Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Zhonghua Wai Ke Za Zhi ; 56(12): 957-960, 2018 Dec 01.
Article in Chinese | MEDLINE | ID: mdl-30497123

ABSTRACT

Acute aortic syndrome(AAS) is a lethal disease with acute onset and a high mortality rate as well as a higher incidence rate especially in an aging population. The diagnostic techniques of AAS have been improving in recent years. Many serum biomarkers have been shown to have the potential of further clinical implication. Advancement of imaging techniques has also improved the accuracy of early diagnosis. Although traditional treatment modality involving open surgery is life-saving, it still has a high mortality rate and a high major morbidity rate. The increasing utilization of endovascular techniques has greatly improved the prognosis of AAS, while it still need further optimization to be applied in different subgroups of patients.


Subject(s)
Aortic Aneurysm , Aortic Dissection , Acute Disease , Aortic Dissection/diagnosis , Aortic Dissection/surgery , Aortic Aneurysm/diagnosis , Aortic Aneurysm/surgery , Aortic Diseases , Hematoma , Humans
2.
Exp Cell Res ; 361(2): 201-209, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29056521

ABSTRACT

Hypertrophic scar (HS) formation is associated with the fibrosis of fibrocytes caused by excessive extracellular matrix (ECM) synthesis and deposition, the initial event of HS formation. Our high throughput screen of miRNA expression profiles identified hsa-miR31-5p, whose transcription level was most differentially in normal skin fibroblasts (NS) and HS among other miRNAs. The level of hsa-miR31-5p in HS was significantly higher than in NS. In-vitro functional experiments showed hsa-miR31-5p knockdown remarkably suppressed the proliferation of hypertrophic scar fibroblasts (HSFBs) under hypoxia, promoted cell invasion, and inhibited the expression of Collagen I and III and Fibronectin (FN), suggesting that hsa-miR31-5p knockdown effectively reduces HS formation caused by excessive ECM synthesis and deposition in HSFBs under hypoxia. Mechanism study showed that the regulation of HS formation by hsa-miR31-5p was mediated by its target gene, factor-inhibiting HIF-1 (FIH): under hypoxia, hsa-miR31-5p down-regulated FIH and thus increased the level of hypoxia inducible factor-1α (HIF-1α), which subsequently activated the HIF-1α fibrosis regulation pathway in HSFBs, and stimulated the proliferation and ECM synthesis in HSFBs, eventually resulting in fibrosis and scar formation. The data also show that knockdown of hsa-miR31-5p in HSFBs impaired the trend of increased proliferation, reduced invasion and excessive ECM synthesis and deposition caused by HIF-1a activation under hypoxia through upregulating FIH, indicating that knockdown of hsa-miR31-5p effectively inhibits the formation of HS. In conclusion, hsa-miR31 -5p plays an important role in HS formation by inhibiting FIH and regulating the HIF-1α pathway. Therefore, hsa-miR31 -5p may be a novel therapeutic target for HS.


Subject(s)
Antagomirs/genetics , Cicatrix, Hypertrophic/genetics , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/genetics , Mixed Function Oxygenases/genetics , Repressor Proteins/genetics , Antagomirs/metabolism , Cell Hypoxia , Cell Movement , Cell Proliferation , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/pathology , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Mixed Function Oxygenases/metabolism , Primary Cell Culture , Repressor Proteins/metabolism , Signal Transduction
3.
Ann Oncol ; 28(9): 2135-2141, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28911083

ABSTRACT

BACKGROUND: Distant metastasis accounts for 90% of deaths from colorectal cancer (CRC). Genomic heterogeneity has been reported in various solid malignancies, but remains largely under-explored in metastatic CRC tumors, especially in primary to metastatic tumor evolution. PATIENTS AND METHODS: We conducted high-depth whole-exome sequencing in multiple regions of matched primary and metastatic CRC tumors. Using a total of 28 tumor, normal, and lymph node tissues, we analyzed inter- and intra-individual heterogeneity, inferred the tumor subclonal architectures, and depicted the subclonal evolutionary routes from primary to metastatic tumors. RESULTS: CRC has significant inter-individual but relatively limited intra-individual heterogeneity. Genomic landscapes were more similar within primary, metastatic, or lymph node tumors than across these types. Metastatic tumors exhibited less intratumor heterogeneity than primary tumors, indicating that single-region sequencing may be adequate to identify important metastasis mutations to guide treatment. Remarkably, all metastatic tumors inherited multiple genetically distinct subclones from primary tumors, supporting a possible polyclonal seeding mechanism for metastasis. Analysis of one patient with the trio samples of primary, metastatic, and lymph node tumors supported a mechanism of synchronous parallel dissemination from the primary to metastatic tumors that was not mediated through lymph nodes. CONCLUSIONS: In CRC, metastatic tumors have different but less heterogeneous genomic landscapes than primary tumors. It is possible that CRC metastasis is, at least partly, mediated through a polyclonal seeding mechanism. These findings demonstrated the rationale and feasibility for identifying and targeting primary tumor-derived metastasis-potent subclones for the prediction, prevention, and treatment of CRC metastasis.


