31 results on '"Kirma NB"'
Search Results
2. Therapeutic targeting of the c-fms oncogene diminishes the growth of breast cancer cells and ductal hyperplasia in the mammary gland of the CSF-1 transgenic mouse model.
- Author
-
Kirma, NB, primary, Perla, RP, additional, Nair, HB, additional, Liu, Y, additional, and Tekmal, RR, additional
- Published
- 2009
- Full Text
- View/download PDF
3. Preclinical modeling of endocrine response: a combination therapy approach with the ERβ agonist, diarryl propionitrile and letrozole restores sensitivity to letrozole-resistant breast cancer cells.
- Author
-
Nair, HB, primary, Jayarajan, R, additional, Saha, SR, additional, Veerapaneni, P, additional, Santhamma, B, additional, Kirma, NB, additional, Perla, RP, additional, Joseph, AK, additional, Evans, DB, additional, Brodie, AH, additional, and Tekmal, RR, additional
- Published
- 2009
- Full Text
- View/download PDF
4. Nano-gossypin, a novel cdk2/VEGF inhibitor formulation against breast cancer.
- Author
-
Nair, HB, primary, Veerapaneni, P, additional, Sophie, DM, additional, Kirma, NB, additional, Perla, RP, additional, and Tekmal, RR, additional
- Published
- 2009
- Full Text
- View/download PDF
5. Increased expression of macrophage colony-stimulating factor and its receptor in patients with endometriosis.
- Author
-
Budrys NM, Nair HB, Liu YG, Kirma NB, Binkley PA, Kumar S, Schenken RS, Tekmal RR, Budrys, Nicole M, Nair, Hareesh B, Liu, Ya-Guang, Kirma, Nameer B, Binkley, Peter A, Kumar, Shantha, Schenken, Robert S, and Tekmal, Rajeshwar Rao
- Abstract
Objective: To investigate the expression and regulation of colony-stimulating factor 1 (CSF-1) and its receptor, C-FMS, in endometriosis.Design: In vivo and vitro study.Setting: University-based academic medical center.Patient(s): Reproductive-age women undergoing surgery for benign conditions.Intervention(s): Peritoneal and endometrial tissue samples were obtained.Main Outcome Measure(s): CSF-1 and C-FMS expression.Result(s): Significantly higher CSF-1 levels were found in peritoneal fluid of patients with endometriosis compared with control subjects. Ectopic endometriotic tissue had 3.5-fold and 1.7-fold increases in CSF-1 and C-FMS expression, respectively, compared with eutopic tissue. Coculture of endometrial cells from either established cell lines or patient samples with peritoneal mesothelial cells (PMCs) led to increased expression of CSF-1 and C-FMS. A higher but nonsignificant increase in levels of C-FMS and CSF-1 was found in cocultures of endometrial epithelial cells from patients with endometriosis compared with those without endometriosis.Conclusion(s): Increased CSF-1 levels may contribute to endometriosis lesion formation and progression. Elevation in CSF-1 after coculture of endometrial cells with PMCs suggests that endometrial tissue may be a source of peritoneal CSF-1. Increased C-FMS expression in endometrial cells from women with endometriosis cocultured with PMCs suggests that endometrial tissue involved in lesion formation is highly responsive to CSF-1 signaling. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
6. PAI-1 uncouples integrin-β1 from restrain by membrane-bound β-catenin to promote collagen fibril remodeling in obesity-related neoplasms.
- Author
-
Lin LL, Nayak B, Osmulski PA, Wang E, Wang CP, Valente PT, Wang CM, Tan X, Santanam N, Wang TL, Gaczynska ME, Kost ER, Huang TH, and Kirma NB
- Subjects
- Humans, Female, Endometrial Neoplasms metabolism, Endometrial Neoplasms pathology, Tissue Inhibitor of Metalloproteinase-2 metabolism, Animals, Osteonectin metabolism, Osteonectin genetics, Collagen metabolism, Endometrium metabolism, Endometrium pathology, Collagen Type I metabolism, Cell Membrane metabolism, Epithelial Cells metabolism, Epithelial Cells pathology, Intercellular Signaling Peptides and Proteins, Obesity metabolism, Obesity pathology, Plasminogen Activator Inhibitor 1 metabolism, beta Catenin metabolism, Integrin beta1 metabolism, Extracellular Matrix metabolism
- Abstract
The paracrine actions of adipokine plasminogen activator inhibitor-1 (PAI-1) are implicated in obesity-associated tumorigenesis. Here, we show that PAI-1 mediates extracellular matrix (ECM) signaling via epigenetic repression of DKK1 in endometrial epithelial cells (EECs). While the loss of DKK1 is known to increase β-catenin accumulation for WNT signaling activation, this epigenetic repression causes β-catenin release from transmembrane integrins. Furthermore, PAI-1 elicits the disengagement of TIMP2 and SPARC from integrin-β1 on the cell surface, lifting an integrin-β1-ECM signaling constraint. The heightened interaction of integrin-β1 with type 1 collagen (COL1) remodels extracellular fibrillar structures in the ECM. Consequently, the enhanced nanomechanical stiffness of this microenvironment is conducive to EEC motility and neoplastic transformation. The formation of extensively branched COL1 fibrils is also observed in endometrial tumors of patients with obesity. The findings highlight PAI-1 as a contributor to enhanced integrin-COL1 engagement and extensive ECM remodeling during obesity-associated neoplastic development., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Phagocytosis-initiated tumor hybrid cells acquire a c-Myc-mediated quasi-polarization state for immunoevasion and distant dissemination.
- Author
-
Chou CW, Hung CN, Chiu CH, Tan X, Chen M, Chen CC, Saeed M, Hsu CW, Liss MA, Wang CM, Lai Z, Alvarez N, Osmulski PA, Gaczynska ME, Lin LL, Ortega V, Kirma NB, Xu K, Liu Z, Kumar AP, Taverna JA, Velagaleti GVN, Chen CL, Zhang Z, and Huang TH
- Subjects
- Humans, Male, Carrier Proteins, Prostatic Neoplasms genetics, Prostatic Neoplasms immunology, Prostatic Neoplasms pathology, Signal Transduction, Tumor Cells, Cultured, CD47 Antigen metabolism, Macrophages metabolism, Phagocytosis, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc immunology, Tumor Escape genetics, Tumor Escape immunology, Neoplasm Metastasis genetics, Neoplasm Metastasis immunology
- Abstract
While macrophage phagocytosis is an immune defense mechanism against invading cellular organisms, cancer cells expressing the CD47 ligand send forward signals to repel this engulfment. Here we report that the reverse signaling using CD47 as a receptor additionally enhances a pro-survival function of prostate cancer cells under phagocytic attack. Although low CD47-expressing cancer cells still allow phagocytosis, the reverse signaling delays the process, leading to incomplete digestion of the entrapped cells and subsequent tumor hybrid cell (THC) formation. Viable THCs acquire c-Myc from parental cancer cells to upregulate both M1- and M2-like macrophage polarization genes. Consequently, THCs imitating dual macrophage features can confound immunosurveillance, gaining survival advantage in the host. Furthermore, these cells intrinsically express low levels of androgen receptor and its targets, resembling an adenocarcinoma-immune subtype of metastatic castration-resistant prostate cancer. Therefore, phagocytosis-generated THCs may represent a potential target for treating the disease., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
8. AXL-initiated paracrine activation of pSTAT3 enhances mesenchymal and vasculogenic supportive features of tumor-associated macrophages.
