Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Neuropathol Exp Neurol ; 80(5): 457-466, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33870420

ABSTRACT

There are few effective preventive or therapeutic strategies to mitigate the effects of catastrophic intracerebral hemorrhage (ICH) in humans. Heme oxygenase is the rate-limiting enzyme in heme metabolism; heme oxygenase-2 (HO-2) is a constitutively expressed heme oxygenase. We explored the involvement of HO-2 in a collagenase-induced mouse model of ICH in C57BL/6 wild-type and HO-2 knockout mice. We assessed oxidative stress injury, blood-brain barrier permeability, neuronal damage, late-stage angiogenesis, and hematoma clearance using immunofluorescence, Western blot, MRI, and special staining methods. Our results show that HO-2 reduces brain injury volume and brain edema, alleviates cytotoxic injury, affects vascular function in the early stage of ICH, and improves hematoma absorbance and angiogenesis in the late stage of ICH in this model. Thus, we found that HO-2 has a protective effect on brain injury after ICH.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Cerebral Hemorrhage/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Neuroprotection/physiology , Animals , Blood-Brain Barrier/pathology , Brain Edema/metabolism , Brain Injuries/pathology , Disease Models, Animal , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Permeability
2.
Aging (Albany NY) ; 12(13): 12869-12895, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32602850

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , MicroRNAs , Animals , Brain/metabolism , Brain/pathology , Cell Line , Heme Oxygenase-1/analysis , Heme Oxygenase-1/genetics , Inflammation , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL
3.
Med Sci Monit ; 25: 794-800, 2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30686819

ABSTRACT

BACKGROUND It is well documented that the Blood-Brain barrier (BBB) can be damaged by matrix metalloproteases (MMPs) after intracerebral hemorrhage (ICH), but little is known about the mechanism of this effect. MATERIAL AND METHODS We established an ICH model in rats by injecting collagenase VII into the striatum. Afterwards, intraperitoneal injection of these rats with 40 mg/kg GM6001 (a MMPs inhibitor). The effects of GM6001 on ICH were investigated by neurological severity score, brain water content, Evans blue staining, hematoxylin-eosin staining, immunohistochemical staining, and Western blot assays. RESULTS We demonstrated that the neurological damage caused by ICH was relieved at 5 and 7 days following administration of GM6001. The impaired BBB induced by ICH was improved in response to GM6001 treatment at around 3 days, as evidenced by alleviated cerebral edema, decreased Evans blue extravasation, and a reduction in inflammatory cellular infiltration. Mechanism analysis revealed that ICH induced the generation of ß-dystroglycan cleavage, which could be suppressed by GM6001 treatment. Furthermore, we found that recombinant MMP2 and MMP9 triggered the cleavage of ß-dystroglycan in vitro, and this action could be inhibited by GM6001 administration. CONCLUSIONS Taken together, our results suggest that MMPs-mediated cleavage on ß-dystroglycan may play an important role in BBB after ICH.


Subject(s)
Blood-Brain Barrier/drug effects , Cerebral Hemorrhage/metabolism , Dystroglycans/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Edema/metabolism , Cerebral Hemorrhage/drug therapy , Collagenases/pharmacology , Dipeptides/pharmacology , Disease Models, Animal , Dystroglycans/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/metabolism , Rats , Rats, Wistar
4.
Epilepsy Res ; 144: 34-42, 2018 08.
Article in English | MEDLINE | ID: mdl-29751355

