Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 207: 115350, 2023 01.
Article in English | MEDLINE | ID: mdl-36435201

ABSTRACT

The pulmonary vascular remodeling (PVR), the pathological basis of pulmonary hypertension (PH), entails pulmonary artery smooth muscle cells (PASMCs) phenotypic switching, but appreciation of the underlying mechanisms is incomplete. Exosomes, a novel transfer machinery enabling delivery of its cargos to recipient cells, have been recently implicated in cardiovascular diseases including PH. The two critical questions of whether plasma-derived exosomes drive PASMCs phenotypic switching and what cargo the exosomes transport, however, remain unclear. Herein, by means of transmission electron microscopy and protein detection, we for the first time, characterized lectin like oxidized low-density lipoprotein receptor-1 (LOX-1) as a novel cargo of plasma-derived exosomes in PH. With LOX-1 knockout (Olr1-/-) rats-derived exosomes, we demonstrated that exosomal LOX-1 could be transferred into PASMCs and thus elicited cell phenotypic switching. Of importance, Olr1-/- rats exhibited no cell phenotypic switching and developed less severe PH, but administration of wild type rather than Olr1-/- exosomes to Olr1-/- rats recapitulated the phenotype of PH with robust PASMCs phenotypic switching. We also revealed that exosomal LOX-1 triggered PASMCs phenotypic switching, PVR and ultimately PH via ERK1/2-KLF4 signaling axis. This study has generated proof that plasma-derived exosomes confer PH by delivering LOX-1 into PASMCs. Hence, exosomal LOX-1 represents a novel exploitable target for PH prevention and treatment.


Subject(s)
Exosomes , Hypertension, Pulmonary , Rats , Animals , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Hypertension, Pulmonary/metabolism , Exosomes/metabolism , Cell Proliferation/physiology , Hypoxia/metabolism , Phenotype , Myocytes, Smooth Muscle/metabolism , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism , Cells, Cultured , Vascular Remodeling/physiology
2.
Biomed Res Int ; 2021: 2202888, 2021.
Article in English | MEDLINE | ID: mdl-34513987

ABSTRACT

The proliferation of pulmonary artery smooth muscle cells (PASMCs) is an important cause of pulmonary vascular remodeling in pulmonary hypertension (PH). It has been reported that miR-137 inhibits the proliferation of tumor cells. However, whether miR-137 is involved in PH remains unclear. In this study, male Sprague-Dawley rats were subjected to 10% O2 for 3 weeks to establish PH, and rat primary PASMCs were treated with hypoxia (3% O2) for 48 h to induce cell proliferation. The effect of miR-137 on PASMC proliferation and calpain-2 expression was assessed by transfecting miR-137 mimic and inhibitor. The effect of calpain-2 on PASMC proliferation was assessed by transfecting calpain-2 siRNA. The present study found for the first time that miR-137 was downregulated in pulmonary arteries of hypoxic PH rats and in hypoxia-treated PASMCs. miR-137 mimic inhibited hypoxia-induced PASMC proliferation and upregulation of calpain-2 expression in PASMCs. Furthermore, miR-137 inhibitor induced the proliferation of PASMCs under normoxia, and knockdown of calpain-2 mRNA by siRNA significantly inhibited hypoxia-induced proliferation of PASMCs. Our study demonstrated that hypoxia-induced downregulation of miR-137 expression promoted the proliferation of PASMCs by targeting calpain-2, thereby potentially resulting in pulmonary vascular remodeling in hypoxic PH.