- Author
-
Hung CN, Chen M, DeArmond DT, Chiu CH, Limboy CA, Tan X, Kusi M, Chou CW, Lin LL, Zhang Z, Wang CM, Chen CL, Mitsuya K, Osmulski PA, Gaczynska ME, Kirma NB, Vadlamudi RK, Gibbons DL, Warner S, Brenner AJ, Mahadevan D, Michalek JE, Huang TH, and Taverna JA
- Subjects
- Humans, Animals, Mice, Endothelial Cells, Signal Transduction, Cell Differentiation, Tumor Microenvironment, Cell Line, Tumor, Tumor-Associated Macrophages, Lung Neoplasms
- Abstract
Tumor-associated macrophages (TAMs) are integral to the development of complex tumor microenvironments (TMEs) and can execute disparate cellular programs in response to extracellular cues. However, upstream signaling processes underpinning this phenotypic plasticity remain to be elucidated. Here, we report that concordant AXL-STAT3 signaling in TAMs is triggered by lung cancer cells or cancer-associated fibroblasts in the cytokine milieu. This paracrine action drives TAM differentiation toward a tumor-promoting "M2-like" phenotype with upregulation of CD163 and putative mesenchymal markers, contributing to TAM heterogeneity and diverse cellular functions. One of the upregulated markers, CD44, mediated by AXL-IL-11-pSTAT3 signaling cascade, enhances macrophage ability to interact with endothelial cells and facilitate formation of primitive vascular networks. We also found that AXL-STAT3 inhibition can impede the recruitment of TAMs in a xenograft mouse model, thereby suppressing tumor growth. These findings suggest the potential application of AXL-STAT3-related markers to quantitatively assess metastatic potential and inform therapeutic strategies in lung cancer., Competing Interests: Declaration of interests S.W. is principal investigator at Sumitomo Dainippon Pharma Oncology., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. 2-Hydroxyglutarate destabilizes chromatin regulatory landscape and lineage fidelity to promote cellular heterogeneity.
- Author
-
Kusi M, Zand M, Lin LL, Chen M, Lopez A, Lin CL, Wang CM, Lucio ND, Kirma NB, Ruan J, Huang TH, and Mitsuya K
- Subjects
- Alcohol Oxidoreductases metabolism, Ascorbic Acid analogs & derivatives, Ascorbic Acid metabolism, Cell Differentiation, Cell Line, Tumor, DNA Repair physiology, Epigenome genetics, Forkhead Transcription Factors genetics, Gene Expression genetics, Gene Expression Regulation genetics, Humans, Isocitrate Dehydrogenase genetics, Neoplasms genetics, Neoplasms metabolism, Nucleosomes metabolism, Repressor Proteins genetics, Chromatin metabolism, Glutarates metabolism
- Abstract
The epigenome delineates lineage-specific transcriptional programs and restricts cell plasticity to prevent non-physiological cell fate transitions. Although cell diversification fosters tumor evolution and therapy resistance, upstream mechanisms that regulate the stability and plasticity of the cancer epigenome remain elusive. Here we show that 2-hydroxyglutarate (2HG) not only suppresses DNA repair but also mediates the high-plasticity chromatin landscape. A combination of single-cell epigenomics and multi-omics approaches demonstrates that 2HG disarranges otherwise well-preserved stable nucleosome positioning and promotes cell-to-cell variability. 2HG induces loss of motif accessibility to the luminal-defining transcriptional factors FOXA1, FOXP1, and GATA3 and a shift from luminal to basal-like gene expression. Breast tumors with high 2HG exhibit enhanced heterogeneity with undifferentiated epigenomic signatures linked to adverse prognosis. Further, ascorbate-2-phosphate (A2P) eradicates heterogeneity and impairs growth of high 2HG-producing breast cancer cells. These findings suggest 2HG as a key determinant of cancer plasticity and provide a rational strategy to counteract tumor cell evolution., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
10. Contacts with Macrophages Promote an Aggressive Nanomechanical Phenotype of Circulating Tumor Cells in Prostate Cancer.
- Author
-
Osmulski PA, Cunsolo A, Chen M, Qian Y, Lin CL, Hung CN, Mahalingam D, Kirma NB, Chen CL, Taverna JA, Liss MA, Thompson IM, Huang TH, and Gaczynska ME
- Subjects
- Cell Line, Tumor, Humans, Male, Phenotype, Macrophages metabolism, Neoplastic Cells, Circulating metabolism, Prostatic Neoplasms immunology
- Abstract
Aggressive tumors of epithelial origin shed cells that intravasate and become circulating tumor cells (CTC). The CTCs that are able to survive the stresses encountered in the bloodstream can then seed metastases. We demonstrated previously that CTCs isolated from the blood of prostate cancer patients display specific nanomechanical phenotypes characteristic of cell endurance and invasiveness and patient sensitivity to androgen deprivation therapy. Here we report that patient-isolated CTCs are nanomechanically distinct from cells randomly shed from the tumor, with high adhesion as the most distinguishing biophysical marker. CTCs uniquely coisolated with macrophage-like cells bearing the markers of tumor-associated macrophages (TAM). The presence of these immune cells was indicative of a survival-promoting phenotype of "mechanical fitness" in CTCs based on high softness and high adhesion as determined by atomic force microscopy. Correlations between enumeration of macrophages and mechanical fitness of CTCs were strong in patients before the start of hormonal therapy. Single-cell proteomic analysis and nanomechanical phenotyping of tumor cell-macrophage cocultures revealed that macrophages promoted epithelial-mesenchymal plasticity in prostate cancer cells, manifesting in their mechanical fitness. The resulting softness and adhesiveness of the mechanically fit CTCs confer resistance to shear stress and enable protective cell clustering. These findings suggest that selected tumor cells are coached by TAMs and accompanied by them to acquire intermediate epithelial/mesenchymal status, thereby facilitating survival during the critical early stage leading to metastasis. SIGNIFICANCE: The interaction between macrophages and circulating tumor cells increases the capacity of tumor cells to initiate metastasis and may constitute a new set of blood-based targets for pharmacologic intervention., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
11. PAI-1-Dependent Inactivation of SMAD4-Modulated Junction and Adhesion Complex in Obese Endometrial Cancer.
- Author
-
Lin LL, Kost ER, Lin CL, Valente P, Wang CM, Kolonin MG, Daquinag AC, Tan X, Lucio N, Hung CN, Wang CP, Kirma NB, and Huang TH
- Subjects
- Adipose Tissue pathology, Down-Regulation genetics, Endometrial Neoplasms complications, Endometrial Neoplasms genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Low Density Lipoprotein Receptor-Related Protein-1 metabolism, Obesity complications, Protein Binding, Proteolysis, Proteomics, Smad4 Protein genetics, Stromal Cells metabolism, Transcription, Genetic, Transforming Growth Factor beta metabolism, Tumor Microenvironment, Ubiquitin metabolism, Endometrial Neoplasms metabolism, Junctional Adhesion Molecules metabolism, Obesity metabolism, Plasminogen Activator Inhibitor 1 metabolism, Smad4 Protein metabolism
- Abstract
While plasminogen activator inhibitor-1 (PAI-1) is known to potentiate cellular migration via proteolytic regulation, this adipokine is implicated as an oncogenic ligand in the tumor microenvironment. To understand the underlying paracrine mechanism, here, we conduct transcriptomic analysis of 1,898 endometrial epithelial cells (EECs) exposed and unexposed to PAI-1-secreting adipose stromal cells. The PAI-1-dependent action deregulates crosstalk among tumor-promoting and tumor-repressing pathways, including transforming growth factor β (TGF-β). When PAI-1 is tethered to lipoprotein receptor-related protein 1 (LRP1), the internalized signaling causes downregulation of SMAD4 at the transcriptional and post-translational levels that attenuates TGF-β-related transcription programs. Repression of genes encoding the junction and adhesion complex preferentially occurs in SMAD4-underexpressed EECs of persons with obesity. The findings highlight a role of PAI-1 signaling that renders ineffective intercellular communication for the development of adiposity-associated endometrial cancer., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
12. Single-Cell Proteomic Profiling Identifies Combined AXL and JAK1 Inhibition as a Novel Therapeutic Strategy for Lung Cancer.