ABSTRACT

OBJECTIVE: Studies have shown that microRNAs play a role in the development of epilepsy by regulating downstream target messenger (m)RNA. The present study aims to determine the changes associated with microRNA-21-5p (miR-21-5p) during epileptogenesis in a kainic acid rat model, and to assess whether the PTEN-mTOR pathway is a target of miR-21-5p. METHOD: Reverse transcription polymerase chain reaction (RT-PCR) was used to examine the quantitative expressions of miR-21-5p and PTEN, and Western blotting was used to test the activity of mTOR in the acute, latent, and chronic stages of epileptogenesis. The antagomir of miR-21-5p was injected into the intracerebroventricular space using a microsyringe. Neuronal death and epilepsy discharge were assessed by Nissl staining and electroencephalography (EEG), respectively. The Morris water maze (MWM) was used to assess the cognitive impairment in rats after status epilepticus (SE). RESULTS: Both miR-21-5p and mTOR were upregulated and PTEN was downregulated in rats during acute, latent, and chronic stages of epileptogenesis when compared with those of the control. After using antagomir miR-21-5p in vivo, miR-21-5p and mTOR decreased and the expression of PTEN increased compared with that in the SE model. The silencing of miR-21-5p diminished the number of abnormal spikes on EEG and decreased the number of neuron deletions on Nissl staining. The cognitive and memory impairment caused by epilepsy could also be improved after miR-21-5p knockdown in vivo. CONCLUSION: The results of the present study demonstrate that PTEN-mTOR is the target of miR-21-5p in a kainic acid model of epilepsy. The knockout of miR-21-5p decreases the neuronal damage in stages of epileptogenesis. The miR-21-5p/PTEN/mTOR axis may be a potential target for preventing and treating seizures and epileptic damage.


Subject(s)
Anticonvulsants/therapeutic use , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Status Epilepticus/drug therapy , TOR Serine-Threonine Kinases/metabolism , Animals , Antagomirs/therapeutic use , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Injections, Intraventricular , Kainic Acid/toxicity , Male , Maze Learning/drug effects , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Rats , Rats, Wistar , Statistics, Nonparametric , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/pathology , TOR Serine-Threonine Kinases/genetics , Time Factors
5.
Oncol Lett ; 15(4): 4550-4556, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29616087

ABSTRACT

The aim of the present study was to identify potential key genes and single nucleotide variations (SNVs) in prostate cancer. RNA sequencing (RNA-seq) data, GSE22260, were downloaded from the Gene Expression Omnibus database, including 4 prostate cancer samples and 4 normal tissues samples. RNA-Seq reads were processed using Tophat and differentially-expressed genes (DEGs) were identified using the Cufflinks package. Gene Ontology enrichment analysis of DEGs was performed. Subsequently, Seqpos was used to identify the potential upstream regulatory elements of DEGs. SNV was analyzed using Genome Analysis Toolkit. In addition, the frequency and risk-level of mutant genes were calculated using VarioWatch. A total of 150 upregulated and 211 downregulated DEGs were selected and 25 upregulated and 17 downregulated potential upstream regulatory elements were identified, respectively. The SNV annotations of somatic mutations revealed that 65% were base transition and 35% were base transversion. At frequencies ≥2, a total of 17 mutation sites were identified. The mutation site with the highest frequency was located in the folate hydrolase 1B (FOLH1B) gene. Furthermore, 20 high-risk mutant genes with high frequency were identified using VarioWatch, including ribosomal protein S4 Y-linked 2 (RPS4Y2), polycystin 1 transient receptor potential channel interacting (PKD1) and FOLH1B. In addition, kallikrein 1 (KLK1) and PKD1 are known tumor suppressor genes. The potential regulatory elements and high-frequency mutant genes (RPS4Y2, KLK1, PKD1 and FOLH1B) may have key functions in prostate cancer. The results of the present study may provide novel information for the understanding of prostate cancer development.

6.
Neuro Oncol ; 20(1): 55-65, 2018 01 10.
Article in English | MEDLINE | ID: mdl-28651374

ABSTRACT

Background: Cancer immunotherapy represents a promising treatment approach for malignant gliomas but is hampered by the limited number of ubiquitously expressed tumor antigens and the profoundly immunosuppressive tumor microenvironment. We identified cluster of differentiation (CD)70 as a novel immunosuppressive ligand and glioma target. Methods: Normal tissues derived from 52 different organs and primary and recurrent low-grade gliomas (LGGs) and glioblastomas (GBMs) were thoroughly evaluated for CD70 gene and protein expression. The association between CD70 and patients' overall survival and its impact on T-cell death was also evaluated. Human and mouse CD70-specific chimeric antigen receptors (CARs) were tested respectively against human primary GBMs and murine glioma lines. The antitumor efficacies of these CARs were also examined in orthotopic xenograft and syngeneic models. Results: CD70 was not detected in peripheral and brain normal tissues but was constitutively overexpressed by isocitrate dehydrogenase (IDH) wild-type primary LGGs and GBMs in the mesenchymal subgroup and recurrent tumors. CD70 was also associated with poor survival in these subgroups, which may link to its direct involvement in glioma chemokine productions and selective induction of CD8+ T-cell death. To explore the potential for therapeutic targeting of this newly identified immunosuppressive axis in GBM tumors, we demonstrate that both human and mouse CD70-specific CAR T cells recognize primary CD70+ GBM tumors in vitro and mediate the regression of established GBM in xenograft and syngeneic models without illicit effect. Conclusion: These studies identify a previously uncharacterized and ubiquitously expressed immunosuppressive ligand CD70 in GBMs that also holds potential for serving as a novel CAR target for cancer immunotherapy in gliomas.