Subject(s)
Colorectal Neoplasms/pathology , Exome Sequencing , Genetic Heterogeneity , Neoplasm Metastasis/genetics , Colorectal Neoplasms/genetics , Humans , Mutation , Neoplasm Seeding
4.
Oncogene ; 36(18): 2577-2588, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28192397

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been recognized as a key element of cell migration and invasion in lung cancer; however, the underlying mechanisms are not fully elucidated. Recently, emerging evidence suggest that miRNAs have crucial roles in control of EMT and EMT-associated traits such as migration, invasion and chemoresistance. Here, we found that miR-218 expression levels were significantly downregulated in lung cancer tissues compared with adjacent non-cancerous tissues, and the levels of miR-218 were significantly associated with histological grades and lymph node metastasis. Overexpression of miR-218 inhibited cell migration and invasion as well as the EMT process. Of particular importance, miR-218 was involved in the metastatic process of lung cancer cells in vivo by suppressing local invasion and distant colonization. We identified Slug and ZEB2 as direct functional targets of miR-218. Inverse correlations were observed between miR-218 levels and Slug/ZEB2 levels in cancer tissue samples. In addition, overexpression of miR-218 in H1299 increased chemosensitivity of cells to cisplatin treatment through suppression of Slug and ZEB2. These findings highlight an important role of miR-218 in the regulation of EMT-related traits and metastasis of lung cancer in part by modulation of Slug/ZEB2 signaling, and provide a potential therapeutic strategy by targeting miR-218 in NSCLC.


Subject(s)
Homeodomain Proteins/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Repressor Proteins/genetics , Snail Family Transcription Factors/genetics , A549 Cells , Animals , Cell Movement/genetics , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Xenograft Model Antitumor Assays , Zinc Finger E-box Binding Homeobox 2
5.
Oncogenesis ; 6(1): e287, 2017 Jan 16.
Article in English | MEDLINE | ID: mdl-28092369

ABSTRACT

A subset of clear cell renal cell carcinoma (ccRCC) tumors exhibit a HIF1A gene mutation, yielding two ccRCC tumor types, H1H2 type expressing both HIF1α and HIF2α, and H2 type expressing HIF2α, but not functional HIF1α protein. However, it is unclear how the H1H2 type ccRCC tumors escape HIF1's tumor-suppressive activity. The polybromo-1 (PBRM1) gene coding for the BAF180 protein, a component of the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex, is inactivated in 40% ccRCCs, the function and mechanism of BAF180 mutation is unknown. Our previous study indicates that BAF180-containing SWI/SNF chromatin remodeling complex is a co-activator for transcription factor HIF to induce HIF target genes. Thus, our questions are if BAF180 is involved in HIF-mediated hypoxia response and if PBRM1/BAF180 mutation has any association with the HIF1A retention in H1H2 type ccRCC. We report here that BAF180 is mutated in H1H2 ccRCC cell lines and tumors, and BAF180 re-expression in H1H2 ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-suppressive role in these cells. However, BAF180 is expressed in HIF1-deficient H2 ccRCC cell lines and tumors, and BAF180 knockdown in H2 type ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-promoting activity in these cells. In addition, our data show that BAF180 functions as co-activator for HIF1- and HIF2-mediated transcriptional response, and BAF180's tumor-suppressive and -promoting activity in ccRCC cell lines depends on co-expression of HIF1 and HIF2, respectively. Thus, our studies reveal that BAF180 function in ccRCC is context dependent, and that mutation of PBRM1/BAF180 serves as an alternative strategy for ccRCC tumors to reduce HIF1 tumor-suppressive activity in H1H2 ccRCC tumors. Our studies define distinct functional subgroups of ccRCCs based on expression of BAF180, and suggest that BAF180 inhibition may be a novel therapeutic target for patients with H2, but not H1H2, ccRCC tumors.

