Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell Death Dis ; 15(3): 212, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38485719

ABSTRACT

During the maturation of hematopoietic stem/progenitor cells (HSPCs) to fully differentiated mature B lymphocytes, developing lymphocytes may undergo malignant transformation and produce B-cell lymphomas. Emerging evidence shows that through the endothelial-hematopoietic transition, specialized endothelial cells called the hemogenic endothelium can differentiate into HSPCs. However, the contribution of genetic defects in hemogenic endothelial cells to B-cell lymphomagenesis has not yet been investigated. Here, we report that mice with endothelial cell-specific deletion of Fbw7 spontaneously developed diffuse large B-cell lymphoma (DLBCL) following Bcl6 accumulation. Using lineage tracing, we showed that B-cell lymphomas in Fbw7 knockout mice were hemogenic endothelium-derived. Mechanistically, we found that FBW7 directly interacted with Bcl6 and promoted its proteasomal degradation. FBW7 expression levels are inversely correlated with BCL6 expression. Additionally, pharmacological disruption of Bcl6 abolished Fbw7 deletion-induced B-cell lymphomagenesis. We conclude that selective deletion of E3 ubiquitin ligase FBW7 in VE-cadherin positive endothelial cells instigates diffuse large B-cell lymphoma via upregulation of BCL6 stability. In addition, the mice with endothelial cell-specific deletion of Fbw7 provide a valuable preclinical platform for in vivo development and evaluation of novel therapeutic interventions for the treatment of DLBCL.


Subject(s)
Antigens, CD , Cadherins , Lymphoma, Large B-Cell, Diffuse , Ubiquitin-Protein Ligases , Animals , Mice , Endothelial Cells/metabolism , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice, Knockout , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
2.
iScience ; 25(1): 103570, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-34988407

ABSTRACT

Overwhelming evidence indicates that infiltration of tumors by Treg cells with elevated levels of FOXP3 suppresses the host antitumor immune response. However, the molecular mechanisms that maintain high expression of FOXP3 in tumor-infiltrating Treg cells remain elusive. Here, we report that AMP-activated protein kinase alpha1 (AMPKα1) enables high FOXP3 expression in tumor-infiltrating Treg cells. Mice with Treg-specific AMPKα1 deletion showed delayed tumor progression and enhanced antitumor T cell immunity. Further experiments showed that AMPKα1 maintains the functional integrity of Treg cells and prevents interferon-γ production in tumor-infiltrating Treg cells. Mechanistically, AMPKα1 maintains the protein stability of FOXP3 in Treg cells by downregulating the expression of E3 ligase CHIP (STUB1). Our results suggest that AMPKα1 activation promotes tumor growth by maintaining FOXP3 stability in tumor-infiltrating Treg cells and that selective inhibition of AMPK in Treg cells might be an effective anti-tumor therapy.

3.
Int J Mol Sci ; 22(15)2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34361033

ABSTRACT

Apolipoprotein E (ApoE), an essential plasma apolipoprotein, has three isoforms (E2, E3, and E4) in humans. E2 is associated with type III hyperlipoproteinemia. E4 is the major susceptibility gene to Alzheimer's disease (AD) and coronary heart disease (CHD). We investigated lipid metabolism and atherosclerotic lesions of novel humanized ApoE knockin (hApoE KI) rats in comparison to wide-type (WT) and ApoE knockout (ApoE KO) rats. The hApoE2 rats showed the lowest bodyweight and white fat mass. hApoE2 rats developed higher serum total cholesterol (TC), total triglyceride (TG), and low- and very low density lipoprotein (LDL-C&VLDL-C). ApoE KO rats also exhibited elevated TC and LDL-C&VLDL-C. Only mild atherosclerotic lesions were detected in hApoE2 and ApoE KO aortic roots. Half of the hApoE2 rats developed hepatic nodular cirrhosis. A short period of the Paigen diet (PD) treatment led to the premature death of the hApoE2 and ApoE KO rats. Severe vascular wall thickening of the coronary and pulmonary arteries was observed in 4-month PD-treated hApoE4 rats. In conclusion, hApoE2 rats develop spontaneous hyperlipidemia and might be suitable for studies of lipid metabolism-related diseases. With the PD challenge, hApoE4 KI rats could be a novel model for the analysis of vascular remodeling.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Hyperlipidemias/genetics , Lipid Metabolism , Liver Cirrhosis/genetics , Animals , Apolipoproteins E/metabolism , Cholesterol/blood , Coronary Vessels/metabolism , Coronary Vessels/pathology , Gene Knock-In Techniques , Humans , Lipoproteins, LDL/blood , Male , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Triglycerides/blood , Vascular Remodeling
4.
Thorac Cancer ; 11(8): 2209-2223, 2020 08.
Article in English | MEDLINE | ID: mdl-32529777