Subject(s)
Calpain/genetics , Hypertension, Pulmonary/genetics , MicroRNAs/genetics , Animals , Calpain/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Hypertension, Pulmonary/pathology , Hypoxia/genetics , Hypoxia/metabolism , Male , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/metabolism , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Vascular Remodeling/genetics
3.
Cell Signal ; 87: 110149, 2021 11.
Article in English | MEDLINE | ID: mdl-34520855

ABSTRACT

Continuous recruitment and inappropriate activation of platelets in pulmonary arteries contribute to pulmonary vascular remodeling in pulmonary hypertension (PH). Our previous study has demonstrated that lectin like oxidized low-density lipoprotein receptor-1 (LOX-1) regulates the proliferation of pulmonary arterial smooth muscle cells (PASMCs). Phosphatidylserine exposed on the surface of activated platelets is a ligand for LOX-1. However, whether hypoxia-activated platelets stimulate the proliferation and migration of PASMCs by phosphatidylserine/LOX-1 signaling-impelled intercellular communication remains unclear. The present study found that rats treated with hypoxia (10% O2) for 21 days revealed PH with the activation of platelets and the recruitment of platelets in pulmonary arteries, and LOX-1 knockout inhibited hypoxia-induced PH and platelets activation. Notably, co-incubation of PASMCs with hypoxic PH rats-derived platelets up-regulated LOX-1 expression in PASMCs leading to the proliferation and migration of PASMCs, which was inhibited by the phosphatidylserine inhibitor annexin V or the LOX-1 neutralizing antibody. LOX-1 knockout led to decreased proliferation and migration of PASMCs stimulated by hypoxia-activated platelets. In rats, hypoxia up-regulated the phosphorylation of signal transducer and activator of transcription 3 (Stat3) and the expression of Pim-1 in pulmonary arteries. Hypoxia-activated platelets also up-regulated the phosphorylation of Stat3 and the expression of Pim-1 in PASMCs, which was inhibited by annexin V, the LOX-1 neutralizing antibody, the protein kinase C inhibitor and LOX-1 knockout. In conclusion, we for the first time demonstrated that hypoxia-activated platelets stimulated the proliferation and migration of PASMCs by phosphatidylserine/LOX-1/PKC/Stat3/Pim-1 signaling-impelled intercellular communication, thereby potentially contributing to hypoxic pulmonary vascular remodeling.


Subject(s)
Hypertension, Pulmonary , Pulmonary Artery , Animals , Blood Platelets/metabolism , Cell Communication , Cell Proliferation/physiology , Cells, Cultured , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphatidylserines/metabolism , Pulmonary Artery/metabolism , Rats , Scavenger Receptors, Class E
4.
Int J Mol Med ; 48(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34318904

ABSTRACT

Meis homeobox 1 (Meis1) was initially discovered in 1995 as a factor involved in leukemia in an animal model. Subsequently, 2 years later, MEIS1, the human homolog, was cloned in the liver and cerebellum, and was found to be highly expressed in myeloid leukemia cells. The MEIS1 gene, located on chromosome 2p14, encodes a 390­amino acid protein with six domains. The expression of homeobox protein MEIS1 is affected by cell type, age and environmental conditions, as well as the pathological state. Certain types of modifications of MEIS1 and its protein interaction with homeobox or pre­B­cell leukemia homeobox proteins have been described. As a transcription factor, MEIS1 protein is involved in cell proliferation in leukemia and some solid tumors. The present review article discusses the molecular biology, modifications, protein­protein interactions, as well as the role of MEIS1 in cell proliferation of cancer cells and MEIS1 inhibitors. It is suggested by the available literature MEIS1 has potential to become a cancer therapeutic target.


Subject(s)
Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy , Myeloid Ecotropic Viral Integration Site 1 Protein/analysis , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Protein Interaction Maps/drug effects
5.
Medicine (Baltimore) ; 99(26): e21018, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32590820