6.
Cell Death Dis ; 7: e2216, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27148689

ABSTRACT

Kv2.1 as a voltage-gated potassium (Kv) channel subunit has a pivotal role in the regulation of glucose-stimulated insulin secretion (GSIS) and pancreatic ß-cell apoptosis, and is believed to be a promising target for anti-diabetic drug discovery, although the mechanism underlying the Kv2.1-mediated ß-cell apoptosis is obscure. Here, the small molecular compound, ethyl 5-(3-ethoxy-4-methoxyphenyl)-2-(4-hydroxy-3-methoxybenzylidene)-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyrimidine-6-carboxylate (SP6616) was discovered to be a new Kv2.1 inhibitor. It was effective in both promoting GSIS and protecting ß cells from apoptosis. Evaluation of SP6616 on either high-fat diet combined with streptozocin-induced type 2 diabetic mice or db/db mice further verified its efficacy in the amelioration of ß-cell dysfunction and glucose homeostasis. SP6616 treatment efficiently increased serum insulin level, restored ß-cell mass, decreased fasting blood glucose and glycated hemoglobin levels, and improved oral glucose tolerance. Mechanism study indicated that the promotion of SP6616 on ß-cell survival was tightly linked to its regulation against both protein kinases C (PKC)/extracellular-regulated protein kinases 1/2 (Erk1/2) and calmodulin(CaM)/phosphatidylinositol 3-kinase(PI3K)/serine/threonine-specific protein kinase (Akt) signaling pathways. To our knowledge, this may be the first report on the underlying pathway responsible for the Kv2.1-mediated ß-cell protection. In addition, our study has also highlighted the potential of SP6616 in the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Potassium Channel Blockers/pharmacology , Pyrimidines/pharmacology , Shab Potassium Channels/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Thiazoles/pharmacology , Animals , CHO Cells , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Calmodulin/metabolism , Cell Line , Cell Survival/drug effects , Cricetulus , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Drug Discovery , Gene Expression Regulation , Glucose Tolerance Test , Hypoglycemic Agents/chemistry , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Potassium Channel Blockers/chemistry , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/chemistry , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , Streptozocin , Thiazoles/chemistry
7.
Oncogene ; 34(43): 5482-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25703326

ABSTRACT

Upregulation of the embryonic M2 isoform of pyruvate kinase (PKM2) emerges as a critical player in the cancer development and metabolism, yet the underlying mechanism of PKM2 overexpression remains to be elucidated. Here we demonstrate that IGF-1/IGF-IR regulates PKM2 expression by enhancing HIF-1α-p65 complex binding to PKM2 promoter. PKM2 expression is regulated by miR-148a/152 suppression. PKM2 directly interacts with NF-κB p65 subunit to promote EGR1 expression for regulating miR-148a/152 feedback circuit in normal cells, but not in cancer cells because of the DNA hypermethylation of miR-148a and miR-152 gene promoters. The silencing of miR-148a/152 contributes to the overexpression of PKM2, NF-κB or/and IGF-IR in some cancer cells. We show that disruption of PKM2/NF-κB/miR-148a/152 feedback loop can regulate cancer cell growth and angiogenesis, and is also associated with triple-negative breast cancer (TNBC) phenotype, which may have clinical implication for providing novel biomarker(s) of TNBC and potential therapeutic target(s) in the future.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , NF-kappa B/genetics , Neovascularization, Pathologic/genetics , Thyroid Hormones/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Biomarkers, Tumor/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation/genetics , Disease Progression , Early Growth Response Protein 1/genetics , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , Up-Regulation/genetics , Thyroid Hormone-Binding Proteins
8.
Cell Death Dis ; 5: e1407, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25188517

ABSTRACT

Ovarian cancer is a leading cause of cancer death as diagnosis is frequently delayed to an advanced stage. Effective biomarkers and screening strategies for early detection are urgently needed. In the current study, we identify PSP94 as a key upstream factor in mediating prostasin (a protein previously reported to be overexpressed in ovarian cancer) signaling that regulates prostasin expression and action in ovarian cancer cells. PSP94 is overexpressed in ovarian cancer cell lines and patients, and is significantly correlated with prostasin levels. Signaling pathway analysis demonstrated that both PSP94 and prostasin, as potential upstream regulators of the Lin28b/Let-7 pathway, regulate Lin28b and its downstream partner Let-7 in ovarian cancer cells. Expression of PSP94 and prostasin show a strong correlation with the expression levels of Lin28b/Let-7 in ovarian cancer patients. Thus, PSP94/prostasin axis appears to be linked to the Lin28b/Let-7 loop, a well-known signaling mechanism in oncogenesis in general that is also altered in ovarian cancer. The findings suggest that PSP94 and PSP94/prostasin axis are key factors and potential therapeutic targets or early biomarkers for ovarian cancer.