Subject(s)
Brain Neoplasms/therapy , CD27 Ligand/immunology , Receptors, Chimeric Antigen/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma , Humans , Immunotherapy, Adoptive/methods , Isocitrate Dehydrogenase/genetics , Mice , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
7.
Front Immunol ; 8: 1451, 2017.
Article in English | MEDLINE | ID: mdl-29163521

ABSTRACT

BACKGROUND: Angiogenesis and immune cell infiltration are key features of gliomas and their manipulation of the microenvironment, but their prognostic significance remains indeterminate. We evaluate the interconnection between tumor-infiltrating lymphocyte (TIL) and tumor blood-vasculatures in the context of glioma progression. METHODS: Paired tumor tissues of 44 patients from three tumor-recurrent groups: diffuse astrocytomas (DA) recurred as DA, DA recurred as glioblastomas (GBM), and GBM recurred as GBM were evaluated by genetic analysis, immunohistochemistry for tumor blood vessel density, TIL subsets, and clinical outcomes. These cells were geographically divided into perivascular and intratumoral TILs. Associations were examined between these TILs, CD34+ tumor blood vessels, and clinical outcomes. To determine key changes in TIL subsets, microarray data of 15-paired tumors from patients who failed antiangiogenic therapy- bevacizumab, and 16-paired tumors from chemo-naïve recurrent GBM were also evaluated and compared. RESULTS: Upon recurrence in primary gliomas, similar kinetic changes were found between tumor blood vessels and each TIL subset in all groups, but only CD4+ including Foxp3+ TILs, positively correlated with the density of tumor blood vessels. CD4 was the predominant T cell population based on the expression of gene-transcripts in primary GBMs, and increased activated CD4+ T cells were revealed in Bevacizumab-resistant recurrent tumors (not in chemo-naïve recurrent tumors). Among these TILs, 2/3 of them were found in the perivascular niche; Foxp3+ T cells in these niches not only correlated with the tumor vessels but were also an independent predictor of shortened recurrence-free survival (RFS) (HR = 4.199, 95% CI 1.522-11.584, p = 0.006). CONCLUSION: The minimal intratumoral T cell infiltration and low detection of CD8 transcripts expression in primary GBMs can potentially limit antitumor response. CD4+ and perivascular Foxp3+ TILs associate with tumor angiogenesis and tumor progression in glioma patients. Our results suggest that combining antiangiogenic agents with immunotherapeutic approaches may help improve the antitumor efficacy for patients with malignant gliomas.

