Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 53
1.
Oncotarget ; 13: 1350-1358, 2022 12 20.
Article En | MEDLINE | ID: mdl-36537914

One of the central challenges for cancer therapy is the identification of factors in the tumor microenvironment that increase tumor progression and immune tolerance. In breast cancer, fibrosis is a histopathologic criterion for invasive cancer and poor survival that results from inflammatory factors and remodeling of the extracellular matrix to produce an immune tolerant microenvironment. To determine whether tolerance is associated with the immune checkpoint, Programmed Cell Death 1 (PD-1), NeuT/ATTAC mice, a conditional model of mammary fibrosis that we recently developed, were administered a murine-specific anti-PD-1 mAb related to pembrolizumab, and drug response was monitored by tumor development, imaging mass cytometry, immunohistochemistry and tumor gene expression by RNAseq. Tumor progression in NeuT/ATTAC mice was unaffected by weekly injection of anti-PD-1 over four months. Insensitivity to anti-PD-1 was associated with several processes, including increased tumor-associated macrophages (TAM), epithelial to mesenchymal transition (EMT), fibroblast proliferation, an enhanced extracellular matrix and the Wnt signaling pathway, including increased expression of Fzd5, Wnt5a, Vimentin, Mmp3, Col2a1 and Tgfß1. These results suggest potential therapeutic avenues that may enhance PD-1 immune checkpoint sensitivity, including the use of tumor microenvironment targeted agents and Wnt pathway inhibitors.


Antineoplastic Agents , Neoplasms , Mice , Animals , Wnt Signaling Pathway , Epithelial-Mesenchymal Transition , Antineoplastic Agents/pharmacology , Macrophages , Tumor Microenvironment , Cell Line, Tumor
2.
PLoS One ; 17(11): e0277098, 2022.
Article En | MEDLINE | ID: mdl-36327243

OBJECTIVE: To determine the expression profile of microRNAs in the peripheral blood of pregnant women with preterm premature rupture of membranes (PPROM) compared to that of healthy pregnant women. STUDY DESIGN: This was a pilot study with case-control design in pregnant patients enrolled between January 2017 and June 2019. Patients with healthy pregnancies and those affected by PPROM between 20- and 33+6 weeks of gestation were matched by gestational age and selected for inclusion to the study. Patients were excluded for multiple gestation and presence of a major obstetrical complication such as preeclampsia, diabetes, fetal growth restriction and stillbirth. A total of ten (n = 10) controls and ten (n = 10) patients with PPROM were enrolled in the study. Specimens were obtained before administration of betamethasone or intravenous antibiotics. MicroRNA expression was analyzed for 800 microRNAs in each sample using the NanoString nCounter Expression Assay. Differential expression was calculated after normalization and log2- transformation using the false discovery rate (FDR) method at an alpha level of 5%. RESULTS: Demographic characteristics were similar between the two groups. Of the 800 miRNAs analyzed, 116 were differentially expressed after normalization. However, only four reached FDR-adjusted statistical significance. Pregnancies affected by PPROM were characterized by upregulation of miR-199a-5p, miR-130a-3p and miR-26a-5p and downregulation of miR-513b-5p (FDR adjusted p-values <0.05). The differentially expressed microRNAs participate in pathways associated with altered collagen and matrix metalloprotease expression in the extracellular matrix. CONCLUSION: Patients with PPROM have a distinct peripheral blood microRNA profile compared to healthy pregnancies as measured by the NanoString Expression Assay.


Fetal Membranes, Premature Rupture , MicroRNAs , Infant, Newborn , Humans , Pregnancy , Female , MicroRNAs/metabolism , Pilot Projects , Fetal Membranes, Premature Rupture/genetics , Gestational Age , Pregnancy, Multiple
3.
PLoS One ; 16(3): e0248996, 2021.
Article En | MEDLINE | ID: mdl-33780491