Subject(s)
Ovarian Neoplasms/pathology , Prostatic Secretory Proteins/metabolism , Serine Endopeptidases/metabolism , Biomarkers, Tumor/blood , Cell Line, Tumor , Female , Humans , MicroRNAs/metabolism , Neoplasm Staging , Ovarian Neoplasms/metabolism , Prostatic Secretory Proteins/antagonists & inhibitors , Prostatic Secretory Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction
9.
Theriogenology ; 80(9): 1088-96, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055399

ABSTRACT

Understanding the performances of cloned pigs and their offspring is critical to evaluate the practical applications of somatic cell nuclear transfer. In this study, genetic polymorphism, growth performance, hematological parameters, and reproduction characteristics of cloned Landrace boars were compared with those of controls. In addition, the growth performance of clone offspring was also evaluated. A total of 479 reconstructed embryos were transferred to five recipient pigs and resulted in the delivery of 14 piglets (overall cloning of 2.9%) from two litters. Analyses of microsatellite markers and polymorphisms of the specific genes confirmed that the 14 clones were genetically identical to the nuclear donor and maintained the desirable genotypes. Growth performance of five healthy, phenotypically normal cloned boars from one litter and eight of their male offspring did not differ from age, breed, and management-matched controls. Although some significant differences were observed between cloned and control boars in hematological and serum enzymes, most of these parameters were within the normal range. Cloned boars had less (P < 0.05) normal sperm in the ejaculated boars than in control boars (71.4% vs. 77.9%, respectively), but sperm production (ejaculate volume, sperm concentration, and total sperm) did not differ between these groups. In addition, use of frozen-thawed semen from cloned boars for insemination produced results that seemed comparable to a control. In conclusion, the present study reported that somatic cell nuclear transfer is effective in reproducing preferred genetic traits and has potential applications to conserve elite bloodlines in a routine pig breeding program.


Subject(s)
Nuclear Transfer Techniques/veterinary , Swine/genetics , Animals , Breeding , Cloning, Organism/veterinary , Embryo Culture Techniques/veterinary , Enzymes/blood , Male , Polymorphism, Genetic , Reproduction , Semen Analysis , Swine/blood
10.
Eur Respir J ; 37(3): 578-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20595149

ABSTRACT

Pulmonary hypertension (PH) and right ventricular (RV) dysfunction associated with heart failure (HF) carry a poor prognosis. Although endothelin receptor antagonists (ERAs) demonstrated benefits in pulmonary arterial hypertension, their efficacy in PH associated with HF was not specifically evaluated. 2 weeks after myocardial infarction (MI) rats received bosentan (100 or 200 mg·kg(-1)·day(-1)) or no treatment for 3 weeks. PH, RV hypertrophy and function as well as lung remodeling and function were evaluated. LV echocardiographic wall motion abnormality and function measured before treatment (2 weeks after MI) and after treatment (5 weeks after MI) were similar in MI control and MI treatment groups. HF induced PH and RV hypertrophy compared with sham: RV systolic pressure 39±5 versus 23±0.8 mmHg and RV/left ventricular+septum weight 52±7 versus 24±0.5% (all p<0.01). Bosentan did not significantly modify these parameters. In addition, bosentan did not improve depressed RV function measured by echocardiograph from the RV myocardial performance index and tricuspid annular plane systolic excursion. The respiratory pressure-volume relationship revealed that HF caused a restrictive lung syndrome with histological lung remodeling and fibrosis, also not improved by bosentan. Dual ERA therapy with bosentan does not reduce PH, RV hypertrophy and lung remodeling and dysfunction associated with ischaemic HF.