- Author
-
Taverna JA, Hung CN, DeArmond DT, Chen M, Lin CL, Osmulski PA, Gaczynska ME, Wang CM, Lucio ND, Chou CW, Chen CL, Nazarullah A, Lampkin SR, Qiu L, Bearss DJ, Warner S, Whatcott CJ, Mouritsen L, Wade M, Weitman S, Mesa RA, Kirma NB, Chao WT, and Huang TH
- Subjects
- Aged, Aged, 80 and over, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Line, Tumor, Drug Resistance, Neoplasm, Drug Synergism, Epithelial-Mesenchymal Transition drug effects, Feasibility Studies, Female, Flow Cytometry methods, Humans, Janus Kinase 1 metabolism, Lung pathology, Lung Neoplasms pathology, Male, Mice, Middle Aged, Nitriles, Protein Kinase Inhibitors therapeutic use, Proteomics methods, Proto-Oncogene Proteins metabolism, Pyrazoles pharmacology, Pyrazoles therapeutic use, Pyrimidines pharmacology, Pyrimidines therapeutic use, RNA-Seq, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction drug effects, Single-Cell Analysis methods, Sulfonamides pharmacology, Sulfonamides therapeutic use, Tissue Array Analysis, Xenograft Model Antitumor Assays, Axl Receptor Tyrosine Kinase, Antineoplastic Combined Chemotherapy Protocols pharmacology, Janus Kinase 1 antagonists & inhibitors, Lung Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins antagonists & inhibitors, Receptor Protein-Tyrosine Kinases antagonists & inhibitors
- Abstract
Cytometry by time-of-flight (CyTOF) simultaneously measures multiple cellular proteins at the single-cell level and is used to assess intertumor and intratumor heterogeneity. This approach may be used to investigate the variability of individual tumor responses to treatments. Herein, we stratified lung tumor subpopulations based on AXL signaling as a potential targeting strategy. Integrative transcriptome analyses were used to investigate how TP-0903, an AXL kinase inhibitor, influences redundant oncogenic pathways in metastatic lung cancer cells. CyTOF profiling revealed that AXL inhibition suppressed SMAD4/TGFβ signaling and induced JAK1-STAT3 signaling to compensate for the loss of AXL. Interestingly, high JAK1-STAT3 was associated with increased levels of AXL in treatment-naïve tumors. Tumors with high AXL, TGFβ, and JAK1 signaling concomitantly displayed CD133-mediated cancer stemness and hybrid epithelial-to-mesenchymal transition features in advanced-stage patients, suggesting greater potential for distant dissemination. Diffusion pseudotime analysis revealed cell-fate trajectories among four different categories that were linked to clinicopathologic features for each patient. Patient-derived organoids (PDO) obtained from tumors with high AXL and JAK1 were sensitive to TP-0903 and ruxolitinib (JAK inhibitor) treatments, supporting the CyTOF findings. This study shows that single-cell proteomic profiling of treatment-naïve lung tumors, coupled with ex vivo testing of PDOs, identifies continuous AXL, TGFβ, and JAK1-STAT3 signal activation in select tumors that may be targeted by combined AXL-JAK1 inhibition. SIGNIFICANCE: Single-cell proteomic profiling of clinical samples may facilitate the optimal selection of novel drug targets, interpretation of early-phase clinical trial data, and development of predictive biomarkers valuable for patient stratification., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
13. Spatial EGFR Dynamics and Metastatic Phenotypes Modulated by Upregulated EphB2 and Src Pathways in Advanced Prostate Cancer.
- Author
-
Liu YL, Horning AM, Lieberman B, Kim M, Lin CK, Hung CN, Chou CW, Wang CM, Lin CL, Kirma NB, Liss MA, Vasisht R, Perillo EP, Blocher K, Horng H, Taverna JA, Ruan J, Yankeelov TE, Dunn AK, Huang TH, Yeh HC, and Chen CL
- Abstract
Advanced prostate cancer is a very heterogeneous disease reflecting in diverse regulations of oncogenic signaling pathways. Aberrant spatial dynamics of epidermal growth factor receptor (EGFR) promote their dimerization and clustering, leading to constitutive activation in oncogenesis. The EphB2 and Src signaling pathways are associated with the reorganization of the cytoskeleton leading to malignancy, but their roles in regulating EGFR dynamics and activation are scarcely reported. Using single-particle tracking techniques, we found that highly phosphorylated EGFR in the advanced prostate cancer cell line, PC3, was associated with higher EGFR diffusivity, as compared with LNCaP and less aggressive DU145. The increased EGFR activation and biophysical dynamics were consistent with high proliferation, migration, and invasion. After performing single-cell RNA-seq on prostate cancer cell lines and circulating tumor cells from patients, we identified that upregulated gene expression in the EphB2 and Src pathways are associated with advanced malignancy. After dasatinib treatment or siRNA knockdowns of EphB2 or Src, the PC3 cells exhibited significantly lower EGFR dynamics, cell motility, and invasion. Partial inhibitory effects were also found in DU145 cells. The upregulation of parts of the EphB2 and Src pathways also predicts poor prognosis in the prostate cancer patient cohort of The Cancer Genome Atlas. Our results provide evidence that overexpression of the EphB2 and Src signaling pathways regulate EGFR dynamics and cellular aggressiveness in some advanced prostate cancer cells., Competing Interests: The authors declare that they have no competing interests.
- Published
- 2019
- Full Text
- View/download PDF
14. A novel algorithm for network-based prediction of cancer recurrence.
- Author
-
Ruan J, Jahid MJ, Gu F, Lei C, Huang YW, Hsu YT, Mutch DG, Chen CL, Kirma NB, and Huang TH
- Subjects
- CpG Islands, DNA, Neoplasm, Epigenomics, Female, Gene Expression Profiling, Gene Regulatory Networks, High-Throughput Nucleotide Sequencing, Humans, Models, Genetic, Prognosis, Protein Interaction Domains and Motifs, Sequence Analysis, DNA, Algorithms, Biomarkers, Tumor, DNA Methylation, Endometrial Neoplasms diagnosis, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology, Neoplasm Recurrence, Local
- Abstract
To develop accurate prognostic models is one of the biggest challenges in "omics"-based cancer research. Here, we propose a novel computational method for identifying dysregulated gene subnetworks as biomarkers to predict cancer recurrence. Applying our method to the DNA methylome of endometrial cancer patients, we identified a subnetwork consisting of differentially methylated (DM) genes, and non-differentially methylated genes, termed Epigenetic Connectors (EC), that are topologically important for connecting the DM genes in a protein-protein interaction network. The ECs are statistically significantly enriched in well-known tumorgenesis and metastasis pathways, and include known epigenetic regulators. Importantly, combining the DMs and ECs as features using a novel random walk procedure, we constructed a support vector machine classifier that significantly improved the prediction accuracy of cancer recurrence and outperformed several alternative methods, demonstrating the effectiveness of our network-based approach., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
15. Adipokines Deregulate Cellular Communication via Epigenetic Repression of Gap Junction Loci in Obese Endometrial Cancer.