One of the central challenges for cancer therapy is the identification of factors in the tumor microenvironment that increase tumor progression and prevent immune surveillance. One such element associated with breast cancer is stromal fibrosis, a histopathologic criterion for invasive cancer and poor survival. Fibrosis is caused by inflammatory factors and remodeling of the extracellular matrix that elicit an immune tolerant microenvironment. To address the role of fibrosis in tumorigenesis, we developed NeuT/ATTAC transgenic mice expressing a constitutively active NeuT/erbB2 transgene, and an inducible, fat-directed caspase-8 fusion protein, which upon activation results in selective and partial ablation of mammary fat and its replacement with fibrotic tissue. Induction of fibrosis in NeuT/ATTAC mice led to more rapid tumor development and an inflammatory and fibrotic stromal environment. In an effort to explore therapeutic options that could reduce fibrosis and immune tolerance, mice were treated with the oxysterol liver X receptor (LXR) pan agonist, N,N-dimethyl-3-ß-hydroxy-cholenamide (DMHCA), an agent known to reduce fibrosis in non-malignant diseases. DMHCA reduced tumor progression, tumor multiplicity and fibrosis, and improved immune surveillance by reducing infiltrating myeloid-derived suppressor cells and increasing CD4 and CD8 effector T cells. These effects were associated with downregulation of an LXR-dependent gene network related to reduced breast cancer survival that included Spp1, S100a9, Anxa1, Mfge8 and Cd14. These findings suggest that the use of DMHCA may be a potentially effective approach to reduce desmoplasia and immune tolerance and increase the efficacy of cancer therapy.


Carcinogenesis/immunology , Carcinogenesis/pathology , Liver X Receptors/agonists , Receptor, ErbB-2/metabolism , Alarmins/genetics , Alarmins/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Carcinogenesis/drug effects , Cholic Acids/pharmacology , Disease Progression , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Fibrosis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver X Receptors/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Proteins/metabolism
4.
Adv Healthc Mater ; 9(15): e1900924, 2020 08.
Article En | MEDLINE | ID: mdl-31846231

Cancer metastases are a challenge for cancer treatment due to their organ specificity and pathophysiological complexity. Engineering 3D in vitro models capable of replicating native cancer dissemination can significantly improve the understanding of cancer biology and can help to guide the development of more effective treatments. In order to better mimic the behavior of native cancer, a triculture metastatic model is created using a stereolithography printing technique with optimized inks for investigating the invasion of breast cancer (BrCa) cells into vascularized bone tissue. The printed system allows to study transendothelial migration and the colony-forming behavior of metastatic BrCa cells. The key steps of BrCa cell progression including expansion, migration, and colonization are continuously monitored and the interactions between cancer cells, vascular cells, and bone cells are systematically investigated. The study results demonstrate that the 3D printed tissue construct by incorporating multiple cells and various favorable ink matrices provides a suitable model to study the interaction between these cells in a complex vascular microenvironment. As such, the 3D printed tricultured model may serve as a valuable tool for studying metastatic breast cancer progression in bone.


Breast Neoplasms , Bone and Bones , Humans , Ink , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tumor Microenvironment
6.
Sci Rep ; 8(1): 5717, 2018 04 09.
Article En | MEDLINE | ID: mdl-29632317

Plac1 is an X-linked trophoblast gene expressed at high levels in the placenta, but not in adult somatic tissues other than the testis. Plac1 however is re-expressed in several solid tumors and in most human cancer cell lines. To explore the role of Plac1 in cancer progression, Plac1 was reduced by RNA interference in EO771 mammary carcinoma cells. EO771 "knockdown" (KD) resulted in 50% reduction in proliferation in vitro and impaired tumor growth in syngeneic mice; however, tumor growth in SCID mice was equivalent to tumor cells expressing a non-silencing control RNA, suggesting that Plac1 regulated adaptive immunity. Gene expression profiling of Plac1 KD cells indicated reduction in several inflammatory and immune factors, including Cxcl1, Ccl5, Ly6a/Sca-1, Ly6c and Lif. Treatment of mice engrafted with wild-type EO771 cells with a Cxcr2 antagonist impaired tumor growth, reduced myeloid-derived suppressor cells and regulatory T cells, while increasing macrophages, dendritic cells, NK cells and the penetration of CD8+ T cells into the tumor bed. Cxcl1 KD phenocopied the effects of Plac1 KD on tumor growth, and overexpression of Cxcl1 partially rescued Plac1 KD cells. These results reveal that Plac1 modulates a tolerogenic tumor microenvironment in part by modulating the chemokine axis.