8.
Int J Cancer ; 141(7): 1434-1444, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28612394

ABSTRACT

Tumor migration/metastasis and immunosuppression are major obstacles in effective cancer therapy. Incidentally, these 2 hurdles usually coexist inside tumors, therefore making therapy significantly more complicated, as both oncogenic mechanisms must be addressed for successful therapeutic intervention. Our recent report highlights that the tumor expression of a TNF family member, CD70, is correlated with poor survival for primary gliomas. In this study, we investigated how CD70 expression by GBM affects the characteristics of tumor cells and the tumor microenvironment. We found that the ablation of CD70 in primary GBM decreased CD44 and SOX2 gene expression, and inhibited tumor migration, growth and the ability to attract monocyte-derived M2 macrophages in vitro. In the tumor microenvironment, CD70 was associated with immune cell infiltrates, such as T cells; myeloid-derived suppressor cells; and monocytes/macrophages based on the RNA-sequencing profile. The CD163+ macrophages were far more abundant than T cells were. This overwhelming level of macrophages was identified only in GBM and not in low-grade gliomas and normal brain specimens, implying their tumor association. CD70 was detected only on tumor cells, not on macrophages, and was highly correlated with CD163 gene expression in primary GBM. Additionally, the co-expression of the CD70 and CD163 genes was found to correlate with decreased survival for patients with primary GBM. Together, these data suggest that CD70 expression is involved in promoting tumor aggressiveness and immunosuppression via tumor-associated macrophage recruitment/activation. Our current efforts to target this molecule using chimeric antigen receptor T cells hold great potential for treating patients with GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CD27 Ligand/metabolism , Glioblastoma/metabolism , Glioblastoma/secondary , Immune Tolerance , Antigens, CD/analysis , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/analysis , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/cytology , Brain Neoplasms/immunology , CD27 Ligand/analysis , CD27 Ligand/genetics , Cell Line, Tumor , Cell Migration Assays, Macrophage/methods , Cell Movement , Gene Expression Regulation, Neoplastic , Glioblastoma/immunology , Glioblastoma/mortality , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunity, Cellular , Macrophages/chemistry , Macrophages/cytology , Macrophages/immunology , Neoplasm Metastasis , Receptors, Cell Surface/analysis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
9.
J Cereb Blood Flow Metab ; 37(1): 25-38, 2017 01.
Article in English | MEDLINE | ID: mdl-27317654

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/enzymology , Heme Oxygenase-1/blood , Aging , Animals , Brain Injuries/chemically induced , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/pathology , Collagenases/pharmacology , Heme Oxygenase-1/metabolism , Mice , Recovery of Function/drug effects , Time Factors
10.
Int J Mol Med ; 38(6): 1831-1838, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27840897

ABSTRACT

Urotensin II (UII) was first recognized for its constrictive and natriuretic properties in fish almost 40 years ago, and recent studies have suggested that it exerts pro-fibrotic effects in a number of cell lines. In this study, we aimed to evaluate the role of UII in extracellular matrix (ECM) synthesis and secretion in advanced glycation end product (AGE)-stimulated rat proximal tubular epithelial cells (NRK-52E cells). UII promoted the proliferation of the NRK-52E cells in a dose-dependent manner over a concentration range of 10-10-10-8 mol/l and this effect was partly inhibited by both nimodipine and EDTA. Furthermore, AGE-BSA promoted the mRNA and protein expression of UII, fibronectin (FN) and collagen IV (ColIV) in the NRK-52E cells in a dose- and time-dependent manner. In addition, UII promoted the mRNA expression and protein secretion of transforming growth factor (TGF)-ß1, FN and Col IV by the NRK-52E cells. Our results suggest that UII promotes the proliferation of NRK-52E cells, an effect which is mediated by the influx of extracellular calcium ions. In addition, our data indicate that AGEs promote UII expression in NRK-52E cells, and that TGF-ß1 signaling is a candidate pathway mediating the involvement of UII in renal fibrosis. Collectively, our data suggest that the UII-TGF-ß1 signaling may be an important factor in tubulointerstitial nephropathy in diabetes.


Subject(s)
Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Glycation End Products, Advanced/metabolism , Kidney Tubules, Proximal/metabolism , Urotensins/metabolism , Animals , Biomarkers , Cell Proliferation , Cells, Cultured , Collagen Type IV/genetics , Collagen Type IV/metabolism , Epithelial Cells/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation/drug effects , Glycation End Products, Advanced/pharmacology , Kidney Tubules, Proximal/drug effects , Rats , Serum Albumin, Bovine/pharmacology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Urotensins/pharmacology
11.
Cell Cycle ; 15(12): 1545-51, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27124741