ABSTRACT

BACKGROUND: The aim of this study was to determine whether progesterone could inhibit the growth of lung adenocarcinoma cells via membrane progesterone receptor alpha (mPRα) and elucidate its potential mechanism. The relationship between mPRα expression and the survival prognosis of lung adenocarcinoma patients was studied. METHODS: A mPRα knockdown lung adenocarcinoma cell line was constructed and treated with P4 and Org (a derivative of P4 and specific agonist of mPRα). Cell proliferation was assessed using CCK-8 and plate colony formation assays. Protein expression was detected by western blotting. A nude mouse model of lung adenocarcinoma was established to assess the antitumor effect of P4/Org in vivo. RESULTS: We initially determined that mPRα could promote the development of lung adenocarcinoma through the following lines of evidence. High expression of mPRα both at the mRNA and protein level was significantly associated with the poor prognosis of lung adenocarcinoma patients. The downregulation of mPRα inhibited the proliferation of lung adenocarcinoma cells. We further showed that mPRα mediates the ability of P4 to inhibit the growth of lung adenocarcinoma cells through the following lines of evidence: P4/Org inhibited the proliferation of lung adenocarcinoma cells; mPRα mediated the ability of P4/Org to inhibit lung adenocarcinoma cell proliferation; mPRα mediated the ability of P4/Org to inhibit the PKA (cAMP-dependent protein kinase)/CREB (cAMP responsive element binding protein) and PKA/ß-catenin signaling pathways; and P4/Org inhibited the growth of a lung adenocarcinoma tumor model in vivo. CONCLUSIONS: In summary, the results of our study show that progesterone can inhibit lung adenocarcinoma cell growth via mPRα.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Progesterone/therapeutic use , Receptors, Progesterone/antagonists & inhibitors , Animals , Female , Humans , Male , Mice , Progesterone/pharmacology , Transfection , Xenograft Model Antitumor Assays
5.
Front Cell Dev Biol ; 8: 18, 2020.
Article in English | MEDLINE | ID: mdl-32154244

ABSTRACT

Obesity is characterized by low-grade chronic inflammation. As an acute-phase reactant to inflammation and infection, C-reactive protein (CRP) has been found to be the strongest factor associated with obesity. Here we show that chronic elevation of human CRP at baseline level causes the obesity. The obesity phenotype is confirmed by whole-body magnetic resonance imaging (MRI), in which the total fat mass is 6- to 9- fold higher in the CRP rats than the control rats. Univariate linear regression analysis showed different growth rates between the CRP rats and the control rats, and that the difference appears around 11 weeks old, indicating that they developed adult-onset obesity. We also found that chronic elevation of CRP can prime molecular changes broadly in the innate immune system, energy expenditure systems, thyroid hormones, apolipoproteins, and gut flora. Our data established a causal role of CRP elevation in the development of adult-onset obesity.

6.
Mol Carcinog ; 59(2): 179-192, 2020 02.
Article in English | MEDLINE | ID: mdl-31777985

ABSTRACT

The discovery of epidermal growth factor receptor (EGFR) mutations has made EGFR tyrosine kinase inhibitors (EGFR-TKIs) a milestone in the treatment for advanced non-small cell lung cancer (NSCLC). However, patients lacking EGFR mutations are not sensitive to EGFR-TKI treatment and the emergence of secondary resistance poses new challenges for the targeted therapy of lung cancer. In this study, we identified that the expression of membrane progesterone receptor α (mPRα) was associated with EGFR mutations in lung adenocarcinoma patients and subsequently affected the efficacy of EGFR-TKIs. Progesterone (P4) or its derivative Org OD02-0 (Org), which is mediated by mPRα, increases the function of EGFR-TKIs to suppress the proliferation, migration, and invasion of lung adenocarcinoma cells in vitro and in vivo. In addition, the mPRα pathway triggers delayed resistance to EGFR-TKIs. Mechanistic investigations demonstrated that the mPRα pathway can crosstalk with the EGFR pathway by activating nongenomic effects to inhibit the EGFR-SRC-ERK1/2 pathway, thereby promoting antitumorigenic effects. In conclusion, our data describe an essential role for mPRα in improving sensitivity to EGFR-TKIs, thus rationalizing its potential as a therapeutic target for lung adenocarcinomas.