Breast Neoplasms/immunology , Breast Neoplasms/pathology , Chemokines/genetics , Pregnancy Proteins/genetics , RNA, Small Interfering/pharmacology , Adaptive Immunity , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mice, SCID , Pregnancy Proteins/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Transplantation, Isogeneic , Tumor Microenvironment
7.
Oncotarget ; 9(8): 8042-8053, 2018 Jan 30.
Article En | MEDLINE | ID: mdl-29487713

One of the central challenges in cancer prevention is the identification of factors in the tumor microenvironment (TME) that increase susceptibility to tumorigenesis. One such factor is stromal fibrosis, a histopathologic negative prognostic criterion for invasive breast cancer. Since the stromal composition of the breast is largely adipose and fibroblast tissue, it is important to understand how alterations in these tissues affect cancer progression. To address this question, a novel transgenic animal model was developed by crossing MMTV-NeuT mice containing a constitutively active ErbB2 gene into the FAT-ATTAC (fat apoptosis through targeted activation of caspase 8) background, which expresses an inducible caspase 8 fusion protein targeted to mammary adipose tissue. Upon caspase 8 activation, lipoatrophy of the mammary gland results in stromal fibrosis and acceleration of mammary tumor development with an increase in tumor multiplicity. Fibrosis was accompanied by an increase in collagen deposition, α-smooth muscle actin and CD31 expression in the tumor stroma as well as an increase in PD-L1-positive tumor cells, and infiltration by regulatory T cells, myeloid-derived suppressor cells and tumor-associated macrophages. Gene expression and signal transduction profiling indicated upregulation of pathways associated with cytokine signaling, inflammation and proliferation. This model should be useful for evaluating new therapies that target desmoplasia in the TME associated with invasive cancer.

8.
PLoS One ; 13(2): e0192106, 2018.
Article En | MEDLINE | ID: mdl-29432428

Placental-specific protein 1 (PLAC1) is an X-linked trophoblast gene that is re-expressed in several malignancies, including breast cancer, and is therefore a potential biomarker to follow disease onset and progression. Sera from 117 preoperative/pretreatment breast cancer patients and 51 control subjects, including those with fibrocystic disease, were analyzed for the presence of PLAC1 protein as well as its expression by IHC in tumor biopsies in a subset of subjects. Serum PLAC1 levels exceeded the mean plus one standard deviation (mean+SD) of the level in control subjects in 67% of subjects with ductal carcinoma in situ (DCIS), 67% with HER2+ tumors, 73% with triple-negative cancer and 73% with ER+/PR+ tumors. Greater sensitivity was achieved using the mean+2 SD of control PLAC1 serum values, where the false positive rate was 3% and was exceeded by 38%, 40%, 60% and 43% of subjects with DCIS, HER2+, TNBC and ER+/PR+/HER2- tumors. PLAC1 was detected in 97% of tumor biopsies, but did not correlate quantitatively with serum levels. There was no significant correlation of serum PLAC1 levels with race, age at diagnosis, body mass index (BMI) or the presence of metastatic disease. It remains to be determined whether PLAC1 serum levels can serve as a diagnostic biomarker for the presence or recurrence of disease post-surgery and/or therapy.


Breast Neoplasms/blood , Pregnancy Proteins/blood , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Middle Aged , Young Adult
9.
Oncotarget ; 9(102): 37808, 2018 12 28.
Article En | MEDLINE | ID: mdl-30701035

[This corrects the article DOI: 10.18632/oncotarget.24233.].

10.
Oncotarget ; 8(31): 50337-50341, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28881566

THE DILEMMA: Estrogen receptora-negative (ER-) breast cancer lacks a specific critical target to control tumor progression. THE OBJECTIVE: To identify mechanisms that enable increased expression of the ER+ lineage in an otherwise ER- breast cancer. PREFACE: The nuclear receptor superfamily members PPARγ and PPARδ regulate gene expression associated with a multitude of pathways, including intermediary metabolism, angiogenesis, proliferation and inflammation (see reviews [1-3]). Recent developments using transgenic and knockout mice, as well as pharmacologic intervention with PPARγ and PPARδ agonists, have revealed a previously unknown relationship between PPARγ suppression and PPARδ activation that leads to the appearance of ER+ tumors, enabling a synthetic lethality approach by anti-ER therapy. The ability to selectively affect the ER+ lineage by modulating PPARγ and PPARδ activity represents a new clinical paradigm and opportunity to treat ER- cancer with PPARγ and PPARδ modulating agents, ultimately rendering them more responsive to adjuvant therapy.