- Author
-
Polusani SR, Huang YW, Huang G, Chen CW, Wang CM, Lin LL, Osmulski P, Lucio ND, Liu L, Hsu YT, Zhou Y, Lin CL, Aguilera-Barrantes I, Valente PT, Kost ER, Chen CL, Shim EY, Lee SE, Ruan J, Gaczynska ME, Yan P, Goodfellow PJ, Mutch DG, Jin VX, Nicholson BJ, Huang TH, and Kirma NB
- Subjects
- Adipose Tissue metabolism, Animals, Cell Movement, Cells, Cultured, Connexin 43 metabolism, Diet, High-Fat adverse effects, Endometrial Neoplasms etiology, Endometrial Neoplasms metabolism, Epithelial Cells metabolism, Epithelial Cells pathology, Female, Gap Junctions, Humans, Mice, Mice, Knockout, Obesity physiopathology, Stromal Cells metabolism, Stromal Cells pathology, Adipokines pharmacology, Adipose Tissue pathology, Cell Communication, Connexin 43 genetics, Endometrial Neoplasms pathology, Epigenetic Repression, Obesity complications
- Abstract
Emerging evidence indicates that adipose stromal cells (ASC) are recruited to enhance cancer development. In this study, we examined the role these adipocyte progenitors play relating to intercellular communication in obesity-associated endometrial cancer. This is particularly relevant given that gap junctions have been implicated in tumor suppression. Examining the effects of ASCs on the transcriptome of endometrial epithelial cells (EEC) in an in vitro coculture system revealed transcriptional repression of GJA1 (encoding the gap junction protein Cx43) and other genes related to intercellular communication. This repression was recapitulated in an obesity mouse model of endometrial cancer. Furthermore, inhibition of plasminogen activator inhibitor 1 (PAI-1), which was the most abundant ASC adipokine, led to reversal of cellular distribution associated with the GJA1 repression profile, suggesting that PAI-1 may mediate actions of ASC on transcriptional regulation in EEC. In an endometrial cancer cohort ( n = 141), DNA hypermethylation of GJA1 and related loci TJP2 and PRKCA was observed in primary endometrial endometrioid tumors and was associated with obesity. Pharmacologic reversal of DNA methylation enhanced gap-junction intercellular communication and cell-cell interactions in vitro . Restoring Cx43 expression in endometrial cancer cells reduced cellular migration; conversely, depletion of Cx43 increased cell migration in immortalized normal EEC. Our data suggest that persistent repression by ASC adipokines leads to promoter hypermethylation of GJA1 and related genes in the endometrium, triggering long-term silencing of these loci in endometrial tumors of obese patients. SIGNIFICANCE: Studies reveal that adipose-derived stem cells in endometrial cancer pathogenesis influence epigenetic repression of gap junction loci, which suggests targeting of gap junction activity as a preventive strategy for obesity-associated endometrial cancer., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
16. Single-Cell RNA-seq Reveals a Subpopulation of Prostate Cancer Cells with Enhanced Cell-Cycle-Related Transcription and Attenuated Androgen Response.
- Author
-
Horning AM, Wang Y, Lin CK, Louie AD, Jadhav RR, Hung CN, Wang CM, Lin CL, Kirma NB, Liss MA, Kumar AP, Sun L, Liu Z, Chao WT, Wang Q, Jin VX, Chen CL, and Huang TH
- Subjects
- Cell Line, Tumor, Humans, Male, Prostatic Neoplasms pathology, Androgens metabolism, Gene Expression Profiling methods, Prostatic Neoplasms genetics, RNA metabolism
- Abstract
Increasing evidence suggests the presence of minor cell subpopulations in prostate cancer that are androgen independent and poised for selection as dominant clones after androgen deprivation therapy. In this study, we investigated this phenomenon by stratifying cell subpopulations based on transcriptome profiling of 144 single LNCaP prostate cancer cells treated or untreated with androgen after cell-cycle synchronization. Model-based clustering of 397 differentially expressed genes identified eight potential subpopulations of LNCaP cells, revealing a previously unappreciable level of cellular heterogeneity to androgen stimulation. One subpopulation displayed stem-like features with a slower cell doubling rate, increased sphere formation capability, and resistance to G
2 -M arrest induced by a mitosis inhibitor. Advanced growth of this subpopulation was associated with enhanced expression of 10 cell-cycle-related genes ( CCNB2, DLGAP5, CENPF, CENPE, MKI67, PTTG1, CDC20, PLK1, HMMR , and CCNB1 ) and decreased dependence upon androgen receptor signaling. In silico analysis of RNA-seq data from The Cancer Genome Atlas further demonstrated that concordant upregulation of these genes was linked to recurrent prostate cancers. Analysis of receiver operating characteristic curves implicates aberrant expression of these genes and could be useful for early identification of tumors that subsequently develop biochemical recurrence. Moreover, this single-cell approach provides a better understanding of how prostate cancer cells respond heterogeneously to androgen deprivation therapies and reveals characteristics of subpopulations resistant to this treatment. Significance: Illustrating the challenge in treating cancers with targeted drugs, which by selecting for drug resistance can drive metastatic progression, this study characterized the plasticity and heterogeneity of prostate cancer cells with regard to androgen dependence, defining the character or minor subpopulations of androgen-independent cells that are poised for clonal selection after androgen-deprivation therapy. Cancer Res; 78(4); 853-64. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
17. EGFR-Dependent Regulated Intramembrane Proteolysis of EpCAM-Response.
- Author
-
Hsu YT, Osmulski P, Wang Y, Huang YW, Liu L, Ruan J, Jin VX, Kirma NB, Gaczynska ME, and Huang TH
- Subjects
- Antigens, Neoplasm, Cell Membrane, ErbB Receptors, Humans, Epithelial Cell Adhesion Molecule, Proteolysis
- Published
- 2017
- Full Text
- View/download PDF
18. EpCAM-Regulated Transcription Exerts Influences on Nanomechanical Properties of Endometrial Cancer Cells That Promote Epithelial-to-Mesenchymal Transition.
- Author
-
Hsu YT, Osmulski P, Wang Y, Huang YW, Liu L, Ruan J, Jin VX, Kirma NB, Gaczynska ME, and Huang TH
- Subjects
- Biomechanical Phenomena, Cell Line, Tumor, Cell Nucleus metabolism, Epidermal Growth Factor pharmacology, Female, Gene Editing, Humans, Lymphoid Enhancer-Binding Factor 1 physiology, Microscopy, Atomic Force, Tight Junctions physiology, Endometrial Neoplasms pathology, Epithelial Cell Adhesion Molecule physiology, Epithelial-Mesenchymal Transition, Transcription, Genetic
- Abstract
Overexpression of epithelial cell adhesion molecule (EpCAM) has been implicated in advanced endometrial cancer, but its roles in this progression remain to be elucidated. In addition to its structural role in modulating cell-surface adhesion, here we demonstrate that EpCAM is a regulatory molecule in which its internalization into the nucleus turns on a transcription program. Activation of EGF/EGFR signal transduction triggered cell-surface cleavage of EpCAM, leading to nuclear internalization of its cytoplasmic domain EpICD. ChIP-seq analysis identified target genes that are coregulated by EpICD and its transcription partner, LEF-1. Network enrichment analysis further uncovered a group of 105 genes encoding functions for tight junction, adherent, and cell migration. Furthermore, nanomechanical analysis by atomic force microscopy revealed increased softness and decreased adhesiveness of EGF-stimulated cancer cells, implicating acquisition of an epithelial-mesenchymal transition (EMT) phenotype. Thus, genome editing of EpCAM could be associated with altering these nanomechanical properties towards a less aggressive phenotype. Using this integrative genomic-biophysical approach, we demonstrate for the first time an intricate relationship between EpCAM-regulated transcription and altered biophysical properties of cells that promote EMT in advanced endometrial cancer. Cancer Res; 76(21); 6171-82. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