Subject(s)
Adenocarcinoma/drug therapy , ErbB Receptors/antagonists & inhibitors , Gefitinib/pharmacology , Lung Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Progesterone/pharmacology , Receptors, Progesterone/metabolism , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mutation , Progesterone/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Receptors, Progesterone/genetics , Survival Analysis , Xenograft Model Antitumor Assays/methods , src-Family Kinases/metabolism
7.
Oncotarget ; 7(36): 57832-57840, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27506936

ABSTRACT

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a known tumor suppressor in non-small cell lung cancer (NSCLC). By performing a systematic review and meta-analysis of the literature, we determined the prognostic value of decreased PTEN expression in patients with NSCLC. We comprehensively and systematically searched through multiple online databases up to May 22, 2016 for NSCLC studies reporting on PTEN expression and patient survival outcome. Several criteria, including the Newcastle-Ottawa Quality Assessment Scale (NOS), were used to discriminate between studies. In total, 23 eligible studies with a total of 2,505 NSCLC patients were included in our meta-analysis. Our results demonstrated that decreased expression of PTEN correlated with poor overall survival in NSCLC patients and was indicative of a poor prognosis for disease-free survival and progression-free survival in patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Disease-Free Survival , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Multivariate Analysis , Prognosis , Treatment Outcome
8.
Sci Rep ; 6: 30437, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27457567

ABSTRACT

The prognostic value of forkhead box protein P1 (FOXP1) protein expression in tumors remains controversial. Therefore, we conducted a systematic review and meta-analysis, searching the PubMed, Embase and Web of Science databases to identify eligible studies. In total, we analyzed 22 articles that examined 9 tumor types and included 2468 patients. Overall, decreased expression of FOXP1 protein was associated with favorable overall survival (OS) in lymphoma patients (HR = 0.38, 95%CI: 0.30-0.48, p < 0.001). In patients with solid tumors, decreased FOXP1 expression correlated with unfavorable OS (HR = 1.82, 95%CI: 1.18-2.83, p = 0.007). However, when FOXP1 protein expression was nuclear, decreased expression was also associated with favorable OS (HR = 0.53, 95%CI: 0.32-0.86, p = 0.011). Furthermore, decreased FOXP1 expression resulted in the best OS in patients with mucosa-associated lymphoid tissue (MALT) lymphomas (HR = 0.26, 95%CI: 0.11-0.59, p = 0.001), but the worst OS was observed in non-small cell lung cancer (NSCLC) patients (HR = 3.11, 95%CI: 1.87-5.17, p < 0.001). In addition, decreased FOXP1 expression was significantly correlated with an unfavorable relapse-free survival (RFS) in breast cancer patients (HR = 1.93, 95%CI: 1.33-2.80, p = 0.001).


Subject(s)
Forkhead Transcription Factors/metabolism , Neoplasms/metabolism , Repressor Proteins/metabolism , Disease-Free Survival , Humans , Neoplasm Recurrence, Local/pathology , Prognosis , Publication Bias
9.
PLoS One ; 11(4): e0152674, 2016.
Article in English | MEDLINE | ID: mdl-27035914