Subject(s)
Heart Failure/drug therapy , Hypertension, Pulmonary/drug therapy , Sulfonamides/pharmacology , Animals , Antihypertensive Agents/pharmacology , Bosentan , Disease Models, Animal , Echocardiography/methods , Endothelin Receptor Antagonists , Heart Failure/complications , Heart Ventricles/pathology , Hemodynamics , Hypertension, Pulmonary/complications , Lung/pathology , Lung/physiopathology , Myocardial Infarction/pathology , Prognosis , Rats
11.
Phytomedicine ; 17(8-9): 606-13, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20092987

ABSTRACT

To elucidate the cytotoxicity mechanism of Ganoderma triterpenes, a chemoproteomic study using five purified ganoderic acids, ganoderic acid F (GAF), ganoderic acid K (GAK), ganoderic B (GAB), ganoderic acid D (GAD) and ganoderic acid AM1 (GAAM1) was conducted. GAF, GAK, GAB, GAD and GAAM1 treatment for 48 h inhibited the proliferation of HeLa human cervical carcinoma cells with IC(50) values of 19.5+/-0.6 microM, 15.1+/-0.5 microM, 20.3+/-0.4 microM, 17.3+/-0.3 microM, 19.8+/-0.7 microM, respectively. The protein expression profiles of HeLa cells treated with each ganoderic acid at dose of 15 microM for 48 h were checked using two-dimensional electrophoresis (2-DE). The possible target-related proteins of ganoderic acids, i.e. proteins with same change tendency in all five ganoderic acids-treated groups compared with control, were identified using MALDI-TOF MS/MS. Twelve proteins including human interleukin-17E, eukaryotic translation initiation factor 5A (eIF5A), peroxiredoxin 2, ubiquilin 2, Cu/Zn-superoxide dismutase, 14-3-3 beta/alpha, TPM4-ALK fusion oncoprotein type 2, PP2A subunit A PR65-alpha isoform, nucleobindin-1, heterogeneous nuclear ribonucleoprotein K, reticulocalbin 1 and chain A of DJ-1 protein were identified. Ganoderic acids might exert their cytotoxicity by altering proteins involved in cell proliferation and/or cell death, carcinogenosis, oxidative stress, calcium signaling and ER stress.


Subject(s)
Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Carcinoma/metabolism , Proteome/drug effects , Reishi/chemistry , Triterpenes/pharmacology , Uterine Cervical Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Carcinoma/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Electrophoresis , Female , HeLa Cells , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Humans , Inhibitory Concentration 50 , Phytotherapy , Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Triterpenes/therapeutic use , Uterine Cervical Neoplasms/drug therapy
12.
Eur Respir J ; 20(1): 52-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12166581

ABSTRACT

The purpose of the present study was to determine the relationship between hypertensive pulmonary vascular remodelling and the changes in mean pulmonary artery pressure (mPAP) during low-dose nitric oxide (NO) inhalation. Rats were exposed to chronic hypobaric hypoxia (air at 50.5 kPa (380 mmHg), 10% oxygen, for 5-29 days) to induce chronic pulmonary hypertension (PH) with pulmonary vascular structural changes. After the chronic hypoxic exposure, the rats had an indwelling pulmonary artery catheter inserted and changes in mPAP with NO were correlated to morphometrical analysis of pulmonary vascular changes. All concentrations of inhaled NO (0.1-2.0 parts per million) reduced mPAP with a similar per cent reduction from baseline mPAP in PH rats, while no changes were observed in control rats. During NO inhalation in PH rats, the absolute value of the decrease in mPAP, but not per cent reduction in mPAP, significantly correlated with baseline mPAP, the percentage of muscularised arteries at the alveolar wall level and at the alveolar duct level, and the per cent medial wall thickness of muscularised arteries. In the chronic hypoxic pulmonary hypertension model, the severity of pulmonary vascular remodelling did not alter the reactivity of the pulmonary arteries to nitric oxide and might, in part, determine the magnitude of nitric-oxide induced absolute reduction in mean pulmonary artery pressure.


Subject(s)
Bronchodilator Agents/administration & dosage , Bronchodilator Agents/pharmacology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Nitric Oxide/administration & dosage , Nitric Oxide/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Administration, Inhalation , Animals , Aorta/drug effects , Aorta/pathology , Aorta/physiopathology , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension, Pulmonary/pathology , Hypoxia/pathology , Male , Pulmonary Artery/pathology , Pulmonary Circulation/drug effects , Pulmonary Circulation/physiology , Rats , Rats, Sprague-Dawley , Severity of Illness Index
13.
Oncogene ; 20(45): 6607-16, 2001 Oct 04.
Article in English | MEDLINE | ID: mdl-11641786