ABSTRACT

To study the correlation between single nucleotide polymorphism (SNP) of the 3' untranslated region (UTR) rs9722 locus in S100B and the risk of chronic heart failure (CHF), plasma levels of S100B protein as well as has-miR-340-3p in a Chinese Han population.A total of 215 patients with CHF (124 ischemic cardiomyopathy (ICM) and 91 dilated cardiomyopathy (DCM)) and 215 healthy controls were recruited to analyze the S100B rs9722 genotype by Sanger sequencing. The levels of hsa-miR-340-3p in the plasma were detected by RT-PCR, and S100B levels were detected by ELISA.The risk of CHF in S100B rs9722 locus T allele carriers was 4.24 times higher than that in those with the C allele (95% CI: 2.84-6.33, P < .001). The association of S100B rs9722 locus SNP with ICM and DCM risk was not affected by factors such as age, gender, and body mass index (BMI). The levels of plasma S100B and hsa-miR-340-3p in patients with ICM and DCM were significantly higher than those in the control group (P < .001). There was no significant difference in plasma S100B levels between patients with ICM and DCM (P > .05). Among ICM, DCM, and control subjects, TT genotype carriers had the highest levels of plasma S100B and hsa-miR-340-3p, followed by the CT genotype and TT genotype, and the difference was statistically significant (P < .05). Plasma hsa-miR-340-3p levels were positively correlated with S100B levels in the control subjects and patients with ICM and DCM.The S100B rs9722 locus SNP is associated with CHF risk in a Chinese Han population.


Subject(s)
3' Untranslated Regions/genetics , Heart Failure/genetics , Polymorphism, Single Nucleotide/genetics , S100 Calcium Binding Protein beta Subunit/analysis , Aged , Asian People/genetics , Biomarkers/analysis , Biomarkers/blood , Body Mass Index , Echocardiography/methods , Female , Heart Failure/epidemiology , Humans , Male , Middle Aged , S100 Calcium Binding Protein beta Subunit/blood
6.
Life Sci ; 255: 117822, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32450174

ABSTRACT

AIM: Proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) are regarded as the primary factors resulting in pulmonary arterial remodeling in pulmonary hypertension (PH). Myeloid ecotropic viral integration site 1 (MEIS1) has been positioned as a negative cardiomyocyte cell cycle regulator and regulates proliferation of multiple kinds of cancer cells. Whether MESI1 is involved in the proliferation and migration of PASMCs deserves to be identified. MAIN METHODS: Sprague Dawley rats were exposed to hypoxia condition (10% O2) for 4 weeks to induce PH and primary rat PASMCs were cultured in hypoxia condition (3% O2) for 48 h to induce proliferation and migration. Immunohistochemistry, immunofluorescence, reverse transcription PCR and Western blot analysis were performed to detect the expressions of target mRNAs and proteins. EDU, CCK8 and wound healing assays were conducted to measure the proliferation and migration of PASMCs. KEY FINDINGS: Hypoxia down-regulated the expression of MEIS1 (both mRNA and protein) in pulmonary arteries and PASMCs. Over-expression of MEIS1 inhibited the proliferation and migration of PASMCs afforded by hypoxia. In contrast, knockdown of MEIS1 under normoxia condition like hypoxia induced the proliferation and migration of PASMCs. MEIS1 mediated hypoxia-induced the proliferation and migration of PASMCs via METTL14/MEIS1/p21 signaling. SIGNIFICANCE: The present study revealed that MEIS1 regulated the proliferation and migration of PASMCs during hypoxia-induced PH. Thus, MEIS1 may be a potential target for PH therapy.


Subject(s)
Hypertension, Pulmonary/physiopathology , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/cytology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Cells, Cultured , Hypertension, Pulmonary/genetics , Hypoxia , Male , Muscle, Smooth, Vascular/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Vascular Remodeling/physiology
7.
Eur J Pharmacol ; 864: 172698, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31563647

ABSTRACT

It has well been demonstrated that E3 ubiquitin ligase cullin7 plays important roles in cancer cell growth control via down-regulating p53 expression. The noncanonical function or the pathogenic role of p53 has more recently been implicated in pulmonary vascular remodeling. Therefore, whether cullin7 participates in hypoxia-induced pulmonary vascular remodeling deserves to be elucidated. The present study found that hypoxia up-regulated the expression of cullin7 mRNA and protein in pulmonary arteries and pulmonary artery smooth muscle cells, and knockdown of cullin7 inhibited hypoxia-induced proliferation and migration of pulmonary artery smooth muscle cells and reversed hypoxia-induced inhibition of p53 expression. Notably, administration of proteasome inhibitor MG132 significantly inhibited the expression of cullin7 and up-regulated the expression of p53 in pulmonary arteries concomitantly with improvement of hypoxia-induced pulmonary vascular remodeling. Our study demonstrated that hypoxia induced up-regulation of cullin7 expression resulting to the proliferation and migration of pulmonary artery smooth muscle cells via down-regulating p53 expression, which contributed to pulmonary vascular remodeling.