11.
ACS Appl Mater Interfaces ; 8(44): 30017-30026, 2016 Nov 09.
Article En | MEDLINE | ID: mdl-27766838

Metastasis is one of the deadliest consequences of breast cancer, with bone being one of the primary sites of occurrence. Insufficient 3D biomimetic models currently exist to replicate this process in vitro. In this study, we developed a biomimetic bone matrix using 3D bioprinting technology to investigate the interaction between breast cancer (BrCa) cells and bone stromal cells (fetal osteoblasts and human bone marrow mesenchymal stem cells (MSCs)). A tabletop stereolithography 3D bioprinter was employed to fabricate a series of bone matrices consisting of osteoblasts or MSCs encapsulated in gelatin methacrylate (GelMA) hydrogel with nanocrystalline hydroxyapatite (nHA). When BrCa cells were introduced into the stromal cell-laden bioprinted matrices, we found that the growth of BrCa cells was enhanced by the presence of osteoblasts or MSCs, whereas the proliferation of the osteoblasts or MSCs was inhibited by the BrCa cells. The BrCa cells co-cultured with MSCs or osteoblasts presented increased vascular endothelial growth factor (VEGF) secretion in comparison to that of monocultured BrCa cells. Additionally, the alkaline phosphatase activity of MSCs or osteoblasts was reduced after BrCa cell co-culture. These results demonstrate that the 3D bioprinted matrix, with BrCa cells and bone stromal cells, provides a suitable model with which to study the interactive effects of cells in the context of an artificial bone microenvironment and thus may serve as a valuable tool for the investigation of postmetastatic breast cancer progression in bone.


Bioprinting , Bone Matrix , Bone and Bones , Breast Neoplasms , Cell Differentiation , Humans , Mesenchymal Stem Cells , Neoplasm Metastasis , Vascular Endothelial Growth Factor A
12.
Nanotechnology ; 27(31): 315103, 2016 08 05.
Article En | MEDLINE | ID: mdl-27346678

Bone metastasis is one of the most prevalent complications of late-stage breast cancer, in which the native bone matrix components, including osteoblasts, are intimately involved in tumor progression. The development of a successful in vitro model would greatly facilitate understanding the underlying mechanism of breast cancer bone invasion as well as provide a tool for effective discovery of novel therapeutic strategies. In the current study, we fabricated a series of in vitro bone matrices composed of a polyethylene glycol hydrogel and nanocrystalline hydroxyapatite of varying concentrations to mimic the native bone microenvironment for the investigation of breast cancer bone metastasis. A stereolithography-based three-dimensional (3D) printer was used to fabricate the bone matrices with precisely controlled architecture. The interaction between breast cancer cells and osteoblasts was investigated in the optimized bone matrix. Using a Transwell® system to separate the two cell lines, breast cancer cells inhibited osteoblast proliferation, while osteoblasts stimulated breast cancer cell growth, whereas, both cell lines increased IL-8 secretion. Breast cancer cells co-cultured with osteoblasts within the 3D bone matrix formed multi-cellular spheroids in comparison to two-dimensional monolayers. These findings validate the use of our 3D printed bone matrices as an in vitro metastasis model, and highlights their potential for investigating breast cancer bone metastasis.

13.
PPAR Res ; 2016: 3082340, 2016.
Article En | MEDLINE | ID: mdl-28077942

PPARδ is a ligand-activated nuclear receptor that regulates the transcription of genes associated with proliferation, metabolism, inflammation, and immunity. Within this transcription factor family, PPARδ is unique in that it initiates oncogenesis in a metabolic and tissue-specific context, especially in mammary epithelium, and can regulate autoimmunity in some tissues. This review discusses its role in these processes and how it ultimately impacts breast cancer.