ABSTRACT

BACKGROUND: Liver kinase B1 (LKB1) is a protein kinase that regulates the growth, integrity and polarity of mammalian cells. Recent studies have reported the prognostic value of decreased LKB1 expression in different tumors. However, the results of these studies remain controversial. Therefore, this meta-analysis was performed to more accurately estimate the role of decreased LKB1 in the prognostication of human solid tumors. METHODS: A systematic literature search in the electronic databases PubMed, Embase, Web of Science and CNKI (updated to October 15, 2015) was performed to identify eligible studies. The overall survival (OS), relapse-free survival (RFS), disease-free survival (DFS) and clinicopathological features data were collected from these studies. The hazard ratios (HRs), odds ratios (ORs) and 95% confidence intervals (CIs) were calculated and pooled with a random-effects models using Stata12.0 software. RESULTS: A total of 14 studies covering 1915 patients with solid tumors were included in this meta-analysis. Decreased LKB1 was associated with poorer OS in both the univariate (HR: 1.86, 95%CI: 1.42-2.42, P<0.001) and multivariate (HR: 1.55, 95%CI: 1.09-2.21, P = 0.015) analyses. A subgroup analysis revealed that the associations between decreased LKB1 and poor OS were significant within the Asian region (HR 2.18, 95%CI: 1.66-2.86, P<0.001) and obvious for lung cancer (HR: 2.16, 95%CI: 1.47-3.18, P<0.001). However, the articles that involved analyses of both RFS and DFS numbered only 3, and no statistically significant correlations of decreased LKB1 with RFS or DFS were observed in this study. Additionally, the pooled odds ratios (ORs) indicated that decreased LKB1 was associated with larger tumor size (OR: 1.60, 95%CI: 1.09-2.36, P = 0.017), lymph node metastasis (OR: 2.41, 95%CI: 1.53-3.78, P<0.001) and a higher TNM stage (OR: 3.35, 95%CI: 2.20-5.09, P<0.001). CONCLUSION: These results suggest that decreased LKB1 expression in patients with solid tumors might be related to poor prognosis and serve as a potential predictive marker of poor clinicopathological prognostic factors. Additional studies are required to verify the clinical utility of decreased LKB1 in solid tumors.


Subject(s)
Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Humans , Prognosis
10.
Biomed Res Int ; 2015: 426429, 2015.
Article in English | MEDLINE | ID: mdl-26075237

ABSTRACT

Basal phenotype breast cancer is one of the most aggressive breast cancers that frequently metastasize to brain. The role of sex hormones and their receptors in development of this disease is largely unclear. We demonstrated that mPRα was expressed at a moderate level in a brain metastatic BPBC cell line MB231Br, which was derived from the parent mPRα undetectable MB231 cells. It functioned as an essential mediator for progesterone induced inhibitory effects on cell migration of MB231Br and, when coincubated with PP1, synergistically enhanced the progesterone's inhibitory effect on cell migration and invasion in vitro. Progesterone and PP1 cotreatment induced a cascade of molecular signaling events, such as dephosphorylation of FAK, downregulation of MMP9, VEGF, and KCNMA1 expressions. Our in vitro study demonstrated that mPRα was expressed and functioned as an essential mediator for progesterone induced inhibitory effects on cell migration and invasion in BPBC cells. This inhibitory effect was enhanced by PP1 via FAK dephosphorylation, MMP9, VEGF, and KCNMA1 downregulation mechanisms. Our study provides a new clue toward the development of novel promising agents and pathways for inhibiting nuclear hormonal receptor-negative and endocrine-resistant breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement/drug effects , Neoplasm Proteins/antagonists & inhibitors , Progesterone/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Signal Transduction/genetics
11.
J Biomed Mater Res A ; 103(3): 887-95, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24829052

ABSTRACT

Viruses are no longer recognized purely for being ubiquitous pathogens, but have served as building blocks for material chemistry and nanotechnology. Thousands of coat protein subunits of a viral particle can be modified chemically and/or genetically. We have previously shown that the three-dimensional porous hydrogels can easily be functionalized by Tobacco mosaic virus (TMV), a rod-like plant virus, using its mutant, RGD-TMV. RGD-TMV hosted bioadhesive peptide (RGD) in the hydrogel, which was shown to enhance cell attachment and promote osteogenic differentiation of cultured stem cell. To translate this technology to potential clinical applications, we sought to study the biocompatibility of the hydrogel. In this paper, the hydrogels were implanted in vivo and assessed for their immunogenicity, toxicity, and biodegradability. Immune response for TMV substantially decreased when incorporated in the hydrogel implants. The implanted TMV hydrogels exhibited no apparent toxicity and were degradable in mice. The results highlighted the feasibility of using TMV incorporated hydrogels as scaffolding materials for regenerative medicine in terms of biocompatibility and biodegradability.


