Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Adv Healthc Mater ; 13(10): e2303593, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38215360

ABSTRACT

Current nucleic acid delivery methods have not achieved efficient, non-toxic delivery of miRNAs with tumor-specific selectivity. In this study, a new delivery system based on light-inducible gold-silver-gold, core-shell-shell (CSS) nanoparticles is presented. This system delivers small nucleic acid therapeutics with precise spatiotemporal control, demonstrating the potential for achieving tumor-specific selectivity and efficient delivery of miRNA mimics. The light-inducible particles leverage the photothermal heating of metal nanoparticles due to the local surface plasmonic resonance for controlled chemical cleavage and release of the miRNA mimic payload. The CSS morphology and composition result in a plasmonic resonance within the near-infrared (NIR) region of the light spectrum. Through this method, exogenous miR-34a-5p mimics are effectively delivered to human squamous cell carcinoma TE10 cells, leading to apoptosis induction without adverse effects on untransformed keratinocytes in vitro. The CSS nanoparticle delivery system is tested in vivo in Foxn1nu athymic nude mice with bilateral human esophageal TE10 cancer cells xenografts. These experiments reveal that this CSS nanoparticle conjugates, when systemically administered, followed by 850 nm light emitting diode irradiation at the tumor site, 6 h post-injection, produce a significant and sustained reduction in tumor volume, exceeding 87% in less than 72 h.


Subject(s)
Esophageal Neoplasms , Metal Nanoparticles , MicroRNAs , Nanoparticles , Animals , Mice , Humans , Mice, Nude , Nanoparticles/chemistry , MicroRNAs/genetics , Metal Nanoparticles/chemistry , Esophageal Neoplasms/drug therapy , Gold/chemistry , Cell Line, Tumor
2.
Dev Biol ; 492: 59-70, 2022 12.
Article in English | MEDLINE | ID: mdl-36179879

ABSTRACT

The IRE1α-XBP1s signaling branch of the unfolded protein response is a well-characterized survival pathway that allows cells to adapt to and resolve endoplasmic reticulum stress. Recent data has broadened our understanding of IRE1α-XBP1s signaling beyond a stress response and revealed a physiological mechanism required for the differentiation and maturation of a wide variety of cell types. Here we provide evidence that the IRE1α-XBP1s signaling pathway is required for the proliferation and maturation of basal keratinocytes in the mouse tongue and esophageal epithelium. Mice with conditional targeted deletion of either Ire1α or Xbp1 in keratin 14 expressing basal keratinocytes displayed severe thinning of the lingual and esophageal mucosa that rendered them unable to eat. In IRE1α null epithelium harvested at an earlier timepoint, genes regulating cell proliferation, cell-cell adhesion, and keratinization were significantly downregulated; indirect immunofluorescence revealed fewer proliferating basal keratinocytes, downregulation of E-cadherin, and thinning of the loricrin-positive granular and cornified layers. The number of Tp63-positive basal keratinocytes was reduced in the absence of IRE1α, and expression of the Wnt pathway transcription factor LEF1, which is required for the proliferation of lingual transit amplifying cells, was also significantly downregulated at the transcript and protein level. Together these results reveal an essential role for IRE1α-XBP1s in the maintenance of the stratified squamous epithelial tissue of the tongue and esophagus.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Mice , Animals , Endoribonucleases/genetics , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/genetics , Unfolded Protein Response/genetics , Endoplasmic Reticulum Stress/genetics , Esophagus , Tongue/metabolism
3.
PLoS One ; 16(11): e0259998, 2021.
Article in English | MEDLINE | ID: mdl-34784403

ABSTRACT

One of the major risk factors for head and neck squamous cell carcinoma (HNSCC) is tobacco smoke exposure, but the mechanisms that can account for disease development remain to be fully defined. Utilizing our HNSCC mouse model, we analyzed oral squamous cell carcinomas (OSCC) induced by the active metabolite of a common smoke constituent, dibenzo[a,l]pyrene diol-epoxide (DBPDE). Analyzing protein expression by either immunofluorescence or immunohistochemistry, we identified biologic processes that are dysregulated in premalignant and invasive cancer lesions induced by DBPDE. Interestingly, p120ctn expression is downregulated in both stages of the disease. In addition to decreased p120ctn expression, there was also increased proliferation (as measured by Ki67), inflammation (as measured by NFkB (p65) expression), neovascularization (as measured by CD31) and recruitment of Ly6G-positive immune cells as well as strong EGFR expression. We also examined the effect of the chemopreventive agent black raspberry (BRB) on p120ctn and EGFR protein expression in DBPDE treated mice. p120ctn, but not EGFR, protein expression increased in mice treated with BRB. Our results suggest that modulation of p120ctn may, in part, account for the mechanism by which BRB inhibits DBPDE induced OSCC in mice.


Subject(s)
Catenins/metabolism , Epoxy Compounds/adverse effects , Mouth Neoplasms/diet therapy , Phytochemicals/administration & dosage , Rubus/chemistry , Squamous Cell Carcinoma of Head and Neck/diet therapy , Animals , Cell Line , Down-Regulation/drug effects , Epoxy Compounds/chemistry , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mouth Neoplasms/chemically induced , Mouth Neoplasms/metabolism , Phytochemicals/pharmacology , Pyrenes/chemistry , Squamous Cell Carcinoma of Head and Neck/chemically induced , Squamous Cell Carcinoma of Head and Neck/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays , Delta Catenin
4.
Sci Rep ; 11(1): 19022, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34561473

ABSTRACT

Phosphatidylinositol glycan anchor biosynthesis class N (PIGN) has been linked to the suppression of chromosomal instability. The spindle assembly checkpoint complex is responsible for proper chromosome segregation during mitosis to prevent chromosomal instability. In this study, the novel role of PIGN as a regulator of the spindle assembly checkpoint was unveiled in leukemic patient cells and cell lines. Transient downregulation or ablation of PIGN resulted in impaired mitotic checkpoint activation due to the dysregulated expression of spindle assembly checkpoint-related proteins including MAD1, MAD2, BUBR1, and MPS1. Moreover, ectopic overexpression of PIGN restored the expression of MAD2. PIGN regulated the spindle assembly checkpoint by forming a complex with the spindle assembly checkpoint proteins MAD1, MAD2, and the mitotic kinase MPS1. Thus, PIGN could play a vital role in the spindle assembly checkpoint to suppress chromosomal instability associated with leukemic transformation and progression.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Leukemia/pathology , Phosphotransferases/physiology , Spindle Apparatus/metabolism , Cell Cycle Proteins/genetics , Cell Line , Disease Progression , Gene Expression , HL-60 Cells , Humans , K562 Cells , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism
5.
Chem Biol Interact ; 333: 109321, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33186600

ABSTRACT

HPV infections in the oral cavity that progress to cancer are on the increase in the USA. Model systems to study co-factors for progression of these infections are lacking as HPVs are species-restricted and cannot grow in preclinical animal models. We have recently developed a mouse papillomavirus (MmuPV1) oral mucosal infection model that provides opportunities to test, for the first time, the hypothesis that tobacco carcinogens are co-factors that can impact the progression of oral papillomas to squamous cell carcinoma (SCC). Four cohorts of mice per sex were included: (1) infected with MmuPV1 and treated orally with DMSO-saline; (2) infected with MmuPV1 and treated orally with the tobacco carcinogen, dibenzo[def,p]chrysene (DBP); (3) uninfected and treated orally with DMSO-saline, and (4) uninfected and treated orally with DBP. Oral swabs were collected monthly for subsequent assessment of viral load. Oral tissues were collected for in situ viral DNA/RNA detection, viral protein staining, and pathological assessment for hyperplasia, papillomas and SCC at study termination. We observed increased rates of SCC in oral tissue infected with MmuPV1 and treated with DBP when compared to mice treated with DBP or virus individually, each of which showed minimal disease. Virally-infected epithelium showed strong levels of viral DNA/RNA and viral protein E4/L1 staining. In contrast, areas of SCC showed reduced viral DNA staining indicative of lower viral copy per nucleus but strong RNA signals. Several host markers (p120 ctn, p53, S100A9) were also examined in the mouse oral tissues; of particular significance, p120 ctn discriminated normal un-infected epithelium from SCC or papilloma epithelium. In summary, we have confirmed that our infection model is an excellent platform to assess the impact of co-factors including tobacco carcinogens for oral PV cancerous progression. Our findings can assist in the design of novel prevention/treatment strategies for HPV positive vs. HPV negative disease.


Subject(s)
Chrysenes/toxicity , Disease Progression , Environmental Pollutants/toxicity , Mouth Neoplasms/pathology , Nicotiana/adverse effects , Papillomaviridae/physiology , Smoke/adverse effects , Animals , Carcinogenesis/drug effects , Female , Genome, Viral/genetics , Male , Mice , Mouth Neoplasms/virology , Papillomaviridae/genetics , Sex Characteristics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/virology
6.
Sci Rep ; 10(1): 18829, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33139779

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is among the most aggressive and fatal cancer types. ESCC classically progresses rapidly and frequently causes mortality in four out of five patients within two years of diagnosis. Yet, little is known about the mechanisms that make ESCC so aggressive. In a previous study we demonstrated that p120-catenin (p120ctn) and EGFR, two genes associated with poor prognosis in ESCC, work together to cause invasion. Specifically, inactivation of p120ctn combined with overexpression of EGFR induces a signaling cascade that leads to hyperactivation of NFkB and a resultant aggressive cell type. The purpose of this present study was to identify targets that are responsive to NFkB when p120ctn and EGFR are modified. Using human esophageal keratinocytes, we have identified Twist2 as an NFkB-responsive gene. Interestingly, we found that when NFkB is hyperactivated in cells with EGFR overexpression and p120ctn inactivation, Twist2 is significantly upregulated. Inhibition of NFkB activity results in nearly complete loss of Twist2 expression, suggesting that this potential EMT-inducing gene, is a responsive target of NFkB. There exists a paucity of research on Twist2 in any cancer type; as such, these findings are important in ESCC as well as in other cancer types.


Subject(s)
Catenins/metabolism , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Keratinocytes/metabolism , NF-kappa B/metabolism , Repressor Proteins/metabolism , Twist-Related Protein 1/metabolism , Catenins/genetics , Cell Line , Disease Progression , ErbB Receptors/genetics , ErbB Receptors/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression , Humans , NF-kappa B/genetics , Repressor Proteins/genetics , Twist-Related Protein 1/genetics , Delta Catenin
7.
PLoS One ; 15(10): e0241299, 2020.
Article in English | MEDLINE | ID: mdl-33112928

ABSTRACT

Epidermal growth factor receptor (EGFR) plays a vital role in cell division and survival signaling pathways. EGFR is activated in nearly every cancer type, and its high expression in tumors is correlated with poor patient outcome. Altogether, EGFR is a prime candidate as a therapeutic target. While targeted EGFR therapy is initially effective in 75% of patients, a majority of patients relapse within the first year due to poorly understood mechanisms of resistance. p120-catenin (p120ctn) has recently been implicated as a biomarker for EGFR therapy. In previous studies, we demonstrated that p120ctn is a tumor suppressor and its loss is capable of inducing cancer. Furthermore, p120ctn down-regulation synergizes with EGFR overexpression to cause a highly invasive cell phenotype. The purpose of this present study was to investigate whether p120ctn down-regulation induced EGFR therapeutic resistance. Using human esophageal keratinocytes, we have found that EGFR-targeting compounds are toxic to cells overexpressing EGFR. Interestingly, these therapies do not cause toxicity in cells with EGFR overexpression and decreased p120ctn expression. These data suggest that decreased p120ctn causes resistance to EGFR therapy. We believe these findings are of utmost importance, as there is an unmet need to discover mechanisms of EGFR resistance.


Subject(s)
Catenins/deficiency , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Molecular Targeted Therapy , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Catenins/metabolism , Cell Line, Tumor , Cetuximab/pharmacology , Down-Regulation/drug effects , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Esophagus/pathology , Gefitinib/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , NF-kappa B/metabolism , Protein Kinase Inhibitors/pharmacology , Treatment Outcome , Delta Catenin
8.
Cancer Prev Res (Phila) ; 13(8): 649-660, 2020 08.
Article in English | MEDLINE | ID: mdl-32434808

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) was the 7th most common malignancy worldwide in 2018 and despite therapeutic advances, the overall survival rate for oral squamous cell carcinoma (OSCC; ∼50%) has remained unchanged for decades. The most common types are OSCC and oropharyngeal squamous cell carcinoma (OPSCC, survival rate ∼85%). Tobacco smoking is a major risk factor of HNSCC. In the developed world, the incidence of OSCC is declining as a result of tobacco cessation programs. However, OPSCC, which is also linked to human papillomavirus (HPV) infection, is on the rise and now ranks as the most common HPV-related cancer. The current state of knowledge indicates that HPV-associated disease differs substantially from other types of HNSCC and distinct biological differences between HPV-positive and HPV-negative HNSCC have been identified. Although risk factors have been extensively discussed in the literature, there are multiple clinically relevant questions that remain unanswered and even unexplored. Moreover, existing approaches (e.g., tobacco cessation, vaccination, and chemoprevention) to manage and control this disease remain a challenge. Thus, in this review, we discuss potential future basic research that can assist in a better understanding of disease pathogenesis which may lead to novel and more effective preventive strategies for OSCC and OPSCC.


Subject(s)
Mouth Neoplasms/prevention & control , Oropharyngeal Neoplasms/prevention & control , Papillomavirus Infections/prevention & control , Squamous Cell Carcinoma of Head and Neck/prevention & control , Alphapapillomavirus/immunology , Animals , Disease Models, Animal , Humans , Incidence , Mass Vaccination/organization & administration , Mice , Microbiota/immunology , Mouth/microbiology , Mouth/pathology , Mouth Neoplasms/epidemiology , Mouth Neoplasms/pathology , Mouth Neoplasms/virology , Oropharyngeal Neoplasms/epidemiology , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/epidemiology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Risk Factors , Squamous Cell Carcinoma of Head and Neck/epidemiology , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/virology , Tobacco Smoking/epidemiology , Tobacco Use Cessation
9.
Exp Dermatol ; 28(7): 867-871, 2019 07.
Article in English | MEDLINE | ID: mdl-31107992

ABSTRACT

Hidradenitis suppurativa (HS) is a chronic, inflammatory skin disease affecting the pilosebaceous units in the axilla, groin and buttocks. While the pathogenesis of HS is not clear, mechanical stress exacerbates HS. In this study, we aimed to determine whether intracellular adhesive junctions may be aberrant in HS patient skin. Strikingly, we observed loss of E-cadherin and p120ctn protein expression, two key adherens junction proteins, in ~85% of HS severe skin lesions. Moreover, loss of protein expression was apparent in non-lesional skin from HS patients and the degree of loss positively correlated with HS Hurley Stage of disease. E-cadherin expression was unaltered in other inflammatory skin conditions including chronic wound epithelium, atopic dermatitis, and acne vulgaris compared with healthy skin suggesting that its loss may be uniquely relevant to HS pathogenesis. A complete loss of α-catenin, ß-catenin and ZO-1 was not observed; however, some cytoplasmic staining of the catenins was noted in HS epithelium. We also demonstrated diminished desmosome size in HS lesional skin. Overall, our data suggested that loss of adherens junction proteins and diminished desmosome size in HS skin contributes to the skin's inability to withstand mechanical stress and provides rationale as to why mechanical stress exacerbates HS symptoms.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Catenins/metabolism , Gene Expression Regulation , Hidradenitis Suppurativa/metabolism , Adherens Junctions , Dermatitis, Atopic/metabolism , Humans , Skin/metabolism , Stress, Mechanical , Zonula Occludens-1 Protein/metabolism , alpha Catenin/metabolism , beta Catenin/metabolism , Delta Catenin
10.
Oncotarget ; 9(13): 11180-11196, 2018 Feb 16.
Article in English | MEDLINE | ID: mdl-29541406

ABSTRACT

Four out of five patients diagnosed with esophageal squamous cell carcinoma (ESCC) will die within five years. This is primarily a result of the aggressive invasive potential of the disease. Our research is focused on the interplay between tumor suppressors and oncogenes in the invasive process. Specifically, EGFR and p120-catenin (p120ctn) are commonly dysregulated genes that are indicative of poor prognosis in ESCC. In a previous study we demonstrated that in our 3D organotypic culture model, only when EGFR overexpression is combined with p120ctn inactivation do the cells transform and invade - as opposed to either event alone. The purpose of this present study was to identify the components of the molecular pathways downstream of p120ctn and EGFR that lead to invasion. Using both human esophageal keratinocytes and human ESCC cells, we have identified NFkB as a central regulator of the invasive process downstream of p120ctn down-regulation and EGFR overexpression. Interestingly, we found that NFkB is hyperactivated in cells with EGFR overexpression and p120ctn inactivation than with either EGFR or p120ctn alone. Inhibition of this NFkB hyperactivation results in complete loss of invasion, suggesting that NFkB signaling is necessary for invasion in this aggressive cell type. Furthermore, we have identified RhoA and Rho-kinase as upstream regulators of NFkB in this process. We believe the cooperation of p120ctn down-regulation and EGFR overexpression is not only important in the aggressive mechanisms of ESCC but could be broadly applicable to many other cancer types in which p120ctn and EGFR are involved.

11.
Mol Cancer Res ; 15(10): 1398-1409, 2017 10.
Article in English | MEDLINE | ID: mdl-28637905

ABSTRACT

Despite recent improvements in treatment for head and neck squamous cell carcinoma (HNSCC), half of all patients with a regional or advanced disease will die within 5 years from diagnosis. Therefore, identification of mechanisms driving the aggressive behavior of HNSCC is of utmost importance. Because p120-catenin (CTNND1/P120CTN) downregulation and PIK3CA mutations are commonly found in HNSCC, the objective of this study was to identify their impact on fundamental processes of metastasis, specifically, migration and invasion. Furthermore, this study aimed to identify the key effector proteins regulated by P120CTN downregulation and PIK3CA mutations. Studies using oral keratinocytes demonstrated that P120CTN downregulation and PIK3CA mutations increased migration and invasion. In addition, P120CTN downregulation and PIK3CA mutations resulted in elevated matrix metallopeptidase 1 (MMP1) levels. Inhibition of MMP1 resulted in decreased invasion, suggesting that MMP1 plays a critical role in HNSCC invasion. Moreover, analysis of HNSCC patient specimens from The Cancer Genome Atlas confirmed these findings. Tumors with low P120CTN and PI3K pathway mutations have higher levels of MMP1 compared to tumors with high P120CTN and no PI3K pathway mutations. In conclusion, this study demonstrates that P120CTN downregulation and PIK3CA mutations promote MMP1-driven invasion, providing a potential novel target for limiting metastasis in HNSCC.Implications: Because of its role in invasion, MMP1 represents a novel, potential target for limiting metastasis in a subset of HNSCCs with P120CTN downregulation and PIK3CA mutations. Mol Cancer Res; 15(10); 1398-409. ©2017 AACR.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Catenins/metabolism , Class I Phosphatidylinositol 3-Kinases/genetics , Down-Regulation , Head and Neck Neoplasms/metabolism , Matrix Metalloproteinase 1/metabolism , Mutation , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Movement , Class I Phosphatidylinositol 3-Kinases/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Neoplasm Invasiveness , Squamous Cell Carcinoma of Head and Neck , Delta Catenin
12.
Diagn Pathol ; 11: 23, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26926447

ABSTRACT

BACKGROUND: Patients with non-dysplastic Barrett's esophagus (ND-BE) and low-grade dysplasia (LGD) are typically monitored by periodic endoscopic surveillance, while those with high-grade dysplasia (HGD) and esophageal adenocarcinoma (EAC) are usually treated by more aggressive interventions like endoscopic mucosal resection, ablation or surgery. Therefore, the accurate grading of dysplasia in Barrett's esophagus (BE) is essential for proper patient care. However, there is significant interobserver and intraobserver variability in the histologic grading of BE dysplasia. The objective of this study was to create an immunohistochemical (IHC) panel that facilitates the grading of BE dysplasia and can be used as an adjunct to histology in challenging cases. METHODS: 100 BE biopsies were re-graded for dysplasia independently by 3 subspecialized gastrointestinal pathologists. IHC staining for CDX2, p120ctn, c-Myc and Jagged1 proteins was then performed and assessed by two separate methods of semi-quantitative scoring. Scores were integrated using a principal component analysis (PCA) and receiver operating characteristic (ROC) curve. RESULTS: Principal component analysis demonstrated the ability of this panel of proteins to segregate ND-BE/LGD and HGD/EAC, as the expression of the four proteins is significantly altered between the two subsets. Analysis of the receiver operating characteristic curve showed that this panel has the potential to aid in the grading of dysplasia in these two subcategories with both high sensitivity and specificity. While not able to discriminate between ND-BE and LGD, this panel of four proteins may be used as an adjunct to help discriminate subsets of ND-BE/LGD from HGD/EAC. CONCLUSIONS: We propose that the maximum utility of this IHC panel of CDX2, p120ctn, c-Myc, and Jagged1 proteins would be to distinguish between LGD and HGD in histologically challenging cases, given the aggressive interventions still used for HGD in many institutions, and hence may aid in the optimal patient management. The results of this initial study are promising, though further validation is needed before this panel can be used clinically, including future randomized prospective studies with larger patient cohorts from diverse locations.


Subject(s)
Adenocarcinoma/chemistry , Barrett Esophagus/metabolism , Biomarkers, Tumor/analysis , Calcium-Binding Proteins/analysis , Catenins/analysis , Esophageal Neoplasms/chemistry , Esophagus/chemistry , Homeodomain Proteins/analysis , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/analysis , Membrane Proteins/analysis , Proto-Oncogene Proteins c-myc/analysis , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Area Under Curve , Barrett Esophagus/pathology , Biopsy , CDX2 Transcription Factor , Consensus , Diagnosis, Differential , Esophageal Neoplasms/pathology , Esophagus/pathology , Female , Humans , Jagged-1 Protein , Male , Middle Aged , Neoplasm Grading , Observer Variation , Predictive Value of Tests , Principal Component Analysis , ROC Curve , Reproducibility of Results , Serrate-Jagged Proteins , Severity of Illness Index , United States , Delta Catenin
13.
Article in English | MEDLINE | ID: mdl-26845434

ABSTRACT

Esophageal squamous cell carcinoma (ESCC), which is the most common subtype of esophageal cancers, is the sixth leading cause of cancer death worldwide with a five-year survival rate of 19%. Identification of efficient biomarkers for early detection and better understanding of the molecular mechanisms of ESCC may offer reduced mortality. However, proper biomarkers for clinical diagnosis and prognosis have not been defined yet. In the presented study, we employed a systematic and integrative 'omics' strategy to reconstruct networks of transcriptional regulation and protein-protein interaction to identify novel biomarkers, potential molecular targets, and mechanisms of transcriptional control in ESCC. Towards this end, we revealed 30 down-regulated and 21 up-regulated genes as ESCC specific biomarkers since these were differentially expressed between 91 ESCC tumor samples compared to normal tissues in five different datasets. We report the association of ACPP, C2orf54, DYNLT3, ENDOU, FMO2, and KANK1 (down-regulated genes) and COL10A1, FNDC3B, HOMER3, MARCKSL1, and RFC4 (up-regulated genes) to ESCC for the first time. Further, the ESCC driven molecular pathways were also constructed to elucidate the molecular mechanism of the disease; specifically several metabolic pathways were down-regulated while the signaling pathways were up-regulated. Additionally, reporter metabolites for ESCC were analyzed and metabolic dysfunction was ascertained in arachidonic acid metabolism and steroid hormone biosynthesis pathways. The multi-omics network strategy presented here may enable discovery of novel biomarkers and targets for personalized medicine in ESCC patients.

14.
Cancer Growth Metastasis ; 8(Suppl 1): 1-15, 2015.
Article in English | MEDLINE | ID: mdl-26380553

ABSTRACT

Mouse models of human cancer play a critical role in understanding the molecular and cellular mechanisms of tumorigenesis. Advances continue to be made in modeling human disease in a mouse, though the relevance of a mouse model often relies on how closely it is able to mimic the histologic, molecular, and physiologic characteristics of the respective human cancer. A classic use of a genetically engineered mouse in studying cancer is through the overexpression or deletion of a gene. However, the manipulation of a single gene often falls short of mimicking all the characteristics of the carcinoma in humans; thus a multiple gene approach is needed. Here we review genetic mouse models of cancers and their abilities to recapitulate human carcinoma with single versus combinatorial approaches with genes commonly involved in cancer.

15.
Am J Pathol ; 185(1): 240-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25529795

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy with a poor prognosis due to its highly invasive and metastatic potential. The molecular pathogenesis underlying the invasive mechanism of ESCC is not well known because of the lack of existing models to study this disease. p120-Catenin (p120ctn) and the epidermal growth factor receptor (EGFR) have each been implicated in several cancers, including ESCC. p120ctn is down-regulated in 60% of ESCC tumors, whereas EGFR is the most commonly overexpressed oncogene in ESCC. For these reasons, we investigated the cooperation between p120ctn and EGFR and its effect on ESCC invasion. We show that p120ctn down-regulation is commonly associated with EGFR overexpression. By using a three-dimensional culture system, we demonstrate that the inverse relationship between p120ctn and EGFR has biological implications. Specifically, p120ctn down-regulation coupled with EGFR overexpression in human esophageal keratinocytes (EPC1-PE) was required to promote invasion. Morphological comparison of EPC1-PE cells grown in three-dimensional culture and human ESCC revealed identical features, including significantly increased cellularity, nuclear grade, and proliferation. Molecular characteristics were measured by keratin expression patterns, which were nearly identical between EPC1-PE cells in three-dimensional culture and ESCC samples. Altogether, our analyses have demonstrated that p120ctn down-regulation and EGFR overexpression are able to mimic human ESCC in a relevant three-dimensional culture model.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Catenins/metabolism , ErbB Receptors/metabolism , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Antigens, CD , Cadherins/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Down-Regulation , Epithelium/metabolism , Epithelium/pathology , Esophageal Squamous Cell Carcinoma , Gene Expression Profiling , Humans , Immunohistochemistry , Keratins/metabolism , Neoplasm Invasiveness , Delta Catenin
16.
Cell Cycle ; 13(24): 3857-66, 2014.
Article in English | MEDLINE | ID: mdl-25558829

ABSTRACT

Barrett's esophagus (BE) is defined as an incomplete intestinal metaplasia characterized generally by the presence of columnar and goblet cells in the formerly stratified squamous epithelium of the esophagus. BE is known as a precursor for esophageal adenocarcinoma. Currently, the cell of origin for human BE has yet to be clearly identified. Therefore, we investigated the role of Notch signaling in the initiation of BE metaplasia. Affymetrix gene expression microarray revealed that BE samples express decreased levels of Notch receptors (NOTCH2 and NOTCH3) and one of the the ligands (JAG1). Furthermore, BE tissue microarray showed decreased expression of NOTCH1 and its downstream target HES1. Therefore, Notch signaling was inhibited in human esophageal epithelial cells by expression of dominant-negative-Mastermind-like (dnMAML), in concert with MYC and CDX1 overexpression. Cell transdifferentiation was then assessed by 3D organotypic culture and evaluation of BE-lineage specific gene expression. Notch inhibition promoted transdifferentiation of esophageal epithelial cells toward columnar-like cells as demonstrated by increased expression of columnar keratins (K8, K18, K19, K20) and glandular mucins (MUC2, MUC3B, MUC5B, MUC17) and decreased expression of squamous keratins (K5, K13, K14). In 3D culture, elongated cells were observed in the basal layer of the epithelium with Notch inhibition. Furthermore, we observed increased expression of KLF4, a potential driver of the changes observed by Notch inhibition. Interestingly, knockdown of KLF4 reversed the effects of Notch inhibition on BE-like metaplasia. Overall, Notch signaling inhibition promotes transdifferentiation of esophageal cells toward BE-like metaplasia in part via upregulation of KLF4. These results support a novel mechanism through which esophageal epithelial transdifferentiation promotes the evolution of BE.


Subject(s)
Esophagus/pathology , Kruppel-Like Transcription Factors/metabolism , Receptors, Notch/metabolism , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cell Culture Techniques , Cell Line , Cell Transdifferentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Esophagus/cytology , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Keratins/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Metaplasia , Mucins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Receptors, Notch/antagonists & inhibitors , Serrate-Jagged Proteins , Signal Transduction , Tissue Array Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Genes Dev ; 26(19): 2154-68, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23028142

ABSTRACT

Extrapituitary prolactin (Prl) is produced in humans and rodents; however, little is known about its in vivo regulation or physiological function. We now report that autocrine prolactin is required for terminal mammary epithelial differentiation during pregnancy and that its production is regulated by the Pten-PI3K-Akt pathway. Conditional activation of the PI3K-Akt pathway in the mammary glands of virgin mice by either Akt1 expression or Pten deletion rapidly induced terminal mammary epithelial differentiation accompanied by the synthesis of milk despite the absence of lobuloalveolar development. Surprisingly, we found that mammary differentiation was due to the PI3K-Akt-dependent synthesis and secretion of autocrine prolactin and downstream activation of the prolactin receptor (Prlr)-Jak-Stat5 pathway. Consistent with this, Akt-induced mammary differentiation was abrogated in Prl(-/-), Prlr(-/-), and Stat5(-/-) mice. Furthermore, cells treated with conditioned medium from mammary glands in which Akt had been activated underwent rapid Stat5 phosphorylation in a manner that was blocked by inhibition of Jak2, treatment with an anti-Prl antibody, or deletion of the prolactin gene. Demonstrating a physiological requirement for autocrine prolactin, mammary glands from lactation-defective Akt1(-/-);Akt2(+/-) mice failed to express autocrine prolactin or activate Stat5 during late pregnancy despite normal levels of circulating serum prolactin and pituitary prolactin production. Our findings reveal that PI3K-Akt pathway activation is necessary and sufficient to induce autocrine prolactin production in the mammary gland, Stat5 activation, and terminal mammary epithelial differentiation, even in the absence of the normal developmental program that prepares the mammary gland for lactation. Together, these findings identify a function for autocrine prolactin during normal development and demonstrate its endogenous regulation by the PI3K-Akt pathway.


Subject(s)
Gene Expression Regulation , Lactation/physiology , Prolactin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Animals , Autocrine Communication/physiology , Cell Differentiation , Cells, Cultured , Down-Regulation , Female , Gene Deletion , Lactation/genetics , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Milk Proteins/metabolism , PTEN Phosphohydrolase/genetics , Pregnancy , Prolactin/genetics , Proto-Oncogene Proteins c-akt/genetics
18.
Patholog Res Int ; 2012: 814146, 2012.
Article in English | MEDLINE | ID: mdl-22701199

ABSTRACT

The incidence of esophageal adenocarcinoma (EAC) has increased exponentially in the last 3 decades. Barrett's esophagus (BE) is the only known precursor of EAC. Patients with BE have a greater than 40 folds higher risk of EAC compared with the general population. Recent years have witnessed a revolution in the clinical and molecular research related to BE. However, several aspects of this condition remain controversial. Data regarding the true prevalence of BE have varied widely. Recent studies have suggested a lower incidence of EAC in nondysplastic BE (NDBE) than previously reported. There is paucity of prospective data showing a survival benefit of screening or surveillance for BE. Furthermore, the ever-increasing emphasis on healthcare cost containment has called for reexamination of the screening and surveillance strategies for BE. There is a need for identification of reliable clinical predictors or molecular biomarkers to risk-stratify patients who might benefit the most from screening or surveillance for BE. Finally, new therapies have emerged for the management of dysplastic BE. In this paper, we highlight the key areas of controversy and uncertainty surrounding BE. The paper discusses, in detail, the current literature about the molecular pathogenesis, biomarkers, histopathological diagnosis, and management strategies for BE.

19.
Neoplasia ; 13(9): 792-805, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21969813

ABSTRACT

The incidence of esophageal adenocarcinoma (EAC) is rising in the United States. An important risk factor for EAC is the presence of Barrett esophagus (BE). BE is the replacement of normal squamous esophageal epithelium with a specialized columnar epithelium in response to chronic acid and bile reflux. However, the emergence of BE from squamous keratinocytes has not yet been demonstrated. Our research has focused on this. Wnt and cyclooxygenase 2 (Cox2) are two pathways whose activation has been associated with BE and progression to EAC, but their role has not been tested experimentally. To explore their contribution, we engineered a human esophageal keratinocyte cell line to express either a dominant-active Wnt effector CatCLef or a Cox2 complementary DNA. In a two-dimensional culture environment, Cox2 expression increases cell proliferation and migration, but neither transgene induces known BE markers. In contrast, when these cells were placed into three-dimensional organotypic culture conditions, we observed more profound effects. CatCLef-expressing cells were more proliferative, developed a thicker epithelium, and upregulated Notch signaling and several BE markers including NHE2. Cox2 expression also increased cell proliferation and induced a thicker epithelium. More importantly, we observed cysts form within the epithelium, filled with intestinal mucins including Muc5B and Muc17. This suggests that Cox2 expression in a three-dimensional culture environment induces a lineage of mucin-secreting cells and supports an important causal role for Cox2 in BE pathogenesis. We conclude that in vitro modeling of BE pathogenesis can be improved by enhancing Wnt signaling and Cox2 activity and using three-dimensional organotypic culture conditions.


Subject(s)
Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Cyclooxygenase 2/metabolism , Esophageal Neoplasms/pathology , Esophagus/pathology , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adenocarcinoma , Barrett Esophagus/genetics , Cell Line , Cell Lineage , Cell Movement , Cell Proliferation , Cyclooxygenase 2/biosynthesis , Humans , Keratinocytes , Mucin-5B/metabolism , Mucins/metabolism , Organ Culture Techniques , Receptors, Notch/metabolism , Sodium-Hydrogen Exchangers/metabolism , TCF Transcription Factors/genetics , Wnt Proteins/biosynthesis , Wnt Proteins/metabolism , beta Catenin/genetics
20.
Cancer Cell ; 19(4): 470-83, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21481789

ABSTRACT

p120-catenin (p120ctn) interacts with E-cadherin, but to our knowledge, no formal proof that p120ctn functions as a bona fide tumor suppressor gene has emerged to date. We report herein that p120ctn loss leads to tumor development in mice. We have generated a conditional knockout model of p120ctn whereby mice develop preneoplastic and neoplastic lesions in the oral cavity, esophagus, and squamous forestomach. Tumor-derived cells secrete granulocyte macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor-α (TNFα). The tumors contain significant desmoplasia and immune cell infiltration. Immature myeloid cells comprise a significant percentage of the immune cells present and likely participate in fostering a favorable tumor microenvironment, including the activation of fibroblasts.


Subject(s)
Carcinoma, Squamous Cell/etiology , Catenins/genetics , Esophageal Neoplasms/etiology , Genes, Tumor Suppressor , Inflammation/etiology , Mouth Neoplasms/etiology , Animals , Cadherins/analysis , Catenins/analysis , Catenins/deficiency , Catenins/physiology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Fibroblasts/physiology , Humans , Mice , Myeloid Cells/physiology , NF-kappa B/physiology , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...