ABSTRACT

The actin filament-associated protein of 110 kDa (AFAP-110) is a Src binding partner that represents a potential modulator of actin filament integrity in response to cellular signals. Previous reports have demonstrated that AFAP-110 is capable of directly binding and altering actin filaments. Deletion of the leucine zipper motif of AFAP-110 (AFAP-110(Deltalzip)) has been shown to induce a phenotype which resembles Src-transformed cells, by repositioning actin filaments into rosettes. This deletion also mimics a conformational change in AFAP-110 that is detected in Src-transformed cells. The results presented here indicate that unlike AFAP-110, AFAP-110(Deltalzip) is capable of activating cellular tyrosine kinases, including Src family members, and that AFAP-110(Deltalzip) itself is hyperphosphorylated. The newly tyrosine phosphorylated proteins and activated Src-family members appear to be associated with actin-rich lamellipodia. A point mutation that alters the SH3-binding motif of AFAP-110(Deltalzip) prevents it from activating tyrosine kinases and altering actin filament integrity. In addition, a deletion within a pleckstrin homology (PH) domain of AFAP-110(Deltalzip) will also revert its effects upon actin filaments. Lastly, dominant-positive RhoA(V14) will block the ability of AFAP-110(Deltalzip) from inducing actin filament rosettes, but does not inhibit Src activation. Thus, conformational changes in AFAP-110 enable it to activate cellular kinases in a mechanism requiring SH3 and/or PH domain interactions. We hypothesize that cellular signals which alter AFAP-110 conformation, enable it to activate cellular kinases such as cSrc, which then direct changes in actin filament integrity in a Rho-dependent fashion.


Subject(s)
Actin Cytoskeleton/ultrastructure , Microfilament Proteins/physiology , Phosphoproteins/physiology , src-Family Kinases/metabolism , Animals , COS Cells , Cytoplasm/metabolism , Cytoskeleton/ultrastructure , Enzyme Activation , Leucine Zippers , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microscopy, Fluorescence , Models, Biological , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Phosphotyrosine/metabolism , Transfection , rhoA GTP-Binding Protein/physiology , src Homology Domains
14.
Mol Cell Biochem ; 222(1-2): 199-204, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11678602

ABSTRACT

While Cr (VI)-containing compounds are well established carcinogens, the mechanisms of their action remain to be investigated. In this study we show that Cr (VI) causes increased tyrosine phosphorylation in human lung epithelial A549 cells in a time-dependent manner. N-acetyl-cysteine (NAC), a general antioxidant, inhibited Cr (VI)-induced tyrosine phosphorylation. Catalase, a scavenger of H2O2, sodium formate and aspirin, scavengers of hydroxyl radical (*OH), also inhibited the increased tyrosine phosphorylation induced by Cr (VI). SOD, an inhibitor of superoxide radical (O2*-), caused less inhibition. ESR study shows that incubation of Cr (VI) with the A549 cells generates *OH radical. The generation of radical was decreased by addition of catalase and sodium formate, while SOD did not have any inhibitory effect. Oxygen consumption measurements show that addition of Cr (VI) to A549 cells resulted in enhanced molecular oxygen consumption. These results indicate that Cr (VI) can induce an increase in tyrosine phosphorylation. H2O2 and *OH radicals generated during the process are responsible for the increased tyrosine phosphorylation induced by Cr (VI).


Subject(s)
Chromium/pharmacology , Hydrogen Peroxide/metabolism , Hydroxyl Radical/metabolism , Protein-Tyrosine Kinases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Catalase/pharmacology , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Formates/pharmacology , Hemostatics/pharmacology , Humans , Lung/cytology , Lung/drug effects , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Time Factors
15.
J Clin Endocrinol Metab ; 86(9): 4284-91, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549663

ABSTRACT

We examined GH and GH receptor expression in human leukemic cell lines and leukocytes of normal subjects to elucidate the cell types expressing GH and GH receptor, the individual variations of their expressions, their correlation and the relationships with serum IgG and IGF-I concentrations. In addition, the expression of GH secretagogue receptor, which enhances GH secretion from the anterior pituitary by synthetic GH secretagogues and that of its endogenous ligand, ghrelin, were also examined in these immune cells. GH expression in human leukemic cell lines was observed mainly in B cell lines at both the mRNA and protein level [3.8 +/- 0.2 pg/10(6) cells in Raji and 19.9 +/- 3.3 pg/10(6) cells in Daudi vs. negligible in T cell lines (Jurkat and Hut-78) and in myeloid cell lines (K-562 and HL-60)]. B cells in normal subjects were also found to be the major immune cells expressing GH mRNA, with significant individual variation. GH receptor mRNA expression was detectable in all human leukemic cell lines, although the expression level varied widely among the cell lines and was weaker than that in the liver. On the other hand, GH receptor mRNA expression was mainly found in B cells, with marked individual variation in normal subjects. There was a positive correlation between the mRNA expressions of GH and GH receptor in B cells of normal subjects (r = 0.89; P < 0.001). Single cell RT-PCR revealed that some B cells expressed both GH and GH receptor transcripts, and others expressed only GH. GH/GH receptor expression levels in B cells did not show any correlation with serum IgG and IGF-I levels in normal subjects. Expression of GH secretagogue receptor and ghrelin was detectable in all immune cells regardless of the maturity and cell types with great individual variations. In summary, GH secreted from B cells may act locally on their own receptors, and their variable expressions may be related to individual immune functions. Widespread distribution of ghrelin and GH secretagogue receptor in human immune cells may indicate unknown biological functions other than enhancing GH secretion in the immune system.