Subject(s)
Biocompatible Materials/chemistry , Capsid Proteins/chemistry , Hydrogels/chemistry , Tissue Engineering/methods , Tissue Scaffolds , Alginates/chemistry , Animals , Bone and Bones/metabolism , Cell Adhesion , Cell Differentiation , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred BALB C , Mutation , Nanostructures/chemistry , Oligopeptides/chemistry , Osteogenesis , Paraffin/chemistry , Peptides/chemistry , Regenerative Medicine , Tobacco Mosaic Virus/chemistry
12.
PLoS One ; 9(9): e107124, 2014.
Article in English | MEDLINE | ID: mdl-25222296

ABSTRACT

PURPOSE: Aberrant promoter DNA methylation can serve as a predictive biomarker for improved clinical responses to certain chemotherapeutics. One of the major advantages of methylation biomarkers is the ease of detection and clinical application. In order to identify methylation biomarkers predictive of a response to a taxane-platinum based chemotherapy regimen in advanced NSCLC we performed an unbiased methylation analysis of 1,536 CpG dinucleotides in cancer-associated gene loci and correlated results with clinical outcomes. METHODS: We studied a cohort of 49 patients (median age 62 years) with advanced NSCLC treated at the Atlanta VAMC between 1999 and 2010. Methylation analysis was done on the Illumina GoldenGate Cancer panel 1 methylation microarray platform. Methylation data were correlated with clinical response and adjusted for false discovery rates. RESULTS: Cav1 methylation emerged as a powerful predictor for achieving disease stabilization following platinum taxane based chemotherapy (p = 1.21E-05, FDR significance  = 0.018176). In Cox regression analysis after multivariate adjustment for age, performance status, gender, histology and the use of bevacizumab, CAV1 methylation was significantly associated with improved overall survival (HR 0.18 (95%CI: 0.03-0.94)). Silencing of CAV1 expression in lung cancer cell lines(A549, EKVX)by shRNA led to alterations in taxane retention. CONCLUSIONS: CAV1 methylation is a predictor of disease stabilization and improved overall survival following chemotherapy with a taxane-platinum combination regimen in advanced NSCLC. CAV1 methylation may predict improved outcomes for other chemotherapeutic agents which are subject to cellular clearance mediated by caveolae.


Subject(s)
Bridged-Ring Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Caveolin 1/genetics , DNA Methylation , Platinum Compounds/therapeutic use , Promoter Regions, Genetic , Taxoids/therapeutic use , Cell Line, Tumor , CpG Islands , Drug Combinations , Epithelial-Mesenchymal Transition/genetics , Humans , Multivariate Analysis , Regression Analysis , Retrospective Studies , Survival Analysis , Treatment Outcome
13.
Oncol Rep ; 29(5): 1873-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23467643

ABSTRACT

Lung cancer is the leading cause of cancer morbidity and mortality in the world. The incidence of lung cancer, particularly lung adenocarcinoma, is increasing in women compared to men. The role of sex hormones in the development of lung cancer has attracted substantial interest, but remains largely unknown. In this study, we demonstrated that membrane progesterone receptor α (mPRα) was expressed in a lung adenocarcinoma cell line, A549, and was located on the cell membrane. In additional experiments, we found that mPRα functioned as an essential mediator for progesterone (P4)-induced inhibitory effects on cell migration and invasion of A549 cells. Furthermore, PP1 (an Src pathway inhibitor), when co-incubated with P4, synchronously enhanced the inhibitory effects of P4 on cell migration and invasion. To explore the mechanisms of inhibition, we found that P4 and PP1 induced a cascade of molecular signaling events, such as dephosphorylation of focal adhesion kinase (FAK) and downregulation of matrix metalloproteinase 9 (MMP-9). Our study provides a mechanistic view on the effects of P4 through mPRα→Src/FAK relevant pathways in human lung adenocarcinoma cells and may aid in the development of novel therapeutic tools for the treatment of lung cancer.


Subject(s)
Cell Movement/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Progesterone/metabolism , Receptors, Progesterone/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , Down-Regulation , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Lung Neoplasms/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Receptors, Progesterone/genetics
14.
PLoS One ; 7(4): e35198, 2012.
Article in English | MEDLINE | ID: mdl-22496908