19. Spatiotemporal control of estrogen-responsive transcription in ERα-positive breast cancer cells.
- Author
-
Hsu PY, Hsu HK, Hsiao TH, Ye Z, Wang E, Profit AL, Jatoi I, Chen Y, Kirma NB, Jin VX, Sharp ZD, and Huang TH
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation genetics, Chromobox Protein Homolog 5, Chromosomal Proteins, Non-Histone metabolism, Chromosomes, Human, Pair 20 genetics, Epigenesis, Genetic, Humans, Janus Kinases metabolism, Kruppel-Like Transcription Factors genetics, STAT Transcription Factors metabolism, Sequence Deletion, Signal Transduction genetics, Spatio-Temporal Analysis, Survival Analysis, Breast Neoplasms pathology, Computational Biology, Estrogen Receptor alpha metabolism, Estrogens metabolism, Response Elements genetics, Transcription, Genetic genetics
- Abstract
Recruitment of transcription machinery to target promoters for aberrant gene expression has been well studied, but underlying control directed by distant-acting enhancers remains unclear in cancer development. Our previous study demonstrated that distant estrogen response elements (DEREs) located on chromosome 20q13 are frequently amplified and translocated to other chromosomes in ERα-positive breast cancer cells. In this study, we used three-dimensional interphase fluorescence in situ hybridization to decipher spatiotemporal gathering of multiple DEREs in the nucleus. Upon estrogen stimulation, scattered 20q13 DEREs were mobilized to form regulatory depots for synchronized gene expression of target loci. A chromosome conformation capture assay coupled with chromatin immunoprecipitation further uncovered that ERα-bound regulatory depots are tethered to heterochromatin protein 1 (HP1) for coordinated chromatin movement and histone modifications of target loci, resulting in transcription repression. Neutralizing HP1 function dysregulated the formation of DERE-involved regulatory depots and transcription inactivation of candidate tumor-suppressor genes. Deletion of amplified DEREs using the CRISPR/Cas9 genomic-editing system profoundly altered transcriptional profiles of proliferation-associated signaling networks, resulting in reduction of cancer cell growth. These findings reveal a formerly uncharacterized feature wherein multiple copies of the amplicon congregate as transcriptional units in the nucleus for synchronous regulation of function-related loci in tumorigenesis. Disruption of their assembly can be a new strategy for treating breast cancers and other malignancies.
- Published
- 2016
- Full Text
- View/download PDF
20. Aromatase expression increases the survival and malignancy of estrogen receptor positive breast cancer cells.
- Author
-
Mukhopadhyay KD, Liu Z, Bandyopadhyay A, Kirma NB, Tekmal RR, Wang S, and Sun LZ
- Subjects
- Animals, Anoikis genetics, Aromatase metabolism, Aromatase Inhibitors pharmacology, Bone Neoplasms drug therapy, Bone Neoplasms enzymology, Bone Neoplasms secondary, Breast Neoplasms drug therapy, Breast Neoplasms enzymology, Breast Neoplasms pathology, Carcinoma, Ductal drug therapy, Carcinoma, Ductal enzymology, Carcinoma, Ductal secondary, Cell Line, Tumor, Estrogen Receptor alpha metabolism, Estrogens biosynthesis, Female, Humans, Letrozole, Mice, Mice, Nude, Neoplasm Transplantation, Nitriles pharmacology, Signal Transduction, Testosterone metabolism, Testosterone pharmacology, Triazoles pharmacology, Tumor Cells, Cultured, Aromatase genetics, Bone Neoplasms genetics, Breast Neoplasms genetics, Carcinoma, Ductal genetics, Estrogen Receptor alpha genetics, Gene Expression Regulation, Neoplastic
- Abstract
In postmenopausal women, local estrogen produced by adipose stromal cells in the breast is believed to support estrogen receptor alpha (ERα) positive breast cancer cell survival and growth. This raises the question of how the ERα positive metastatic breast cancer cells survive after they enter blood and lymph circulation, where estrogen level is very low in postmenopausal women. In this study, we show that the aromatase expression increased when ERα positive breast cancer cells were cultured in suspension. Furthermore, treatment with the aromatase substrate, testosterone, inhibited suspension culture-induced apoptosis whereas an aromatase inhibitor attenuated the effect of testosterone suggesting that suspended circulating ERα positive breast cancer cells may up-regulate intracrine estrogen activity for survival. Consistent with this notion, a moderate level of ectopic aromatase expression rendered a non-tumorigenic ERα positive breast cancer cell line not only tumorigenic but also metastatic in female nude mice without exogenous estrogen supplementation. The increased malignant phenotype was confirmed to be due to aromatase expression as the growth of orthotopic tumors regressed with systemic administration of an aromatase inhibitor. Thus, our study provides experimental evidence that aromatase plays an important role in the survival of metastatic ERα breast cancer cells by suppressing anoikis.
- Published
- 2015
- Full Text
- View/download PDF
21. Genome-wide DNA methylation analysis reveals estrogen-mediated epigenetic repression of metallothionein-1 gene cluster in breast cancer.
- Author
-
Jadhav RR, Ye Z, Huang RL, Liu J, Hsu PY, Huang YW, Rangel LB, Lai HC, Roa JC, Kirma NB, Huang TH, and Jin VX
- Abstract
Background: Recent genome-wide analysis has shown that DNA methylation spans long stretches of chromosome regions consisting of clusters of contiguous CpG islands or gene families. Hypermethylation of various gene clusters has been reported in many types of cancer. In this study, we conducted methyl-binding domain capture (MBDCap) sequencing (MBD-seq) analysis on a breast cancer cohort consisting of 77 patients and 10 normal controls, as well as a panel of 38 breast cancer cell lines., Results: Bioinformatics analysis determined seven gene clusters with a significant difference in overall survival (OS) and further revealed a distinct feature that the conservation of a large gene cluster (approximately 70 kb) metallothionein-1 (MT1) among 45 species is much lower than the average of all RefSeq genes. Furthermore, we found that DNA methylation is an important epigenetic regulator contributing to gene repression of MT1 gene cluster in both ERα positive (ERα+) and ERα negative (ERα-) breast tumors. In silico analysis revealed much lower gene expression of this cluster in The Cancer Genome Atlas (TCGA) cohort for ERα + tumors. To further investigate the role of estrogen, we conducted 17β-estradiol (E2) and demethylating agent 5-aza-2'-deoxycytidine (DAC) treatment in various breast cancer cell types. Cell proliferation and invasion assays suggested MT1F and MT1M may play an anti-oncogenic role in breast cancer., Conclusions: Our data suggests that DNA methylation in large contiguous gene clusters can be potential prognostic markers of breast cancer. Further investigation of these clusters revealed that estrogen mediates epigenetic repression of MT1 cluster in ERα + breast cancer cell lines. In all, our studies identify thousands of breast tumor hypermethylated regions for the first time, in particular, discovering seven large contiguous hypermethylated gene clusters.