Subject(s)
Cell Movement , Cullin Proteins/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/complications , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology , Up-Regulation , Animals , Cell Proliferation , Cullin Proteins/genetics , Gene Knockdown Techniques , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/metabolism , Leupeptins/pharmacology , Male , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Vascular Remodeling/drug effects
8.
Eur J Pharmacol ; 863: 172673, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31542480

ABSTRACT

Pulmonary arterial remodeling is a crucial cause of increased pulmonary artery pressure during pulmonary hypertension (PH). Recently, growing evidence has upheld the contribution of endothelial-mesenchymal transition (EndMT) to pulmonary arterial remodeling, but the underlying mechanisms remain largely unaddressed. miR-204 has been implicated in PH, being anti-proliferative and pro-apoptotic in pulmonary artery smooth muscles cells (PASMCs), but its role in EndMT is still unknown. Here we found that miR-204 was down-regulated by hypoxia in rat pulmonary arterial intima and human pulmonary artery endothelial cells (HPAECs), and its further down-regulation by using miR-204 inhibitor suppressed hypoxia-induced EndMT. Moreover, autophagy, evoked by hypoxia in rat pulmonary arterial intima and HPAECs, suppressed hypoxia-induced EndMT via p62-dependent degradation of Snail and Twist. Additionally, autophagy was regulated by miR-204 targeting ATG7. While down-regulation of miR-204 in PASMCs reportedly promoted monocrotaline-induced pulmonary arterial hypertension via increased cell proliferation, our data suggested an important, albeit dichotomous, role of miR-204 down-regulation in endothelial cells in the process of EndMT that it attenuated EndMT by enhancing autophagy, thereby ameliorating hypoxia-induced PH to some extent.


Subject(s)
Autophagy/genetics , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Autophagy-Related Protein 7/genetics , Cell Hypoxia/genetics , Cell Line , Humans , Male , Proteolysis , Rats , Rats, Sprague-Dawley , Snail Family Transcription Factors/metabolism , Twist-Related Protein 1/metabolism
9.
Life Sci ; 227: 64-73, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31004656

ABSTRACT

AIM: Growing evidence suggests that endothelial-mesenchymal transition (EndMT) play key roles in pulmonary arterial remodeling during pulmonary arterial hypertension (PAH), but the underlying mechanisms have yet to be fully understood. miR-27a has been shown to promote proliferation of pulmonary arterial cells during PAH, but its role in EndMT remains unexplored. This study was designed to investigate the role and underlying mechanism of miR-27a in EndMT during PAH. MAIN METHODS: Rats were exposed in hypoxia (10% O2) for 3 weeks to induce PAH, and human pulmonary artery endothelial cells (HPAECs) were exposed in hypoxia (1% O2) for 48 h to induce EndMT. Immunohistochemistry, in situ hybridization, immunofluorescence, real-time PCR and Western blot were conducted to detect the expressions of RNAs and proteins, and luciferase assay was used to verify the putative binding site of miR-27a. KEY FINDINGS: We found that hypoxia up-regulated miR-27a in the tunica intima of rat pulmonary arteries and HPAECs, and that inhibition of miR-27a suppressed hypoxia-induced EndMT. Furthermore, elevated expression of miR-27a suppressed bone morphogenetic protein (BMP) signaling by targeting Smad5, thereby lessening Id2-mediated repression of the 2 critical mediators of EndMT (Snail and Twist). SIGNIFICANCE: Our data unveiled a novel role of miR-27a in EndMT during hypoxia-induced PAH. Thus, targeting of miR-27a-related pathway may be therapeutically harnessed to treat PAH.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Hypertension, Pulmonary/genetics , MicroRNAs/physiology , Animals , Cell Culture Techniques , Cell Proliferation , Endothelial Cells/metabolism , Endothelium/metabolism , Gene Expression Regulation , Humans , Hypertension, Pulmonary/physiopathology , Hypoxia/genetics , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , Vascular Remodeling/genetics
10.
Asian Pac J Trop Med ; 7(8): 663-667, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25149383