14.
Nanomedicine ; 12(1): 69-79, 2016 Jan.
Article En | MEDLINE | ID: mdl-26472048

Bone is one of the most common metastatic sites of breast cancer, but the underlying mechanisms remain unclear, in part due to an absence of advanced platforms for cancer culture and study that mimic the bone microenvironment. In the present study, we integrated a novel stereolithography-based 3D printer and a unique 3D printed nano-ink consisting of hydroxyapatite nanoparticles suspended in hydrogel to create a biomimetic bone-specific environment for evaluating breast cancer bone invasion. Breast cancer cells cultured in a geometrically optimized matrix exhibited spheroid morphology and migratory characteristics. Co-culture of tumor cells with bone marrow mesenchymal stem cells increased the formation of spheroid clusters. The 3D matrix also allowed for higher drug resistance of breast cancer cells than 2D culture. These results validate that our 3D bone matrix can mimic tumor bone microenvironments, suggesting that it can serve as a tool for studying metastasis and assessing drug sensitivity. From the Clinical Editor: Cancer remains a major cause of mortality for patients in the clinical setting. For breast cancer, bone is one of the most common metastatic sites. In this intriguing article, the authors developed a bone-like environment using 3D printing technology to investigate the underlying biology of bone metastasis. Their results would also allow a new model for other researchers who work on cancer to use.


Batch Cell Culture Techniques/methods , Bone Matrix/chemistry , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Nanocomposites/chemistry , Biomimetic Materials/chemical synthesis , Cell Line, Tumor , Humans , Materials Testing , Nanocomposites/ultrastructure , Neoplasm Invasiveness , Particle Size , Printing, Three-Dimensional , Tumor Microenvironment
15.
Antivir Chem Chemother ; 24(1): 19-27, 2015 Feb.
Article En | MEDLINE | ID: mdl-26149263

BACKGROUND: Current approved nucleoside therapies for genital herpes simplex virus (HSV) infections are effective but improved therapies are needed for treatment of both acute and recurrent diseases. METHODS: The effects of N-methanocarbathymidine were evaluated and compared to acyclovir using guinea pig models of acute and recurrent infection. For acute disease following intravaginal inoculation of 10(6 )pfu HSV-2 (MS strain), animals were treated intraperitoneally beginning 24 h post-infection, and the effects on disease severity, vaginal virus replication, subsequent recurrences, and latent virus loads were evaluated. For evaluation of recurrent infection, animals were treated for 21 days beginning 14 days after infection and disease recurrence and recurrent shedding were evaluated. RESULTS: Treatment of the acute disease with N-methanocarbathymidine significantly reduced the severity of acute disease and decreased acute vaginal virus shedding more effectively than acyclovir. Significantly, none of the animals developed visible disease in the high-dose N-methanocarbathymidine group and this was the only group in which the number of days with recurrent virus shedding was reduced. Treatment of recurrent disease was equivalent to acyclovir when acyclovir was continuously supplied in the drinking water. CONCLUSION: N-methanocarbathymidine was effective as therapy for acute and recurrent genital HSV-2 disease in the guinea pig models.


Antiviral Agents/pharmacology , Herpes Genitalis/drug therapy , Herpes Genitalis/virology , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/physiology , Thymidine/analogs & derivatives , Virus Shedding/drug effects , Acute Disease , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Disease Models, Animal , Female , Guinea Pigs , Recurrence , Thymidine/pharmacokinetics , Thymidine/pharmacology , Thymidine/therapeutic use
16.
PLoS One ; 10(2): e0116755, 2015.
Article En | MEDLINE | ID: mdl-25714835

Co-culture of human primary epithelial cells with irradiated 3T3 fibroblast feeder cells (J2 cells) and the Rho kinase inhibitor Y-27632 (Y) allows for the unrestricted growth of cells of epithelial origin by the process termed conditional reprogramming. To better understand the nature of the signaling processes associated with conditionally reprogrammed cells, the effect of the two critical components of the co-culture conditions, J2 cells and Y, on the growth of human foreskin keratinocytes (HFKs) was evaluated by gene expression profiling, reverse-phase protein arrays and siRNA screening. J2 cells and Y acted cooperatively to down-regulate differentiation, and upregulate proliferation and cell adhesion, including increased pT308Akt and pERK, and reduced TGF-ß pathway signaling. These findings establish a mechanistic basis for the unlimited growth potential of human epithelial cells that will be invaluable to assess the effect of genetic changes in pathologic tissues and their response to therapeutic agents.