- Published
- 2015
- Full Text
- View/download PDF
22. Promoter hypomethylation of EpCAM-regulated bone morphogenetic protein gene family in recurrent endometrial cancer.
- Author
-
Hsu YT, Gu F, Huang YW, Liu J, Ruan J, Huang RL, Wang CM, Chen CL, Jadhav RR, Lai HC, Mutch DG, Goodfellow PJ, Thompson IM, Kirma NB, and Huang TH
- Subjects
- Antigens, Neoplasm metabolism, Base Sequence, Cell Adhesion Molecules metabolism, Cell Line, Tumor, CpG Islands genetics, Disease-Free Survival, Epidermal Growth Factor metabolism, Epidermal Growth Factor pharmacology, Epithelial Cell Adhesion Molecule, Epithelial-Mesenchymal Transition drug effects, Female, Humans, Neoplasm Recurrence, Local genetics, Promoter Regions, Genetic, RNA Interference, RNA, Small Interfering, Sequence Analysis, DNA, Survival, Transcriptional Activation drug effects, Antigens, Neoplasm genetics, Bone Morphogenetic Proteins genetics, Cell Adhesion Molecules genetics, DNA Methylation, Endometrial Neoplasms genetics
- Abstract
Purpose: Epigenetic regulation by promoter methylation plays a key role in tumorigenesis. Our goal was to investigate whether altered DNA methylation signatures associated with oncogenic signaling delineate biomarkers predictive of endometrial cancer recurrence., Experimental Design: Methyl-CpG-capture sequencing was used for global screening of aberrant DNA methylation in our endometrial cancer cohort, followed by validation in an independent The Cancer Genome Atlas (TCGA) cohort. Bioinformatics as well as functional analyses in vitro, using RNA interference (RNAi) knockdown, were performed to examine regulatory mechanisms of candidate gene expression and contribution to aggressive phenotype, such as epithelial-mesenchymal transition (EMT)., Results: We identified 2,302 hypermethylated loci in endometrial tumors compared with control samples. Bone morphogenetic protein (BMP) family genes, including BMP1, 2, 3, 4, and 7, were among the frequently hypermethylated loci. Interestingly, BMP2, 3, 4, and 7 were less methylated in primary tumors with subsequent recurrence and in patients with shorter disease-free interval compared with nonrecurrent tumors, which was validated and associated with poor survival in the TCGA cohort (BMP4, P = 0.009; BMP7, P = 0.007). Stimulation of endometrial cancer cells with epidermal growth factor (EGF) induced EMT and transcriptional activation of these genes, which was mediated by the epithelial cell adhesion molecule (EpCAM). EGF signaling was implicated in maintaining the promoters of candidate BMP genes in an active chromatin configuration and thus subject to transcriptional activation., Conclusions: Hypomethylation signatures of candidate BMP genes associated with EpCAM-mediated expression present putative biomarkers predictive of poor survival in endometrial cancer., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
23. Amplification of distant estrogen response elements deregulates target genes associated with tamoxifen resistance in breast cancer.
- Author
-
Hsu PY, Hsu HK, Lan X, Juan L, Yan PS, Labanowska J, Heerema N, Hsiao TH, Chiu YC, Chen Y, Liu Y, Li L, Li R, Thompson IM, Nephew KP, Sharp ZD, Kirma NB, Jin VX, and Huang TH
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Breast Neoplasms metabolism, Cell Line, Tumor, Drug Resistance, Neoplasm, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Female, Gene Amplification, Genomics, Humans, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Estrogens metabolism, Gene Expression Regulation, Neoplastic, Response Elements, Tamoxifen pharmacology
- Abstract
A causal role of gene amplification in tumorigenesis is well known, whereas amplification of DNA regulatory elements as an oncogenic driver remains unclear. In this study, we integrated next-generation sequencing approaches to map distant estrogen response elements (DEREs) that remotely control the transcription of target genes through chromatin proximity. Two densely mapped DERE regions located on chromosomes 17q23 and 20q13 were frequently amplified in estrogen receptor-α-positive luminal breast cancer. These aberrantly amplified DEREs deregulated target gene expression potentially linked to cancer development and tamoxifen resistance. Progressive accumulation of DERE copies was observed in normal breast progenitor cells chronically exposed to estrogenic chemicals. These findings may extend to other DNA regulatory elements, the amplification of which can profoundly alter target transcriptome during tumorigenesis., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
24. Epigenetic deregulation of the anaplastic lymphoma kinase gene modulates mesenchymal characteristics of oral squamous cell carcinomas.
- Author
-
Huang TT, Gonzales CB, Gu F, Hsu YT, Jadhav RR, Wang CM, Redding SW, Tseng CE, Lee CC, Thompson IM, Chen HR, Huang TH, and Kirma NB
- Subjects
- Anaplastic Lymphoma Kinase, Carcinogenesis genetics, Carcinogenesis metabolism, Carcinogenesis pathology, Carcinoma, Squamous Cell metabolism, Cell Growth Processes physiology, Cell Line, Tumor, CpG Islands, DNA Methylation, Disease Progression, Epigenesis, Genetic, Humans, Lymphatic Metastasis, Mesoderm metabolism, Mouth Mucosa metabolism, Mouth Mucosa pathology, Mouth Neoplasms metabolism, Neoplasm Invasiveness, Promoter Regions, Genetic, Receptor Protein-Tyrosine Kinases metabolism, Transcriptional Activation, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Mesoderm pathology, Mouth Neoplasms genetics, Mouth Neoplasms pathology, Receptor Protein-Tyrosine Kinases genetics
- Abstract
DNA hypermethylation of promoter CpG islands is associated with epigenetic silencing of tumor suppressor genes in oral squamous cell carcinomas (OSCCs). We used a methyl-CpG-binding domain protein capture method coupled with next-generation sequencing (MBDCap-seq) to survey global DNA methylation patterns in OSCCs with and without nodal metastasis and normal mucosa (total n = 58). Of 1462 differentially methylated CpG islands identified in OSCCs relative to normal controls, MBDCap-seq profiling uncovered 359 loci linked to lymph node metastasis. Interactive network analysis revealed a subset of these loci (n = 23), including the anaplastic lymphoma kinase (ALK) gene, are potential regulators and effectors of invasiveness and metastatic progression. Promoter methylation of ALK was preferentially observed in OSCCs without node metastasis, whereas relatively lower methylation levels were present in metastatic tumors, implicating an active state of ALK transcription in the latter group. The OSCC cell line, SCC4, displayed reduced ALK expression that corresponded to extensive promoter CpG island methylation. SCC4 treatment with demethylating agents induced ALK expression and increased invasion and migration characteristics. Inhibition of ALK activity in OSCC cells with high ALK expression (CAL27, HSC3 and SCC25), decreased cell growth and resulted in changes in invasive potential and mesenchymal marker expression that were cell-line dependent. Although ALK is susceptible to epigenetic silencing during oral tumorigenesis, overwriting this default state may be necessary for modulating invasive processes involved in nodal metastases. Given the complex response of OSCC cells to ALK inhibition, future studies are required to assess the feasibility of targeting ALK to treat invasive OSCCs.
- Published
- 2013
- Full Text
- View/download PDF
25. Comprehensive methylome analysis of ovarian tumors reveals hedgehog signaling pathway regulators as prognostic DNA methylation biomarkers.