ABSTRACT

Silent information regulator type-1 (SIRT1) is the best-studied member of the Sirtuin (Sir2) family of nicotinamide dinucleotide (NAD)-dependent class III histone deacetylases (HDACs). Rrecently, it is suggested that SIRT1 may be involved in the development of malignant tumors including mouse lymphoma, but has not yet been explored in Angioimmunoblastic T-cell lymphoma (AITL). Therefore, we investigated the prevalence and the prognostic impact of SIRT1 expression in AITL. Immunohistochemical expression of SIRT1, p53 were evaluated by using a 2 mm core from 45 AITL patients. Positive expression of SIRT1 was seen in 71.11% (32 of 45) of patients and p53 expression were seen in 53.33% (24 of 45). SIRT1 and p53 expression were significantly associated with shorter PFS by univariate analysis (P=0.009 and P < 0.001, respectively), multivariate analysis also shows that SIRT1 expression relate to worse prognosis. We also suggest inferior survival in AITL with the combined expression of SIRT1 and clinical characteristics of high IPI scores, high clinical stage, increased serum LDH, decreased HGB and increased γ-Globulin. In conclusion, our results indicate that SIRT1 is strongly expressed in AITL and it act as a clinically significant prognostic indicator for AITL patients, may also serve as a therapeutic target in AITL.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Immunoblastic Lymphadenopathy/genetics , Lymphoma, T-Cell, Peripheral/genetics , Sirtuin 1/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cyclophosphamide/therapeutic use , Doxorubicin/therapeutic use , Female , Humans , Immunoblastic Lymphadenopathy/diagnosis , Immunoblastic Lymphadenopathy/drug therapy , Immunoblastic Lymphadenopathy/mortality , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/genetics , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/mortality , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prednisone/therapeutic use , Prognosis , Sirtuin 1/metabolism , Survival Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vincristine/therapeutic use , gamma-Globulins/genetics , gamma-Globulins/metabolism
12.
Zhonghua Bing Li Xue Za Zhi ; 44(5): 323-8, 2015 May.
Article in Chinese | MEDLINE | ID: mdl-26178214

ABSTRACT

OBJECTIVE: To study biological effect of recombinant human erythropoietin (RhEPO) on the expression of oligodendrocyte in the neuron glia antigen 2(NG2), Nogo receptor-interacting protein 1(LINGO-1), myelin basic protein (MBP) and myelin associated glycoprotein (MAG), and to explore the protective mechanism of RhEPO for oligodendrocyte after cerebral infarction. METHODS: Experimental rats were randomly divided into the treatment group (RhEPO at a dose of 3 000 U/kg) or saline control group. Both groups received intraperitoneal injection of RhEPO after cerebral ischemia in 30 min, 3 h, 6 h, 12 h and 24 h, which was administered daily for 7 days. The modified neurological severity score (mNSS) and histology were analyzed, and immunohistochemistry was used to detect the protein expression of NG2, MAG, MBP and LINGO-1. RESULTS: The overall mNSS of RhEPO treatment group significantly decreased compared with the saline control group on the seventh day after cerebral infarction (P<0.05). Such treatment effect was more obvious in the treatment group at 30 min and 3 h (P<0.01). Compared with the saline control group, the numbers of NG2 positive cells increased in RhEPO treatment group. In contrast, the expression of LINGO-1 protein significantly decreased (P<0.05), with a dramatic decrease observed at 30 min and 3 h (P<0.01). However, the expression of MBP protein decreased more significantly in saline control group, while the level of the MAG protein expression increased. The differences were statistically significant (P<0.05), especially at 30 min (P<0.01). CONCLUSIONS: After cerebral ischemia, RhEPO promotes the proliferation of NG2 positive cells, and inhibits the expression of LINGO-1 and MAG proteins. RhEPO improves the proliferation and differentiation of oligodendrocyte precursor cells, which in turn protects neuronal function, particularly at the early phase of ischemia.


Subject(s)
Cerebral Infarction/drug therapy , Erythropoietin/pharmacology , Oligodendroglia/drug effects , Animals , Antigens/metabolism , Cell Proliferation/drug effects , Cerebral Infarction/metabolism , Humans , Membrane Proteins/metabolism , Myelin Basic Protein/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Proteoglycans/metabolism , Random Allocation , Rats , Recombinant Proteins/pharmacology , Time Factors
13.
Tumour Biol ; 35(11): 11621-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25139094