ABSTRACT

OBJECTIVE: To investigate the expression of soluble vascular endothelial growth factor receptor-1 (sFlt-1) and placental growth factor (PLGF) in the fetal growth restriction (FGR) cases and the intervention mechanism of tetramethylpyrazine. METHODS: A total of 60 fetal growth restriction cases that admitted to our hospital were randomly divided into ligustrazine intervention group (group A) and nutritional support group (group B). A total of 50 healthy pregnant women were also enrolled as control group (group C). Expression level of maternal serum sFlt1, PLGF and fetal growth parameters including HC, AC, FL, BPD, EFW as well as placenta PLGF, sFlt-1 mRNA expression were recorded and compared among the three groups. A total of 15 SD rats were selected and were divided into three groups, TMP group, alcohol and tobacco group and blank control group. Three groups of rats were dissected on the twentieth day of gestation. RESULTS: Expression level of sFlt-1 and PLGF in group A was not significantly different from that of group C (P>0.05); but significant difference in SFlt1 and PLGF expression level was observed between group C and group B (P<0.05). Before treatment, HC, AC, FL, BPD and EFW of group A and group B were significant lower than those of group C, but after treatment, those parameters in group A were significantly improved (P<0.05). In the animal experiment there was no significant difference in sFlt-1 between treatment group and FGR group without treatment or control group (P>0.05). There was significant difference in PLGF between FGR group with treatment and FGR group without treatment or control group (P<0.01). CONCLUSIONS: PLGF level is decreased and sFlt-1 increased in patients suffered from fetal growth restriction, and FGR rats show increased sFlt-1 and decreased PLGF, thus they can be indicator of the fetal growth restriction. Ligustrazine can effectively improve sFlt-1, PLGF expression level in fetal growth restriction cases, which can be used as treatment for FGR.


Subject(s)
Fetal Development/drug effects , Fetal Growth Retardation/drug therapy , Pregnancy Proteins/blood , Pyrazines/pharmacology , Vascular Endothelial Growth Factor Receptor-1/blood , Animals , Female , Fetal Growth Retardation/metabolism , Humans , Placenta/drug effects , Placenta/metabolism , Placenta Growth Factor , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Pyrazines/therapeutic use , RNA, Messenger/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use
11.
Eur J Gynaecol Oncol ; 35(6): 724-7, 2014.
Article in English | MEDLINE | ID: mdl-25556281

ABSTRACT

OBJECTIVE: The aim of this study was to compare the characteristics of two orthotopic xenograft models established with human epithelial ovarian cancer solid tumor tissue slices and human ovarian carcinoma cell line OVCAR-3. MATERIALS AND METHODS: Tumor tissues and cell line OVCAR3 of human epithelial ovarian cancer were grown in subcutaneous tissue and the subcutaneous tumor source was fetched and inoculated in ovarian capsule of nude mice under microscope to establish the orthotopic implantation model. At four and eight weeks after modeling, the orthotopic tumor formation rate, tumor diameter, metastasis rate outside the ovary, incidence rate of ascites, and CA125 levels in the two models were observed. RESULTS: The orthotopic tumor formation rate in the solid tumor slices group (60.0%) was significantly lower than that in the cell line group (85.0%, p < 0.05). However, the tumor diameter, metastasis rate outside the ovary, incidence rate of ascites, and CA125 levels in the solid tumor slices group (2.4 +/- 0.61 cm, 75.0%, 50.0%, and 80.13 +/- 11.26 U/ml, respectively) were remarkably higher than those in the cell line group (1.6 +/- 0.53 cm, 52.9%, 29.4%, and 36.5 +/- 6.71 U/ml, respectively) (p < 0.05, respectively). CONCLUSION: There are differences between the two orthotopic xenograft models established with human epithelial ovarian cancer solid tumor tissue slices and human ovarian carcinoma cell line OVCAR-3. The biological characteristics of the solid tumor slices model are more similar to human ovarian cancer.