Subject(s)
B-Lymphocytes/metabolism , Human Growth Hormone/metabolism , Neutrophils/metabolism , Peptide Hormones , Peptides/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Receptors, Somatotropin/metabolism , T-Lymphocytes/metabolism , Blotting, Northern , Cells, Cultured , Chromatography, Gel , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Ghrelin , Humans , Immunoglobulin G/metabolism , Insulin-Like Growth Factor I/metabolism , RNA, Messenger/biosynthesis , Receptors, Ghrelin , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Growth Differ ; 12(7): 363-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11457733

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) signaling pathway has inherent oncogenic potential. It is up-regulated in diverse human cancers by either a gain of function in PI3K itself or in its downstream target Akt or by a loss of function in the negative regulator PTEN. However, the complete consequences of this up-regulation are not known. Here we show that insulin and epidermal growth factor or an inactivating mutation in the tumor suppressor PTEN specifically increase the protein levels of hypoxia-inducible factor (HIF) 1alpha but not of HIF-1beta in human cancer cell lines. This specific elevation of HIF-1alpha protein expression requires PI3K signaling. In the prostate carcinoma-derived cell lines PC-3 and DU145, insulin- and epidermal growth factor-induced expression of HIF-1alpha was inhibited by the PI3K-specific inhibitors LY294002 and wortmannin in a dose-dependent manner. HIF-1beta expression was not affected by these inhibitors. Introduction of wild-type PTEN into the PTEN-negative PC-3 cell line specifically inhibited the expression of HIF-1alpha but not that of HIF-1beta. In contrast to the HIF-1alpha protein, the level of HIF-1alpha mRNA was not significantly affected by PI3K signaling. Vascular endothelial growth factor reporter gene activity was induced by insulin in PC-3 cells and was inhibited by the PI3K inhibitor LY294002 and by the coexpression of a HIF-1 dominant negative construct. Vascular endothelial growth factor reporter gene activity was also inhibited by expression of a dominant negative PI3K construct and by the tumor suppressor PTEN.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Transcription Factors , Tumor Suppressor Proteins , Androstadienes/pharmacology , Blotting, Northern , Cell Fractionation , Chromones/pharmacology , Culture Media, Serum-Free , Endothelial Growth Factors/metabolism , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunoblotting , Insulin/pharmacology , Lymphokines/metabolism , Morpholines/pharmacology , PTEN Phosphohydrolase , Phosphoinositide-3 Kinase Inhibitors , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wortmannin
17.
Proc Natl Acad Sci U S A ; 97(4): 1749-53, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677529

ABSTRACT

Phosphatidylinositol 3-kinase (PI 3-kinase) is a signaling molecule that controls numerous cellular properties and activities. The oncogene v-p3k is a homolog of the gene coding for the catalytic subunit of PI 3-kinase, p110alpha. P3k induces transformation of cells in culture, formation of hemangiosarcomas in young chickens, and myogenic differentiation in myoblasts. Here, we describe a role of PI 3-kinase in angiogenesis. Overexpression of the v-P3k protein or of cellular PI 3-kinase equipped with a myristylation signal, Myr-P3k, can induce angiogenesis in the chorioallantoic membrane (CAM) of the chicken embryo. This process is characterized by extensive sprouting of new blood vessels and enlargement of preexisting vessels. Overexpression of the myristylated form of the PI 3-kinase target Akt, Myr-Akt, also induces angiogenesis. Overexpression of the tumor suppressor PTEN or of dominant-negative constructs of PI 3-kinase inhibits angiogenesis in the yolk sac of chicken embryos, suggesting that PI 3-kinase and Akt signaling is required for normal embryonal angiogenesis. The levels of mRNA for vascular endothelial growth factor (VEGF) are elevated in cells expressing activated PI 3-kinase or Myr-Akt. VEGF mRNA levels are also increased by insulin treatment through the PI 3-kinase-dependent pathway. VEGF mRNA levels are decreased in cells treated with the PI 3-kinase inhibitor LY294002 and restored by overexpression of v-P3k or Myr-Akt. Overexpression of VEGF by the RCAS vector induces angiogenesis in chicken embryos. These results suggest that PI 3-kinase plays an important role in angiogenesis and regulates VEGF expression.