Cellular Reprogramming , Keratinocytes/cytology , Keratinocytes/metabolism , Amides/pharmacology , Animals , Cell Adhesion , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Cells, Cultured , Cluster Analysis , Coculture Techniques , Feeder Cells , Gene Expression Profiling , Gene Expression Regulation , Humans , Keratinocytes/drug effects , Protein Processing, Post-Translational , Pyridines/pharmacology , RNA, Small Interfering
17.
Cancer Res ; 73(14): 4349-61, 2013 Jul 15.
Article En | MEDLINE | ID: mdl-23811944

The peroxisome proliferator-activated receptor-δ (PPARδ) regulates a multitude of physiological processes associated with glucose and lipid metabolism, inflammation, and proliferation. One or more of these processes are potential risk factors for the ability of PPARδ agonists to promote tumorigenesis in the mammary gland. In this study, we describe a new transgenic mouse model in which activation of PPARδ in the mammary epithelium by endogenous or synthetic ligands resulted in progressive histopathologic changes that culminated in the appearance of estrogen receptor- and progesterone receptor-positive and ErbB2-negative infiltrating ductal carcinomas. Multiparous mice presented with mammary carcinomas after a latency of 12 months, and administration of the PPARδ ligand GW501516 reduced tumor latency to 5 months. Histopathologic changes occurred concurrently with an increase in an inflammatory, invasive, metabolic, and proliferative gene signature, including expression of the trophoblast gene, Plac1, beginning 1 week after GW501516 treatment, and remained elevated throughout tumorigenesis. The appearance of malignant changes correlated with a pronounced increase in phosphatidylcholine and lysophosphatidic acid metabolites, which coincided with activation of Akt and mTOR signaling that were attenuated by treatment with the mTOR inhibitor everolimus. Our findings are the first to show a direct role of PPARδ in the pathogenesis of mammary tumorigenesis, and suggest a rationale for therapeutic approaches to prevent and treat this disease.


Carcinogenesis/metabolism , Inflammatory Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , PPAR delta/metabolism , Receptors, Estrogen/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Ductal/genetics , Carcinoma, Ductal/metabolism , Epithelium/metabolism , Female , Gene Expression , Genes, erbB-2 , Inflammation/genetics , Inflammation/metabolism , Inflammatory Breast Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Metabolomics/methods , Mice , Mice, Transgenic , PPAR delta/genetics , Phenotype , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , TOR Serine-Threonine Kinases/genetics , Thiazoles/pharmacology
18.
Oncotarget ; 4(5): 739-50, 2013 May.
Article En | MEDLINE | ID: mdl-23715514

Lung cancer remains one of the leading causes of cancer-related deaths worldwide with a 5-year survival rate of less than 20%. One approach to improving survival is the identification of biomarkers to detect early stage disease. In this study, we investigated the potential of the stem cell and progenitor cell marker, Musashi1 (Msi1), as a diagnostic marker and potential therapeutic target for lung cancer. Functional studies in A549 bronchioalveolar carcinoma and NCI-H520 squamous cell carcinoma cells revealed that Msi1 was enriched in spheroid cultures of tumor cells and in the CD133+ cell population. Downregulation of Msi1 by lentivirus-mediated expression of an Msi1 shRNA reduced spheroid colony proliferation. Growth inhibition was associated with reduced nuclear localization of ß-catenin and inhibition of the processing of intracellular Notch. In primary lung cancer, Msi1 protein expression was elevated in 86% of 202 tissue microarray specimens, and Msi1 mRNA was increased in 80% of 118 bronchoscopic biopsies, including metastatic disease, but was rarely detected in adjacent normal lung tissue and in non-malignant diseased tissue. Msi1 was expressed in a diffuse pattern in most tumor subtypes, except in squamous cell carcinomas, where it appeared in a focal pattern in 50% of specimens. Thus, Msi1 is a sensitive and specific diagnostic marker for all lung cancer subtypes.