Subject(s)
Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Animals , CA-125 Antigen/blood , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Transplantation, Heterologous
12.
Asian Pac J Cancer Prev ; 13(9): 4669-75, 2012.
Article in English | MEDLINE | ID: mdl-23167400

ABSTRACT

OBJECTIVE: Nude mice with orthotopic transplantation of human ovarian epithelial cancer were used to investigate screening criteria for paraneoplastic normal ovarian tissue and the security of the freezing and thawing for ovarian tissue transplantation. METHODS: Expression of CK-7, CA125, P53, survivin, MMP-2/TIMP- 2 in paraneoplastic normal ovarian tissues were detected by RT-PCR as well as immunohistochemistry. The tissues of the groups with all negative indicators of RT-PCR, all negative indicators of immunohistochemistry, negative expression of CK-7, CA125 and survivin, positive expression of CK-7, CA125 and survivin, cancer tissues and normal ovarian tissues of nude mice were used for freezing and thawing transplantation, to analyze overt and occult carcinogenesis rates after transplantation. RESULTS: When all indicators or the main indicators, CK-7, CA125 and survivin, were negative, tumorigenesis did not occur after transplantation. In addition the occult carcinogenesis rate was lower than in the group with positive expression of CK-7, CA125 and survivin (P<0.01). After subcutaneous and orthotopic transplantation of ovarian tissues, rates did not change (P>0.05). There was no statistical significance among rates after transplantation of ovarian tissues which were obtained under different severity conditions (P>0.05). CONCLUSION: Negative expression of CK-7, CA125 and survivin can be treated as screening criteria for security of ovarian tissues for transplantation. Immunohistochemical methods can be used as the primary detection approach. Both subcutaneous and orthotopic transplantation are safe. The initial severity does not affect the carcinogenesis rate after tissue transplantation. Freezing and thawing ovarian tissue transplantation in nude mice with human epithelial ovarian carcinoma is feasible and safe.


Subject(s)
Cryopreservation , Neoplasms, Glandular and Epithelial/diagnosis , Ovarian Neoplasms/diagnosis , Ovary/pathology , Replantation/adverse effects , Animals , CA-125 Antigen/genetics , CA-125 Antigen/metabolism , Carcinoma, Ovarian Epithelial , Female , Fertility Preservation , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Keratin-7/genetics , Keratin-7/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Glandular and Epithelial/etiology , Neoplasms, Glandular and Epithelial/surgery , Ovarian Neoplasms/etiology , Ovarian Neoplasms/surgery , Ovary/metabolism , Ovary/surgery , Reverse Transcriptase Polymerase Chain Reaction , Survivin , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Yao Xue Xue Bao ; 47(5): 609-13, 2012 May.
Article in Chinese | MEDLINE | ID: mdl-22812004

ABSTRACT

This study is to investigate the effect and mechanism of puerarin on DNA damage of HaCaT cells induced by UVB. Puerarin pre-treated cells were irradiated with UVB at 30 mJ x cm(-2). Twenty four hours after irradiation, DNA damage was detected by comet assay, ceramide was measured by thin layer chromatography and gas chromatography, intracellular free calcium ion was analyzed by flow cytometry, the phosphorylation level of p38 protein was examined by Western blotting method. Levels of DNA damage, ceramide, free calcium ion and p-p38 protein were elevated in UVB model cells. Contrary to the model group, all indicators above were reduced in all groups pre-treated by puerarin. Puerarin restrains the ceramide accumulation to block downstream p38 MAPK pathway and calcium ion rising, therefore reduces DNA damage in HaCaT cells induced by UVB.


Subject(s)
Ceramides/metabolism , DNA Damage/drug effects , Isoflavones/pharmacology , Keratinocytes/cytology , Calcium/metabolism , Cell Line , DNA Damage/radiation effects , Down-Regulation , Humans , Keratinocytes/metabolism , Phosphorylation , Signal Transduction/drug effects , Ultraviolet Rays/adverse effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...