ABSTRACT

Classically, the actions of progesterone (P4) are attributed to the binding of nuclear progesterone receptor (PR) and subsequent activation of its downstream target genes. These mechanisms, however, are not applicable to PR- or basal phenotype breast cancer (BPBC) due to lack of PR in these cancers. Recently, the function of membrane progesterone receptor alpha (mPRα) in human BPBC cell lines was studied in our lab. We proposed that the signaling cascades of P4→mPRα pathway may play an essential role in controlling cell proliferation and epithelial mesenchymal transition (EMT) of breast cancer. Using human breast cancer tissue microarrays, we found in this study that the average intensity of mPRα expression, but not percentage of breast cancer with high level of mPRα expression (mPRα-HiEx), was significantly lower in the TNM stage 4 patients compared to those with TNM 1-3 patients; and both average intensities of mPRα expression and mPRα-HiEx rates were significantly higher in cancers negative for ER, as compared with those cancers with ER+. However, after adjusting for age at diagnosis and/or TNM stage, only average intensities of mPRα expression were associated with ER status. In addition, we found that the rates of mPRα-HiEx were significantly higher in cancers with epithelial growth factor receptor-1 (EGFR+) and high level of Ki67 expression, indicating positive correlation between mPRα over expression and EGFR or Ki67. Further analysis indicated that both mPRα-HiEx rate and average intensity of mPRα expression were significantly higher in HER2+ subtype cancers (i.e. HER2+ER-PR-) as compared to ER+ subtype cancers. These data support our hypothesis that P4 modulates the activities of the PI3K and cell proliferation pathways through the caveolar membrane bound growth factor receptors such as mPRα and growth factor receptors. Future large longitudinal studies with larger sample size and survival outcomes are necessary to confirm our findings.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Carcinoma/mortality , Receptors, Progesterone/metabolism , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/pathology , ErbB Receptors/analysis , Female , Humans , Ki-67 Antigen/analysis , Middle Aged , Neoplasm Staging , Progesterone/metabolism , Prognosis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Retrospective Studies
15.
J Bone Miner Res ; 26(10): 2537-47, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21773989

ABSTRACT

The heterogeneous nuclear ribonucleoprotein (hnRNP)-like estrogen response element-binding protein (ERE-BP) competes with estrogen receptor α (ERα) for occupancy of estrogen response elements (EREs). Here we report that ERE-BP potently stimulates osteoclastogenesis. ERE-BP mRNA and protein were found to be expressed ubiquitously in bone. Overexpression of ERE-BP in cultured osteoblasts stimulated expression of the receptor activator of NF-κB ligand (RANKL) and decreased osteoprotegerin (OPG). The effect of ERE-BP on RANKL was shown to be transcriptional in transient transfection assay and competed with via the ER. Constitutive expression of ERE-BP increased the sensitivity of cells toward 1,25-dihydroxyvitamin D(3) stimulation of RANKL expression. In contrast, knockdown of ERE-BP in stromal ST-2 cells decreased basal RANKL promoter activity. Cocultures of ERE-BP lentivirus-transduced ST-2 cells with spleen monocytes induced formation of multinucleated osteoclasts (OCs) characterized by tartrate-resistant acid phosphatase, calcitonin receptors, and functional calcium resorption from bone slices. Although ERα competed with ERE-BP for an ERE in a dose-dependent manner, ERE-BP was an independent and potent regulator of RANKL and osteoclastogenesis. In preosteoclastic RAW cells, overexpression of ERE-BP increased RANK, upregulated NF-κB signaling, and enhanced differentiation toward a mature OC phenotype independent of RANKL. These results identify ERE-BP as a potent modulator of osteoclastogenesis. We hypothesize that ERE-BP may play a critical role in the regulation of bone homeostasis as a modulator of estrogen sensitivity as well as by direct action on the transcription of critical osteoclastogenic genes.


Subject(s)
Bone and Bones/metabolism , Estrogens/physiology , Osteoclasts/cytology , Animals , Base Sequence , Cell Differentiation , Coculture Techniques , DNA Primers , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , RANK Ligand/genetics , RANK Ligand/metabolism , Real-Time Polymerase Chain Reaction
16.
J Bone Miner Res ; 26(1): 209-19, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20683885