- Author
-
Huang RL, Gu F, Kirma NB, Ruan J, Chen CL, Wang HC, Liao YP, Chang CC, Yu MH, Pilrose JM, Thompson IM, Huang HC, Huang TH, Lai HC, and Nephew KP
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Carcinoma, Ovarian Epithelial, Cell Line, Tumor, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Disease-Free Survival, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Neoplasms, Glandular and Epithelial drug therapy, Neoplasms, Glandular and Epithelial pathology, Nerve Tissue Proteins metabolism, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Promoter Regions, Genetic, Proportional Hazards Models, Survival Analysis, Transcription Factors metabolism, DNA Methylation, Hedgehog Proteins metabolism, Neoplasms, Glandular and Epithelial genetics, Neoplasms, Glandular and Epithelial metabolism, Nerve Tissue Proteins genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Signal Transduction, Transcription Factors genetics
- Abstract
Women with advanced stage ovarian cancer (OC) have a five-year survival rate of less than 25%. OC progression is associated with accumulation of epigenetic alterations and aberrant DNA methylation in gene promoters acts as an inactivating "hit" during OC initiation and progression. Abnormal DNA methylation in OC has been used to predict disease outcome and therapy response. To globally examine DNA methylation in OC, we used next-generation sequencing technology, MethylCap-sequencing, to screen 75 malignant and 26 normal or benign ovarian tissues. Differential DNA methylation regions (DMRs) were identified, and the Kaplan-Meier method and Cox proportional hazard model were used to correlate methylation with clinical endpoints. Functional role of specific genes identified by MethylCap-sequencing was examined in in vitro assays. We identified 577 DMRs that distinguished (p < 0.001) malignant from non-malignant ovarian tissues; of these, 63 DMRs correlated (p < 0.001) with poor progression free survival (PFS). Concordant hypermethylation and corresponding gene silencing of sonic hedgehog pathway members ZIC1 and ZIC4 in OC tumors was confirmed in a panel of OC cell lines, and ZIC1 and ZIC4 repression correlated with increased proliferation, migration and invasion. ZIC1 promoter hypermethylation correlated (p < 0.01) with poor PFS. In summary, we identified functional DNA methylation biomarkers significantly associated with clinical outcome in OC and suggest our comprehensive methylome analysis has significant translational potential for guiding the design of future clinical investigations targeting the OC epigenome. Methylation of ZIC1, a putative tumor suppressor, may be a novel determinant of OC outcome.
- Published
- 2013
- Full Text
- View/download PDF
26. Raf-1, a potential therapeutic target, mediates early steps in endometriosis lesion development by endometrial epithelial and stromal cells.
- Author
-
De La Garza EM, Binkley PA, Ganapathy M, Krishnegowda NK, Tekmal RR, Schenken RS, and Kirma NB
- Subjects
- Animals, Cell Line, Cells, Cultured, Endometriosis pathology, Female, Humans, Indoles pharmacology, Mice, Phenols pharmacology, Phosphorylation drug effects, Proto-Oncogene Proteins c-raf antagonists & inhibitors, Proto-Oncogene Proteins c-raf genetics, Transforming Growth Factor beta pharmacology, Endometriosis metabolism, Endometrium metabolism, Endometrium pathology, Epithelial Cells metabolism, Epithelial Cells pathology, Proto-Oncogene Proteins c-raf metabolism, Stromal Cells metabolism, Stromal Cells pathology
- Abstract
Endometriosis is a hormone-sensitive gynecological disorder characterized by the benign growth of endometrial-like tissue in the pelvic cavity. Endometriotic lesions composed of endometrial stromal cells (ESC) and glandular epithelial cells (EEC) are thought to arise from menstrual endometrial tissue reaching the pelvic cavity via retrograde menstruation. The cause of endometriotic lesion formation is still not clear. Recent evidence suggest that cytokines may play a role in the early development of endometriosis lesions. Because cytokines and growth factors signal via the v-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) kinase pathway, we have examined the role of Raf-1 in early steps of endometriosis lesion formation, specifically attachment of endometrial cells to peritoneal mesothelial cells (PMC) and invasion of endometrial cells through PMC (trans-mesothelial invasion). Raf-1 antagonist GW5074 decreased attachment to PMC and trans-mesothelial invasion by primary EEC and ESC. Raf-1 also mediated TGFβ-induced trans-mesothelial invasion by the established, low-invasive EEC line EM42. TGFβ treatment of EEC resulted in Raf-1 phosphorylation at S338 and phosphorylation of ERK, suggesting that TGFβ activates Raf-1 signaling in these cells. GW5074 had little effect on ESC proliferation but inhibited EEC growth significantly under reduced serum conditions. Antagonizing Raf-1 activity and expression via GW5074 and specific Raf-1 small interfering RNA, respectively, did not alter EEC resistance to growth inhibition by TGFβ. Raf-1 inhibition blocked induction of EEC growth by epidermal growth factor. Our data suggest that Raf-1 may mediate pathologic steps involved in early endometriosis lesion formation and may be a mediator of TGFβ and epidermal growth factor actions in endometriosis.
- Published
- 2012
- Full Text
- View/download PDF
27. Estrogen receptor-beta mediates the protective effects of aromatase induction in the MMTV-Her-2/neu x aromatase double transgenic mice.
- Author
-
Nair HB, Perla RP, Kirma NB, Krishnegowda NK, Ganapathy M, Rajhans R, Nair SS, Saikumar P, Vadlamudi RK, and Tekmal RR
- Subjects
- Animals, Aromatase metabolism, Cell Cycle genetics, Cell Proliferation, Disease-Free Survival, Female, Gene Expression Regulation, Genes, Tumor Suppressor, Mammary Neoplasms, Animal mortality, Mammary Neoplasms, Animal pathology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental mortality, Mice, Mice, Transgenic, Mitogen-Activated Protein Kinase Kinases metabolism, Phosphorylation, Signal Transduction genetics, Aromatase genetics, Estrogen Receptor beta metabolism, Genes, erbB-2
- Abstract
Breast cancers amplified for the tyrosine kinase receptor Her-2/neu constitute ~30% of advanced breast cancer cases, and are characterized by hormone independence and aggressive growth, implicating this pathway in breast oncogenesis. The induction of Her-2/neu leads to tumor development in 60% of transgenic mice. We have previously examined the effects of estrogen in the MMTV-Her-2/neu background by generating the MMTV-Her-2/neu x aromatase double transgenic mouse strain. MMTV-Her-2/neu x aromatase mice developed fewer mammary tumors than the Her-2/neu parental strain. Our present data show the induction of several estrogen-related genes, including the tumor suppressors BRCA1 and p53, and a decrease in several angiogenic factors. The phosphorylated forms of MAPK p42/44 and AKT were lower in the MMTV-Her-2/neu x aromatase double transgenic mice compared to the MMTV-Her-2/neu parental strain; conversely, phospho-p38 levels were higher in the double transgenic strain. The ERβ-selective antagonist THC reversed these changes. The regulation of these factors by ERβ was confirmed in clones of MCF7 breast cancer cells overexpressing Her-2/neu in combination with ERβ, suggesting that ERβ may play a direct role in regulating MAPK and AKT pathways. In summary, the data suggest that ERβ may play a major role in decreasing tumorigenesis and that it may affect breast cancer cell proliferation and survival by altering MAPK and AKT activation as well as modulation of tumor suppressor and angiogenesis factors. Treatment with selective ERβ agonist may provide therapeutic advantages for the treatment and prevention of breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
28. Colony-stimulating factor-1 exerts direct effects on the proliferation and invasiveness of endometrial epithelial cells.
- Author
-
Aligeti S, Kirma NB, Binkley PA, Schenken RS, and Tekmal RR
- Subjects
- Analysis of Variance, Autocrine Communication, Cell Adhesion, Cell Line, Coculture Techniques, Endometriosis pathology, Endometrium pathology, Epithelial Cells pathology, Female, Humans, Macrophage Colony-Stimulating Factor genetics, Peritoneum metabolism, Peritoneum pathology, RNA Interference, Time Factors, Cell Movement, Cell Proliferation, Endometriosis metabolism, Endometrium metabolism, Epithelial Cells metabolism, Macrophage Colony-Stimulating Factor metabolism
- Abstract
Although macrophage colony-stimulating factor (CSF-1) has been suggested to play a role in maintaining the chronic inflammatory response in endometriosis, our data suggest that CSF-1 may also play a role in early endometriosis lesion formation. We have shown that CSF-1, in an autocrine fashion, has a direct effect on endometrial epithelial cell proliferation and attachment to peritoneal mesothelial cells, early steps in endometriosis lesion formation on the peritoneum., (Copyright © 2011 American Society for Reproductive Medicine. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
29. Imatinib decreases endometrial stromal cell transmesothial migration and proliferation in the extracellular matrix of modeled peritoneum.