Carcinoma, Non-Small-Cell Lung/diagnosis , Lung Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Small Cell Lung Carcinoma/diagnosis , Spheroids, Cellular/metabolism , AC133 Antigen , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Glycoproteins/metabolism , Humans , Lung Neoplasms/diagnosis , Male , Middle Aged , Neoplasms, Squamous Cell/metabolism , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Peptides/metabolism , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Receptors, Notch/metabolism , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
19.
Oncotarget ; 3(3): 345-56, 2012 Mar.
Article En | MEDLINE | ID: mdl-22538444

The peroxisome proliferator-activated receptorγ (PPARγ) is a key regulator of metabolism, proliferation, inflammation and differentiation, and upregulates tumor suppressor genes, such as PTEN, BRCA1 and PPARγ itself. Examination of mammary carcinogenesis in transgenic mice expressing the dominant-negative Pax8PPARγ fusion protein revealed that tumors were estrogen receptorα (ER)-positive and sensitive to the ER antagonist, fulvestrant. Here we evaluated whether administration of an irreversible PPARγ inhibitor in vivo could similarly induce ER expression in otherwise ER-negative mammary tumors following induction of carcinogenesis, and sensitize them to the antitumor effects of fulvestrant. In addition, we wished to determine whether the effect of GW9662 was associated with a PPAR-selective gene expression profile. Mammary carcinogenesis was induced in wild-type FVB mice by treatment with medroxyprogesterone and dimethylbenz(a)anthracene (DMBA) that were subsequently maintained on a diet supplemented with 0.1% GW9662, and tumorigenesis and gene expression profiling of the resulting tumors were determined. Administration of GW9962 resulted in ER+ tumors that were highly sensitive to fulvestrant. Tumors from GW9662-treated animals exhibited reduced expression of a metabolic gene profile indicative of PPARγ inhibition, including PPARγ itself. Additionally, GW9662 upregulated the expression of several genes associated with the transcription, processing, splicing and translation of RNA. This study is the first to show that an irreversible PPARγ inhibitor can mimic a dominant-negative PPARγ transgene to elicit the development of ER-responsive tumors. These findings suggest that it may be possible to pharmacologically influence the responsiveness of tumors to anti-estrogen therapy.


Anilides/administration & dosage , Carcinoma/prevention & control , Estradiol/analogs & derivatives , Mammary Neoplasms, Experimental/prevention & control , PPAR gamma/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene , Anilides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacology , Carcinogens , Carcinoma/chemically induced , Carcinoma/genetics , Carcinoma/mortality , Chemoprevention/methods , Drug Synergism , Estradiol/administration & dosage , Estradiol/pharmacology , Estrogen Antagonists/administration & dosage , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/mortality , Mice , Microarray Analysis , Molecular Targeted Therapy
20.
Cancer Prev Res (Phila) ; 5(5): 754-64, 2012 May.
Article En | MEDLINE | ID: mdl-22427346

Progesterone receptor (PR) antagonists are potent antitumor agents in carcinogen and progestin-dependent mammary tumorigenesis models through both PR- and non-PR-mediated mechanisms. The PR antagonist mifepristone/RU486 has been used primarily as an abortifacient possessing high affinity for both the PR and glucocorticoid receptors (GR). To determine whether mifepristone would be effective as a chemopreventive agent, we assessed its effect on progestin/7,12-dimethylbenz(a)anthracene (DMBA)-induced mammary carcinogenesis in wild-type (WT) and estrogen receptor-α-positive (ER(+)) transgenic mice expressing the dominant-negative Pax8PPARγ (Pax8) fusion protein. Mifepristone administered at a dose of 2.5 mg significantly delayed mammary tumorigenesis in WT, but not in Pax8 mice, whereas, a three-fold higher dose almost completely blocked tumorigenesis in both WT and Pax8 mice. The sensitivity of WT mice to 2.5 mg mifepristone correlated with an expression profile of 79 genes in tumors, 52 of which exhibited the opposite response in Pax8 mice, and corresponded primarily to the downregulation of genes associated with metabolism, inflammation, and invasion. These results suggest that the chemopreventive activity of mifepristone in WT mice correlates with a specific gene expression signature that is associated with multiple nuclear receptor signaling pathways.


Carcinoma/genetics , Carcinoma/prevention & control , Inflammation/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/prevention & control , Mifepristone/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Carcinogens , Carcinoma/chemically induced , Carcinoma/pathology , Chemoprevention/methods , Drug Evaluation, Preclinical , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hormone Antagonists/pharmacology , Hormone Antagonists/therapeutic use , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Microarray Analysis , Mifepristone/pharmacology , Neoplasm Invasiveness/genetics , PAX8 Transcription Factor , Paired Box Transcription Factors/genetics
...