ABSTRACT

Tumor necrosis factor α (TNF-α) promotes bone loss and inhibits bone formation. Osterix (Osx, SP7) is a transcription factor required for osteoblast (OB) differentiation because deletion results in a cartilaginous skeleton. We previously described a TNF suppressor element in the Osx promoter that was used to isolate nuclear proteins mediating TNF inhibition of OB differentiation. Nuclear extracts from TNF-treated pre-OBs were incubated with the TNF suppressor element for protein pull-down, and tryptic fragments were analyzed by mass spectrometry. Chromatin immunoprecipitation (ChIP) assay confirmed eight bound transcription factors. One protein, the paired related homeobox protein (Prx1), had been shown previously to have a critical role in limb bud formation and skeletal patterning. PCR revealed Prx1 expression in primary stromal cells (MSCs), C3H10T1/2 cells, and MC3T3 preosteoblasts. TNF stimulated a 14-fold increase in mRNA for Prx1, rapid cell accumulation in MC3T3 cells, and expression in periosteal and trabecular lining cells in vivo. Transient expression of Prx inhibited transcription of Osx and RUNX2. Expression of the Prx1b isoform or Prx2 decreased Osx and RUNX2 mRNA and OB differentiation in preosteoblasts. Silencing of Prx1 with siRNA abrogated TNF suppression of Osx mRNA and increased basal Osx expression. Electrophoretic mobility shift revealed Prx1b as the preferred isoform binding the Osx promoter. These results identify the homeobox protein Prx1 as an obligate mediator of TNF inhibition of Osx and differentiation of OB progenitors. Activation of Prx1 by TNF may contribute to reduced bone formation in inflammatory arthritis, menopause, and aging.


Subject(s)
Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Homeodomain Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mice , Osteoblasts/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sp7 Transcription Factor , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects
17.
Breast Cancer Res ; 12(3): R34, 2010.
Article in English | MEDLINE | ID: mdl-20540763

ABSTRACT

INTRODUCTION: Basal phenotype breast cancers (BPBC) are often associated with apparent epithelial to mesenchymal transition (EMT). The role of progesterone (P4) in regulating EMT of BPBC has not been reported. METHODS: The EMT relevant biology was investigated in vitro using human BPBC cell models (MDA-MB468 and MDA-MB231) with P4, PR agonist (RU486), and PR antagonist (R5020) treatments. The essential role of membrane progesterone receptor alpha (mPRalpha) in the P4-regulated EMT was demonstrated by knocking down the endogenous gene and/or stably transfecting exogenous mPRalpha gene in the BPBC cell models. RESULTS: The expression of snail and down-stream EMT proteins such as occludin, fibronectin, and E-cadherin was significantly regulated by P4 incubation, which was accompanied by cell morphological reversion from mesenchymal to epithelial phenotypes. In searching for the cell mediator of P4' action in the MDA-MB468 (MB468) cells, it was found that mPRalpha but not the nuclear PR has an essential role in the P4 mediated EMT inhibition. Knocking down the expression of mPRalpha with specific siRNA blocked the P4's effects on expression of the EMT proteins. In another BPBC cell line--MDA-MB231 (MB231), which is mPRalpha negative by Western blotting--P4 treatment did not alter cell proliferation and EMT protein expressions. Introduction of the exogenous mPRalpha cDNA into these cells caused cell proliferation, but not EMT, to become responsive to P4 treatment. In further studies, it was found that activation of the PI3K/Akt pathway is necessary for the P4-induced EMT reversion. To define the potential inter-mediate steps between mPRalpha and PI3K, we demonstrated that mPRalpha, caveolin-1 (Cav-1), and epidermal growth factor receptor (EGFR) are colocalized in the membrane of caveolar vesicle and the P4-repressed EMT in MB468 cells can be blocked by EGFR inhibitor (AG1478) and PI3K inhibitor (wortmannin). CONCLUSIONS: Our data suggest that the signaling cascade of P4 induced mesenchymal repression is mediated through mPRalpha and other caveolae bound signaling molecules namely Cav-1, EGFR, and PI3K. This novel finding may have great impact on fully understanding the pathogenesis of BPBC and provide an essential clue for developing a targeted therapeutic strategy for treatment of BPBC.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Membrane/metabolism , Mesoderm/metabolism , Neoplasms, Basal Cell/metabolism , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cadherins/metabolism , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/pathology , Caveolae/metabolism , Caveolin 1/metabolism , Cell Dedifferentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Mesoderm/pathology , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/pathology , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Progestins/pharmacology , RNA, Small Interfering/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Tissue Array Analysis
18.
Bioconjug Chem ; 21(7): 1369-77, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20499838

ABSTRACT

The M13 bacteriophage has been demonstrated to be a robust scaffold for bionanomaterial development. In this paper, we report on the chemical modifications of three kinds of reactive groups, i.e., the amino groups of lysine residues or N-terminal, the carboxylic acid groups of aspartic acid or glutamic acid residues, and the phenol group of tyrosine residues, on M13 surface. The reactivity of each group was identified through conjugation with small fluorescent molecules. Furthermore, the regioselectivity of each reaction was investigated by HPLC-MS-MS. By optimizing the reaction condition, hundreds of fluorescent moieties could be attached to create a highly fluorescent M13 bacteriophage. In addition, cancer cell targeting motifs such as folic acid could also be conjugated onto the M13 surface. Therefore, dual-modified M13 particles with folic acid and fluorescent molecules were synthesized via the selective modification of two kinds of reactive groups. Such dual-modified M13 particles showed very good binding affinity to human KB cancer cells, which demonstrated the potential applications of M13 bacteriophage in bioimaging and drug delivery.