- Author
-
Griffith JS, Binkley PA, Kirma NB, Schenken RS, Witz CA, and Tekmal RR
- Subjects
- Antineoplastic Agents pharmacology, Benzamides, Cell Adhesion drug effects, Cell Adhesion physiology, Cell Culture Techniques, Cells, Cultured, Down-Regulation drug effects, Drug Evaluation, Preclinical, Endometrium pathology, Endometrium physiology, Epithelium drug effects, Epithelium pathology, Extracellular Matrix metabolism, Extracellular Matrix physiology, Female, Humans, Imatinib Mesylate, Models, Biological, Peritoneum physiology, Stromal Cells pathology, Stromal Cells physiology, Cell Proliferation drug effects, Endometrium drug effects, Extracellular Matrix drug effects, Peritoneum drug effects, Piperazines pharmacology, Pyrimidines pharmacology, Stromal Cells drug effects, Transendothelial and Transepithelial Migration drug effects
- Abstract
Objective: To characterize imatinib's effect on endometrial stromal cell (ESC) attachment, proliferation, and invasion in modeled peritoneum., Design: In vitro study., Setting: Academic medical center., Patient(s): Twelve normally cycling women., Intervention(s): Imatinib treatment in ESCs from women without endometriosis., Main Outcome Measure(s): Rate of ESC attachment, proliferation, and invasion., Result(s): Imatinib treatment at 10 μM had no effect on ESC attachment. Treatment with 0.5 μM, 2 μM, and 10 μM of imatinib reduced ESC proliferation by 30%, 72%, and 76%, respectively. The 0.1 μM dose of imatinib had no effect on proliferation. Treatment with 5 μM and 10 μM of imatinib reduced ESC invasion by 30% and 73%, respectively. The 2 μM dose had no effect on invasion., Conclusion(s): Imatinib treatment reduces ESC proliferation and invasion in modeled peritoneum without altering attachment. Imatinib may have a therapeutic role in endometriosis treatment., (Copyright © 2010 American Society for Reproductive Medicine. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
30. Induction of endometrial epithelial cell invasion and c-fms expression by transforming growth factor beta.
- Author
-
Liu YG, Tekmal RR, Binkley PA, Nair HB, Schenken RS, and Kirma NB
- Subjects
- Cell Adhesion drug effects, Cell Line, Cells, Cultured, Chromatin Immunoprecipitation, Enzyme Inhibitors pharmacology, Epithelial Cells drug effects, Female, Flow Cytometry, Gene Expression drug effects, Humans, Imidazoles pharmacology, Promoter Regions, Genetic genetics, Pyridines pharmacology, Receptor, Macrophage Colony-Stimulating Factor genetics, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta physiology, Endometrium cytology, Epithelial Cells cytology, Receptor, Macrophage Colony-Stimulating Factor metabolism, Transforming Growth Factor beta pharmacology
- Abstract
Transforming growth factor beta 1 (TGF-beta1) levels are increased in the peritoneal fluid of endometriosis patients, and endometrial cells express TGF-beta signaling components; however, little is known regarding the role of TGF-beta in endometriosis. Our objective was to examine the effects of TGF-beta1 on (i) the expression of macrophage colony-stimulating factor receptor encoded by the c-fms gene, (ii) transmesothelial invasiveness of endometrial cells, (iii) cellular proliferation and (iv) attachment to peritoneal mesothelial cells (PMCs). Effects of TGF-beta1 on c-fms mRNA expression were determined by real-time RT-PCR and c-fms cell-surface expression by flow cytometry. Effects of TGF-beta1 on the invasiveness of the immortalized endometrial epithelial cell (EEC) line EM42 and primary EECs were examined using a three-dimensional in vitro system modeling the peritoneum. Cellular proliferation and attachment to PMCs were also examined using established techniques. TGF-beta1 had little or no effect on cellular proliferation and endometrial cell attachment to PMCs. TGF-beta1 significantly induced the expression of c-fms mRNA and c-fms cell-surface expression. TGF-beta1 enhanced transmesothelial invasion by EM42 cells and EECs. Antagonists of TGF-beta1 signaling significantly inhibited both the induction of c-fms expression and cellular invasiveness, suggesting that additional studies are warranted to assess the therapeutic potential of TGF-beta antagonists in endometriosis.
- Published
- 2009
- Full Text
- View/download PDF
31. Modulation of in situ estrogen synthesis by proline-, glutamic acid-, and leucine-rich protein-1: potential estrogen receptor autocrine signaling loop in breast cancer cells.
- Author
-
Rajhans R, Nair HB, Nair SS, Cortez V, Ikuko K, Kirma NB, Zhou D, Holden AE, Brann DW, Chen S, Tekmal RR, and Vadlamudi RK
- Subjects
- Aromatase biosynthesis, Aromatase genetics, Aromatase metabolism, Autocrine Communication, Breast Neoplasms enzymology, Breast Neoplasms genetics, Carcinoma, Ductal, Breast enzymology, Carcinoma, Ductal, Breast genetics, Cell Line, Tumor, Chromatin Immunoprecipitation, Co-Repressor Proteins, Female, Gene Expression Regulation, Neoplastic, Humans, Immunohistochemistry, Middle Aged, RNA, Messenger biosynthesis, RNA, Messenger genetics, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Reverse Transcriptase Polymerase Chain Reaction, Tissue Array Analysis, Trans-Activators biosynthesis, Trans-Activators genetics, Transcription Factors, Breast Neoplasms metabolism, Carcinoma, Ductal, Breast metabolism, Estrogens biosynthesis, Trans-Activators metabolism
- Abstract
In situ estrogen synthesis is implicated in tumor cell proliferation through autocrine or paracrine mechanisms especially in postmenopausal women. Several recent studies demonstrated activity of aromatase, an enzyme that plays a critical role in estrogen synthesis in breast tumors. Proline-, glutamic acid-, and leucine-rich protein-1 (PELP1/MNAR) is an estrogen receptor (ER) coregulator, and its expression is deregulated in breast tumors. In this study, we examined whether PELP1 promotes tumor growth by promoting local estrogen synthesis using breast cancer cells (MCF7) that stably overexpress PELP1. Immunohistochemistry revealed increased aromatase expression in MCF7-PELP1-induced xenograft tumors. Real-time PCR analysis showed enhanced activation of the aromatase promoter in MCF7-PELP1 clones compared with MCF7 cells. Using a tritiated-water release assay, we demonstrated that MCF7-PELP1 clones exhibit increased aromatase activity compared with control MCF-7 cells. PELP1 deregulation uniquely up-regulated aromatase expression via activation of aromatase promoter I.3/II, and growth factor signaling enhanced PELP1 activation of aromatase. PELP1-mediated induction of aromatase requires functional Src and phosphatidylinositol-3-kinase pathways. Mechanistic studies revealed that PELP1 interactions with ER-related receptor-alpha and proline-rich nuclear receptor coregulatory protein 2 lead to activation of aromatase. Immunohistochemistry analysis of breast tumor array showed increased expression of aromatase in ductal carcinoma in situ and node-positive tumors compared with no or weak expression in normal breast tissue. Fifty-four percent (n = 79) of PELP1-overexpressing tumors also overexpressed aromatase compared with 36% (n = 47) in PELP1 low-expressing tumors. Our results suggest that PELP1 regulation of aromatase represents a novel mechanism for in situ estrogen synthesis leading to tumor proliferation by autocrine loop and open a new avenue for ablating local aromatase activity in breast tumors.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.