Subject(s)
Endothelial Growth Factors/genetics , Endothelium, Vascular/embryology , Lymphokines/genetics , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases , Signal Transduction , Animals , Chick Embryo , Chorion/metabolism , Endothelial Growth Factors/metabolism , Fibroblasts , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Immunohistochemistry , Lymphokines/metabolism , Oncogenes/genetics , Phosphatidylinositol 3-Kinases/metabolism , Plasmids , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Retroviridae/genetics , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Proc Natl Acad Sci U S A ; 96(5): 2077-81, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051597

ABSTRACT

The oncogene p3k, coding for a constitutively active form of phosphatidylinositol 3-kinase (PI 3-kinase), strongly activates myogenic differentiation. Inhibition of endogenous PI 3-kinase activity with the specific inhibitor LY294002, or with dominant-negative mutants of PI 3-kinase, interferes with myotube formation and with the expression of muscle-specific proteins. Here we demonstrate that a downstream target of PI 3-kinase, serine-threonine kinase Akt, plays an important role in myogenic differentiation. Expression of constitutively active forms of Akt dramatically enhances myotube formation and expression of the muscle-specific proteins MyoD, creatine kinase, myosin heavy chain, and desmin. Transdominant negative forms of Akt inhibit myotube formation and the expression of muscle-specific proteins. The inhibition of myotube formation and the reduced expression of muscle-specific proteins caused by the PI 3-kinase inhibitor LY294002 are completely reversed by constitutively active forms of Akt. Wild-type cellular Akt effects a partial reversal of LY294002-induced inhibition of myogenic differentiation. This result suggests that Akt can substitute for PI 3-kinase in the stimulation of myogenesis; Akt may be an essential downstream component of PI 3-kinase-induced muscle differentiation.


Subject(s)
Cell Differentiation , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Chick Embryo , Chromones/pharmacology , Creatine Kinase/genetics , Desmin/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Morpholines/pharmacology , Muscle, Skeletal/physiology , MyoD Protein/genetics , Myosin Heavy Chains/genetics , Phosphatidylinositol 3-Kinases/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , Transfection
19.
J Virol ; 73(4): 3071-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074157

ABSTRACT

Some human adenoviruses are tumorigenic in rodents. Subgroup A and B human adenoviruses generally induce sarcomas in both male and female animals, and the gene products encoded within viral early region 1 (E1 region) are both necessary and sufficient for this tumorigenicity. In contrast, subgroup D human adenovirus type 9 (Ad9) induces estrogen-dependent mammary tumors in female rats and requires the E4 region-encoded ORF1 oncoprotein for its tumorigenicity. Considering the established importance of the viral E1 region for tumorigenesis by adenoviruses, we investigated whether this viral transcription unit is also necessary for Ad9 to generate mammary tumors. The nucleotide sequence of the Ad9 E1 region indicated that the gene organization and predicted E1A and E1B polypeptides of Ad9 are closely related to those of other human adenovirus E1 regions. In addition, an Ad9 E1 region plasmid demonstrated focus-forming activity in both low-passage-number and established rat embryo fibroblasts, whereas a large deletion within either the E1A or E1B gene of this plasmid diminished transforming activity. Surprisingly, we found that introducing the same transformation-inactivating E1A and E1B deletions into Ad9 results in mutant viruses that retain the ability to elicit mammary tumors in rats. These results are novel in showing that Ad9 represents a unique oncogenic adenovirus in which the E4 region, rather than the E1 region, encodes the major oncogenic determinant in the rat.


Subject(s)
Adenovirus E1 Proteins/genetics , Adenoviruses, Human/genetics , Cell Transformation, Neoplastic/genetics , Mammary Neoplasms, Experimental/virology , Amino Acid Sequence , Animals , Base Sequence , Female , Gene Expression Regulation, Viral , Humans , Male , Mammary Neoplasms, Experimental/pathology , Molecular Sequence Data , Rats , Sequence Alignment , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...