ABSTRACT

Dystroglycan (DG), a multifunctional protein dimer of non-covalently linked α and ß subunits, is best known as an adhesion and transduction molecule linking the cytoskeleton and intracellular signaling pathways to extracellular matrix proteins. Loss of DG binding, possibly by degradation or disturbed glycosylation, has been reported in a variety of cancers. DG is abundant at astroglial endfeet forming the blood-brain barrier (BBB) and glia limitans; so, we examined if loss of expression is associated with glioma. Expression levels of α-DG and ß-DG were assessed by immunohistochemistry in a series of 78 glioma specimens to determine the relationship with tumor grade and possible prognostic significance. α-DG immunostaining was undetectable in 44 of 49 high-grade specimens (89.8%) compared to 15 of 29 low-grade specimens (51.72%) (P<0.05). Moreover, loss of α-DG expression was an independent predictor of shorter disease-free survival (DFS) (hazards ratio (HR) = 0.142, 95% confidence interval (CI) 0.033-0.611, P=0.0088). Reduced expression of both α-DG and ß-DG was also a powerful negative prognostic factor for DFS (HR=2.556, 95% CI 1.403-4.654, P=0.0022) and overall survival (OS) (HR=2.193, 95% CI 1.031-4.666, P=0.0414). Lack of α-DG immunoreactivity is more frequent in high-grade glioma and is an independent predictor of poor clinical outcome. Similarly, lack of both α-DG and ß-DG immunoreactivity is a strong independent predictor of clinical outcome.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Dystroglycans/metabolism , Glioma/metabolism , Neoplasm Recurrence, Local/metabolism , Adult , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Female , Follow-Up Studies , Glioma/mortality , Glioma/pathology , Humans , Immunoenzyme Techniques , Male , Matrix Metalloproteinase 9/metabolism , Neoplasm Grading , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Prognosis , Retrospective Studies , Survival Rate
14.
Indian J Dermatol ; 59(3): 225-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24891649

ABSTRACT

BACKGROUND: Caveolin-1 is a key structural and functional protein. Caveolin-1 is known to modulate multiple signal-transducing pathways involved in cell differentiation and proliferation. Psoriasis is viewed as a multifactorial pathology characterized by keratinocyte hyperproliferation and abnormal cell maturation. OBJECTIVES: To examine the expression of caveolin-1 in skin biopsies from normal subjects, patients, and subjects with the three respective isoforms of psoriasis (psoriasis vulgaris, localized pustular psoriasis, and erythrodermic psoriasis). The expression level of caveolin-1 was compared among psoriasis vulgaris, localized pustular psoriasis, erythrodermic psoriasis, and normal subjects. MATERIALS AND METHODS: Using immunohistochemical methods, caveolin-1 protein expression was assayed in four groups. An analysis was conducted on skin samples obtained from 22 normal subjects and 28 patients with psoriasis vulgaris, 22 patients with localized pustular psoriasis, and 16 patients with erythrodermic psoriasis. The statistical analysis of the scoring criteria reflecting the level of Caveolin-1 immunostaining between different groups was determined using the Mann-Whitney U-test. RESULTS: In the normal skin, intense and consistent caveolin-1 staining was present in 22 cases. The Caveolin-1 protein was significantly reduced and showed very weak or absent staining within the tissues of psoriasis vulgaris, localized pustular psoriasis, and erythrodermic psoriasis (respective P < 0.001). Caveolin-1 protein expression in psoriasis vulgaris was higher than that in localized pustular psoriasis and erythrodermic psoriasis (respective P < 0.05). Caveolin-1 protein expression was no different in localized pustular psoriasis and erythrodermic psoriasis (P > 0.05). CONCLUSION: The finding of this study was consistent with a downregulation of Caveolin-1, which might serve as an etiological factor in the development of psoriasis vulgaris, localized pustular psoriasis, and erythrodermic psoriasis. Further mechanistic investigations are required to prove that Caveolin-1 protein has the potential and may be a novel target for therapy of psoriasis vulgaris, localized pustular psoriasis, and erythrodermic psoriasis.

