebral ischemia, and to elucidate the roles they played in angiogenesis and vascular permeability. METHODS: Rats were subjected to either middle cerebral artery occlusion (MCAO) or sham operation. Reverse transcriptase-polymerase chain reaction, Western blotting, and immunohistochemistry were used to detect the expression of VEGF, Ang-1 and Ang-2 at different time points after ischemia. CD31 was used to label endothelial cells after MCAO. Vascular permeability was determined by Evans blue. RESULTS: VEGF was markedly increased at 2 h, had an initial peak at 12 h (0.7249 ± 0.1933, P < 0.01), and a second peak at 7 days (0.5264 ± 0.1519, P < 0.01). Ang-2 mRNA and protein significantly increased after MCAO, both of them peaked at 12 h (0.6747 ± 0.2416, P < 0.01; 1.1197 ± 0.1780, P < 0.01). In contrast, Ang-1 mRNA and protein gradually decreased after MCAO, respectively reaching a minimum at 3 d (0.3220 ± 0.1427, P < 0.01) and 1 d (0.1298 ± 0.0293, P < 0.01). Changes in the expression of these factors correlated with the progress of angiogenesis and vascular permeability. Evans blue test revealed that the vascular permeability gradually increased, and peaked at day 1 after ischemia [(6.219 ± 0.887) µg/g, P < 0.01]. CONCLUSION: Dynamic temporal changes in VEGF, Ang-1 and Ang-2 expression stimulate the cerebral angiogenesis after focal cerebral ischemia.


Assuntos
Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Angiopoietina-1/genética , Angiopoietina-2/genética , Animais , Western Blotting , Permeabilidade Capilar , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Neovascularização Fisiológica , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética
18.
Chin Med J (Engl) ; 123(20): 2832-5, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21034592

RESUMO

BACKGROUND: Hyalinizing trabecular tumor (HTT) is a rare thyroid neoplasm, which shares some histologic features with thyroid papillary carcinoma (TPC). Clinically, it is frequently misdiagnosed as papillary carcinoma, even for some experienced pathologists. The aim of this study was to investigate whether HTT is variant of TPC or HTT is an independent entity of thyroid neoplasm. METHODS: The expression of CK19, galectin-3, HBME-1 and MIB-1 was detected by immunohistochemical staining in 12 cases of hyalinizing trabecular tumor and 20 cases of thyroid papillary carcinoma. RESULTS: Two of the 12 HTT samples were positive or focally positive for CK19. Four of the 12 samples of HTT presented positive to galectin-3; 3 were stained strongly and the other one was focally positive. None of the 12 samples of HTT was positive for HBME-1. Five in 12 HTT samples were stained in nucleus for MIB-1. Almost all the 20 cases of thyroid papillary carcinoma were intensely stained for CK19, galectin-3 and HBME-1. Fifteen in 20 cases of thyroid papillary carcinoma showed nuclear staining for MIB-1. CONCLUSIONS: HTT is an independent thyroid neoplasm, not a variant of TPC. This study could help in the differential diagnosis of HTT from TPC. CK19, galectin-3 and HBME-1 are adequate to identify HTT and TPC, but MIB-1 does not play an important role in discrimination between HTT and TPC.


Assuntos
Carcinoma Papilar/química , Neoplasias da Glândula Tireoide/química , Adolescente , Adulto , Idoso , Biomarcadores Tumorais/análise , Carcinoma Papilar/diagnóstico , Criança , Diagnóstico Diferencial , Feminino , Galectina 3/análise , Humanos , Imuno-Histoquímica , Queratina-19/análise , Masculino , Pessoa de Meia-Idade , Neoplasias da Glândula Tireoide/diagnóstico , Ubiquitina-Proteína Ligases/análise
19.
Chin Med J (Engl) ; 123(15): 2077-81, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20819545

RESUMO

BACKGROUND: Intracerebral hemorrhage (ICH) can cause brain damage through a number of pathways. The purpose of the study was to explore the effect of thrombin, protease nexin-1 (PN-1) and protease activated receptor-1 (PAR-1) in rat and human cerebellum after ICH. METHODS: A model of ICH was produced in adult Sprague-Dawley rats by direct injection of autologous blood (50 microl) into caudate nucleus. Patients with injured hemorrhage were also enrolled in this study. Different expressions of thrombin, PAR-1, PN-1 were detected in rat and human cerebellum by immunohistochemistry and in situ hybridization. RESULTS: In rat cerebellum, thrombin protein significantly increased at 6 hours and reached the maximum 2 days after ICH. The expression of PAR-1 protein reached the maximum at 24 - 48 hours, and then began to decrease. The expression of PN-1 protein reached the maximum at 3 hours, decreased somewhat after that and increased a little at 5 days after ICH. While in human cerebellum, the changing tendency of thrombin, PAR-1 and PN-1 was almost conform to the rat. CONCLUSION: In cerebellum, thrombin can activate PAR-1 expression after ICH, and PN-1 appears quickly after ICH in order to control the deleterious effect of thrombin.


Assuntos
Cerebelo/metabolismo , Hemorragia Cerebral/metabolismo , Receptor PAR-1/metabolismo , Serpina E2/metabolismo , Trombina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Receptor PAR-1/genética , Serpina E2/genética , Trombina/genética
20.
Neurochem Int ; 57(3): 248-53, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20541575

RESUMO

Intracerebral hemorrhage (ICH) can cause secondary brain damage through inflammation-related pathways. Thrombin and one of its receptors, protease activated receptor-1 (PAR-1); matrix metalloproteinase (MMP)-9; and aquaporin (AQP)-4 are stroke-related inflammatory mediators that have been implicated in ICH pathology. To further characterize the inflammatory response after ICH, we studied the temporal profile of the expression of these inflammatory mediators and assessed their potential correlation with brain edema formation after brain hemorrhage in rats. ICH was modeled by infusing autologous blood into the striatum. Then mRNA and protein expression was assessed over the course of 5 days. We found that the mRNA and/or protein expression of thrombin, PAR-1, AQP-4, and MMP-9 was upregulated between 2h and 5 days after ICH. Each reached a maximal level at day 2, except for AQP-4 protein, which peaked at day 5. Brain water content after ICH presented a similar trend; it was increased at 2h, peaked at day 2, and then decreased but remained elevated at day 5. Our data provide novel evidence that upregulation of these selected inflammatory mediators occurs very early and persists for several days after ICH, and that temporal patterns of expression of thrombin and AQP-4 are associated with brain edema formation. These findings have important implications for efforts to reduce secondary brain damage after ICH.


Assuntos
Edema Encefálico/metabolismo , Edema Encefálico/patologia , Mediadores da Inflamação/metabolismo , Hemorragias Intracranianas/metabolismo , Hemorragias Intracranianas/patologia , Animais , Aquaporina 4/biossíntese , Western Blotting , Água Corporal/metabolismo , Química Encefálica/fisiologia , Imuno-Histoquímica , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Neostriado/patologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptor PAR-1/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombina/biossíntese , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA