Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Transl Res ; 13(12): 13854-13861, 2021.
Article in English | MEDLINE | ID: mdl-35035725

ABSTRACT

OBJECTIVE: To investigate the clinical efficacy of coenzyme Q10 (CoQ10) plus trimetazidine (TMZ) in treating acute viral myocarditis (AVMC) and the combination's influence on the oxidative stress markers and the patients' quality of life (QoL). METHODS: This retrospective analysis enrolled 156 patients with AVMC admitted to the Department of Cardiology of the Affiliated Hospital of Chengdu University of Traditional Chinese Medicine between February 2018 and February 2019. Based on the treatment method each patient was administered, the patients were classified into a control group (n=72, CoQ10 therapy) and a combination group (n=84, CoQ10+TMZ therapy). The clinical effectiveness was observed in the two groups two weeks after the treatment, and the changes in the patients' serum inflammatory factor levels, oxidative stress indexes, myocardial enzyme levels, and cardiac function were compared. RESULTS: The combination group had a far superior total effective rate than the control group (90.5% vs. 77.8%, P<0.05). After the treatment, the serum inflammatory factor levels, including tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and C-reactive protein (CRP), decreased in both groups, and the index levels in the combination group were significantly better than they were in the control group (P<0.05). The oxidative stress indicators, such as superoxide dismutase (SOD), malondialdehyde (MDA) and nitric oxide (NO), improved more significantly in the combination group compared to the control group (P<0.05). The myocardial zymogram creatine kinase (CK), cardiac troponin (cTnI), creatine kinase isoenzyme MB (CK-MB), and lactate dehydrogenase (LDH) levels were reduced in the two groups, with lower levels in the combination group. The left ventricular systolic function and the patients' QoL were better in the combination group compared with the control group (P<0.05). CONCLUSIONS: CoQ10 plus TMZ yields a favorable clinical effectiveness in the treatment of AVMC, and it can effectively promote cardiac function recovery, alleviate oxidative stress and inflammatory reactions, and bolster patients' QoL.

2.
J Biol Chem ; 285(20): 15137-15148, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20223821

ABSTRACT

We have previously shown a novel link between hPar-1 (human protease-activated receptor-1) and beta-catenin stabilization. Although it is well recognized that Wnt signaling leads to beta-catenin accumulation, the role of PAR1 in the process is unknown. We provide here evidence that PAR1 induces beta-catenin stabilization independent of Wnt, Fz (Frizzled), and the co-receptor LRP5/6 (low density lipoprotein-related protein 5/6) and identify selective mediators of the PAR1-beta-catenin axis. Immunohistological analyses of hPar1-transgenic (TG) mouse mammary tissues show the expression of both Galpha(12) and Galpha(13) compared with age-matched control counterparts. However, only Galpha(13) was found to be actively involved in PAR1-induced beta-catenin stabilization. Indeed, a dominant negative form of Galpha(13) inhibited both PAR1-induced Matrigel invasion and Lef/Tcf (lymphoid enhancer factor/T cell factor) transcription activity. PAR1-Galpha(13) association is followed by the recruitment of DVL (Dishevelled), an upstream Wnt signaling protein via the DIX domain. Small interfering RNA-Dvl silencing leads to a reduction in PAR1-induced Matrigel invasion, inhibition of Lef/Tcf transcription activity, and decreased beta-catenin accumulation. It is of note that PAR1 also promotes the binding of beta-arrestin-2 to DVL, suggesting a role for beta-arrestin-2 in PAR1-induced DVL phosphorylation dynamics. Although infection of small interfering RNA-LRP5/6 or the use of the Wnt antagonists, SFRP2 (soluble Frizzled-related protein 2) or SFRP5 potently reduced Wnt3A-mediated beta-catenin accumulation, no effect was observed on PAR1-induced beta-catenin stabilization. Collectively, our data show that PAR1 mediates beta-catenin stabilization independent of Wnt. We propose here a novel cascade of PAR1-induced Galpha(13)-DVL axis in cancer and beta-catenin stabilization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Phosphoproteins/metabolism , Receptor, PAR-1/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Arrestins/metabolism , Cell Line , Dishevelled Proteins , Gene Silencing , Humans , Immunoprecipitation , Mice , Mice, Transgenic , Phosphoproteins/genetics , beta-Arrestin 2 , beta-Arrestins
3.
J Cell Physiol ; 218(3): 512-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19040205

ABSTRACT

Despite extensive efforts toward elucidation of the molecular pathway controlling cytotrophoblast (CTB) invasion to the uterine decidua, it remains poorly defined. There are striking similarities between tumor cell invasion and cytotrophoblast implantation to the deciduas whereby the role of Protease Activated Receptors (PARs) and wnt signaling is well recognized. We examine here consequences of modulation of PAR1 and PAR2 expression and function on CTB invasion and beta-catenin stabilization. Toward this end, we utilized a model system of extravillous trophoblast (EVT) organ culture and various placenta cell lines (e.g., JAR and HTR-8/Svneo). Activation of PAR1 induces EVT invasion while hPar1-SiRNA and PAR1 antagonist SCH79797--effectively inhibited it. In parallel, the Wnt inhibitor Dickkopf-1 (Dkk1) similarly inhibited it. Nuclear localization of beta-catenin is seen only after PAR1 activation, and is markedly reduced following the application of hPar1-SiRNA construct and PAR1 antagonist in CTBs. In contrast, PAR2 elicited a low cytoplasmic beta-catenin level as also proliferation and invasion. In the non-activated CTBs in-comparison, beta-catenin appeared limited to the membrane pools. Concomitantly, a temporal regulated pattern of Wnt-4, 5a, 7b, 10a, 10b expression is seen along PAR1 appearance. Enforced expression of Wnt antagonists, Secreted Frizzled Related Proteins; SFRP2 & 5; into HTR-8/Svneo, resulted with a markedly reduced nuclear beta-catenin levels, similar to the effect obtained by hPar1-SiRNA treatment. Identification of PAR1 downstream target/s may nonetheless contribute to the formation of a future platform system for eliciting a firm placenta-uterus interactions and to the definition of late pregnancy outcomes.


Subject(s)
Cell Movement , Receptor, PAR-1/metabolism , Trophoblasts/cytology , beta Catenin/metabolism , Cell Line , Cell Proliferation , Female , Gene Expression Regulation, Developmental , Gene Silencing , Humans , Ki-67 Antigen/metabolism , Organ Culture Techniques , Pregnancy , Pregnancy Trimester, First , Protein Stability , Protein Transport , RNA, Small Interfering/metabolism , Receptor, PAR-1/genetics , Trophoblasts/metabolism , Wnt Proteins/metabolism
4.
Cancer Res ; 66(10): 5224-33, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707447

ABSTRACT

Protease-activated receptor 1 (PAR1) is emerging with distinct assignments in tumor biology. We show that tissue targeted overexpression of hPar1 in mice mammary glands results in precocious hyperplasia, characterized by a dense network of ductal side branching and accelerated proliferation. These glands exhibit increased levels of wnt-4 and wnt-7b and a striking beta-catenin stabilization. Nuclear localization of beta-catenin is observed in hPar1 transgenic mouse tissue sections but not in the wild-type, age-matched counterparts. PAR1 induces beta-catenin nuclear localization also in established epithelial tumor cell lines of intact beta-catenin system (transformed on the background of mismatch repair system; RKO cells). We propose hereby that PAR1-mediated beta-catenin stabilization is taking place primarily via the increase of Wnt expression. Enforced expression of a specific Wnt antagonist family member, secreted frizzled receptor protein 5 (SFRP5), efficiently inhibited PAR1-induced beta-catenin stabilization. Likewise, application of either SFRP2 or SFRP5 on epithelial tumor cells completely abrogated PAR1-induced beta-catenin nuclear accumulation. This takes place most likely via inhibition of Wnt signaling at the level of cell surface (forming a neutralizing complex of "Receptors-SFRP-Wnt"). Furthermore, depletion of hPar1 by small interfering RNA (siRNA) vectors markedly inhibited PAR1-induced Wnt-4. The striking stabilization of beta-catenin, inhibited by SFRPs on one hand and Wnt-4 silencing by hPar1 siRNA on the other hand, points to a novel role of hPar1 in Wnt-mediated beta-catenin stabilization. This link between PAR1 and beta-catenin may bear substantial implications both in developmental and tumor progression processes.


Subject(s)
Mammary Glands, Animal/metabolism , Receptor, PAR-1/biosynthesis , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Female , Gene Silencing , Glycoproteins/biosynthesis , Glycoproteins/genetics , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Male , Mammary Glands, Animal/pathology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/genetics , Wnt Proteins/biosynthesis , Wnt Proteins/genetics , Wnt4 Protein
5.
Arterioscler Thromb Vasc Biol ; 23(6): 940-4, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12637343

ABSTRACT

While protease-activated receptors (PARs) play a traditional role in vascular biology, they emerge with surprisingly new assignments in tumor biology. PAR1 expression correlates with the invasion properties of breast carcinoma, whereas human PAR1 antisense reduces their ability to migrate through Matrigel. Part of the molecular mechanism of PAR1 invasion involves the formation of focal contact complexes on PAR1 activation. PAR1 induces angiogenesis in animal models in vivo and exhibits an oncogenic phenotype of enhanced ductal complexity when overexpressed in mouse mammary glands.


Subject(s)
Epithelial Cells/metabolism , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Receptor, PAR-1/physiology , Animals , Breast/blood supply , Breast/growth & development , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division , Cytoskeleton/ultrastructure , Epithelial Cells/pathology , Female , Humans , Integrins/physiology , Mice , Mice, Knockout , Morphogenesis , Neovascularization, Pathologic/physiopathology , Oligonucleotides, Antisense/pharmacology , Placenta/blood supply , Pregnancy , Receptors, Vitronectin/physiology , Thrombin/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
6.
FASEB J ; 17(2): 163-74, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12554695

ABSTRACT

The formation of new blood vessels is a critical determinant of tumor progression. We find that Par1 gene expression plays a central role in blood vessel recruitment in animal models. By in vivo injection of either Matrigel plugs containing Par1-expressing cells or of rat prostatic carcinoma cells transfected with tetracycline-inducible Par1 expression vectors, we show that Par1 significantly enhances both angiogenesis and tumor growth. Several vascular endothelial growth factor (VEGF) splice forms are induced in cells expressing Par1. Activation of PAR1 markedly augments the expression of VEGF mRNAs and of functional VEGFs as determined by in vitro assays for endothelial tube alignment and bovine aortic endothelial cell proliferation. Because neutralizing anti-VEGF antibodies potently inhibited Par1-induced endothelial cell proliferation, we conclude that Par1-induced angiogenesis requires VEGF. Specific inhibitors of protein kinase C (PKC), Src, and phosphatidylinositol 3-kinase (PI3K) inhibit Par1-induced VEGF expression, suggesting the participation of these kinases in the process. We also show that oncogenic transformation by genes known to be part of PAR1 signaling machinery is sufficient to increase VEGF expression in NIH 3T3 cells. These data support the novel notion that initiation of cell signaling either by activating PAR1 or by the activated forms of oncogenes is sufficient to induce VEGF and hence angiogenesis.


Subject(s)
Cell Transformation, Neoplastic , Neoplasms/blood supply , Neovascularization, Pathologic/physiopathology , Receptors, Thrombin/physiology , 3T3 Cells , Animals , Blotting, Northern , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Gene Expression , Genes, ras/genetics , Genes, src/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oncogenes/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, PAR-1 , Receptors, Thrombin/genetics , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...