15.
Can J Neurol Sci ; 41(4): 498-503, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24878476

ABSTRACT

BACKGROUND: The objective of this study was to explore the expression of urotensin II (UII), its receptor (GPR14), and vascular endothelial growth factor (VEGF), as well as their associations in the ischaemic brains of rats with focal cerebral ischaemia, under normal and diabetic conditions. METHODS: Diabetes mellitus (DM) was induced by injection of streptozotocin (STZ) into Sprague - Dawley rats. Focal cerebral ischaemia was induced by middle cerebral artery occlusion (MCAO) four weeks after DM onset by STZ. Rats (n=80) were divided into four groups: normal control, DM, MCAO, and DM/MCAO. Immunohistochemistry and reverse-transcriptase-polymerase chain reaction (RT-PCR) were used to detect the expression of UII, GPR14 and VEGF in the diabetic and ischaemic brain. RESULTS: Expression of UII and GPR14 was increased at mRNA and protein levels in the DM and MCAO group compared with controls. In the DM/MCAO group, expression of UII and GPR14 was increased significantly in the ischaemic brain, and was accompanied by a significantly increased VEGF expression. CONCLUSION: Diabetes mellitus was seen to aggravate brain lesions after ischaemia, and UII may have an important role.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Receptors, G-Protein-Coupled/biosynthesis , Urotensins/biosynthesis , Animals , Gene Expression Regulation , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley
16.
Breast Cancer (Auckl) ; 6: 1-8, 2012.
Article in English | MEDLINE | ID: mdl-22259247

ABSTRACT

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.

17.
J Neurol Sci ; 270(1-2): 172-83, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18442833

ABSTRACT

Brain edema is one of the most frequent and serious complications of intracerebral hemorrhage (ICH), but how the ICH cause brain edema is unknown. Our studies were designed to investigate the regulation and distribution of protease nexin-1 (PN-1), thrombin and aquaporin-4 (AQP-4) in brain edema after ICH in rat and human brain in vivo. Our result showed that the severity of cerebral edema resulted from an acute stage of ICH. The PN-1-thrombin system modulated cerebral edema after ICH. Thrombin and AQP-4 increased to aggregate cerebral edema after ICH. In order to control the deleterious effect of thrombin's overexpression, PN-1 appeared quickly and abundantly to inhibit thrombin and lessen the cerebral edema. PN-1 was distributed in neurons and glial cells of cerebral cortex, hippocampus, thalamencephalon, basal ganglia, cerebellum and circum-encephalocoele in rat and human brain. The expression of AQP-4 is different between human and rat. Thus, we demonstrated that the animal experimental approach was, however, not sufficient by itself and needed to be corroborated by observations on human brains.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain Edema/etiology , Brain Edema/metabolism , Cerebral Hemorrhage/complications , Gene Expression Regulation/physiology , Receptors, Cell Surface/metabolism , Adult , Aged , Amyloid beta-Protein Precursor/genetics , Animals , Aquaporin 4/genetics , Aquaporin 4/metabolism , Cell Count/methods , Humans , Male , Middle Aged , Protease Nexins , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Serpin E2 , Statistics as Topic , Thrombin/genetics , Thrombin/metabolism , Time Factors , Water/metabolism
18.
J Alzheimers Dis ; 12(4): 335-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18198420

ABSTRACT

Growing evidence suggests a synergistic and perhaps etiological relationship between vascular disease and Alzheimer's disease (AD), which is characterized by the progressive accumulation of amyloid-beta peptide (Abeta). Moreover, apolipoprotein E (ApoE) has also been shown to be associated with AD and cerebral ischemia. It seems that cerebral ischemia may play an important, both direct and indirect, role in the pathogenesis of AD. We investigated the expression and distribution of Abeta1-40, beta1-42 and ApoE in human hippocampus after cerebral ischemia in this study to determine the role of cerebral ischemia in Alzheimer's disease. Our study has demonstrated that the accumulation of both Abeta1-40 and beta1-42 were increased dramatically and consistently after cerebral ischemia. Neuronal ApoE immunoreactivity was also significantly increased in all ischemic groups compared with controls. The most likely stimulus for the increased Abeta1-40, Abeta1-42 and ApoE immunoreactivity in the CA1 and CA3 neurons is the ischemic conditions, and their upregulation, in turn, may partly explain the contribution of cerebral ischemia to the pathogenesis of AD. Therefore our observations provide a basis for establishing therapeutic strategies aimed at preventing ischemic insults and subsequent neurodegeneration in AD.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Hippocampus/metabolism , Adult , Aged , Alzheimer Disease/pathology , Brain Ischemia/pathology , Female , Humans , Male , Middle Aged , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...