Subject(s)
Bacteriophage M13/chemistry , Fluorescent Dyes/chemistry , Molecular Imaging/methods , Neoplasms/pathology , Fluorescent Dyes/chemical synthesis , Folic Acid/chemistry , HeLa Cells , Humans , Molecular Structure , Particle Size , Stereoisomerism , Surface Properties , Tumor Cells, Cultured
19.
Int J Nanomedicine ; 5: 221-9, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20463938

ABSTRACT

Sex hormonal milieus during the female fertility cycle modulate the tumor vascular permeability of breast cancer. It has been proposed that the liposomal formulated doxorubicin (ie, Doxil), given at the menstrual/estrous stage with the predicted highest tumor vascular permeability, allows significantly increased drug retention in the breast tumor. In the current study, syngeneic murine 4T1 mammary tumors were established on the backs of female BALB/c mice and Doxil was administered at particular mouse estrous cycle stages. The results indicated that Doxil administration during certain times in the mouse estrous cycle was crucial for drug retention in 4T1 tumor tissues. Significantly higher drug concentrations were detected in the tumor tissues when Doxil was administered during the diestrus stage, as compared to when the drug injection was given at all other estrous stages. Our study also showed that the tumor-bearing mice exhibited nearly normal rhythmicity of the estrous cycle post drug injection, indicating the feasibility of continual injection of Doxil at the same estrous cycle stage. By using 4T1 cells cultured in vitro, we showed that progesterone (P4) significantly inhibited cell proliferation and the production of six tumor-derived cytokines, eg, sTNF-RI, CXCL-16, GM-CSF, MIP-1alpha, MIP-1gamma, and Flt3-L. Some of these factors have been shown to be vascular modulators in diverse tissues. In this report, we demonstrated that the concentration of P4 in the plasma and/or estrous cycle stage of 4T1 tumor-bearing mice can be used to select the best time for administrating the liposomal anticancer drugs.


Subject(s)
Doxorubicin/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Dose-Response Relationship, Drug , Female , Injections, Intralesional/methods , Mice , Mice, Inbred BALB C , Treatment Outcome
20.
J Mol Histol ; 41(2-3): 101-10, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20473635

ABSTRACT

The broad tissue distribution of membrane progesterone receptor alpha (mPRalpha) in vertebrates suggests multiple physiological functions of the receptor. Current knowledge regarding the receptor distribution, however, is largely obtained via non-histological assays. In this study, the tissue distribution of mPRalpha in mice of both sexes was described using both histological and non-histological methods. Immunohistochemical analysis revealed that abundant expression of mPRalpha was consistently detected in the cytoplasm and membrane of smooth muscles in vasculatures, gastro-intestines, and uterus. It was also observed in myoepithelial cells of mammary gland and intra-ovarian myofibroblasts. These findings suggest that mPRalpha may function as a mediator of P4 in regulating function of smooth muscles or smooth muscle-like cells in numerous physiological processes such as vasodilation, transportation of contents within luminary organs, relaxation of the uterine myometrium during pregnancy, release of oocytes, and milk secretion. In addition, strong mPRalpha expression was identified in the parietal cells of gastric glands, indicating the potential roles of P4/mPRalpha signaling in the modulation of gastric acid secretion. Surprisingly, in the testis of male mice mPRalpha was mainly seen in the nuclei, rather than cytoplasm and/or membrane, of the primary and secondary spermatocytes, suggesting a direct role of the receptor in gene regulation. Our results indicate that mPRalpha may function as a key modulator of P4 in the modulation of multiple physiological functions in normal mice.


Subject(s)
Cell Membrane/metabolism , Receptors, Progesterone/biosynthesis , Animals , Blotting, Western , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Organ Specificity , Receptors, Progesterone/physiology , Reproduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...