Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Sci Rep ; 13(1): 22626, 2023 12 18.
Article in English | MEDLINE | ID: mdl-38114532

ABSTRACT

Oral squamous cell carcinomas unusually show distant metastasis to the lung after primary treatment, which can be difficult to differentiate from primary squamous cell carcinoma of the lung. While the location and number of tumor nodules is helpful in diagnosing cases, differential diagnosis may be difficult even with histopathological examination. Therefore, we attempted to identify molecules that can facilitate accurate differential diagnosis. First, we performed a comprehensive gene expression analysis using microarray data for OSCC-LM and LSCC, and searched for genes showing significantly different expression levels. We then identified KRT13, UPK1B, and nuclear receptor subfamily 0, group B, member 1 (NR0B1) as genes that were significantly upregulated in LSCC and quantified the expression levels of these genes by real-time quantitative RT-PCR. The expression of KRT13 and UPK1B proteins were then examined by immunohistochemical staining. While OSCC-LM showed no KRT13 and UPK1B expression, some tumor cells of LSCC showed KRT13 and UPK1B expression in 10 of 12 cases (83.3%). All LSCC cases were positive for at least one of these markers. Thus, KRT13 and UPK1B might contribute in differentiating OSCC-LM from LSCC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Lung Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Mouth Neoplasms/diagnosis , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Diagnosis, Differential , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Lung/pathology , Head and Neck Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Uroplakin Ib/genetics , Uroplakin Ib/metabolism , Keratin-13/genetics , Keratin-13/metabolism
2.
Comput Biol Med ; 158: 106795, 2023 05.
Article in English | MEDLINE | ID: mdl-36989746

ABSTRACT

Neoadjuvant chemotherapy (NAC) prior to surgery and immune checkpoint therapy (ICT) has revolutionized bladder cancer (BCa) treatment. Patients likely to benefit from these therapies need to be accurately stratified; however, this remains a major clinical challenge. In the present study, single-cell RNA sequencing was used to evaluate the predictive ability of an epithelial cell population highly expressing keratin 13 (KRT13) to assess therapeutic response in BCa. The presence of KRT13-enriched tumors indicated favorable outcomes after NAC and superior response to ICT in patients with BCa. Furthermore, KRT13 population characteristics appeared to be closely related to changes in the immune microenvironment in the vicinity of this cell population. We constructed a prognostic model using an artificial neural network based on the gene signatures in the KRT13 population; the model demonstrated strong robustness and superiority. Additionally, a user-friendly and open-access web application named BCa database was developed for researchers to study BCa by mining the connective map database.


Subject(s)
Keratin-13 , Urinary Bladder Neoplasms , Humans , Keratin-13/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Epithelial Cells/pathology , Immunotherapy , Tumor Microenvironment
3.
APMIS ; 131(2): 77-91, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36269681

ABSTRACT

Cytokeratin 13 (CK13) is a type I acidic low molecular weight cytokeratin, which is mainly expressed in urothelium and in the squamous epithelium of various sites of origin. Loss of CK13 has been implicated in the development and progression of squamous epithelial neoplasms. To comprehensively determine CK13 expression in normal and neoplastic tissues, a tissue microarray containing 10,439 samples from 131 different tumor types and subtypes as well as 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry. CK13 immunostaining was detectable in 42 (32.1%) of the 131 tumor categories including 24 (18.3%) tumor types with at least one strongly positive case. The highest rate of positive staining was found in various urothelial neoplasms (52.1-92.3%) including Brenner tumor of the ovary (86.8%) and in squamous cell carcinomas from various sites of origin (39.1-77.6%), Warthin tumors of parotid glands (66.7%), adenosquamous carcinomas of the cervix (33.3%), thymomas (16.0%), and endometroid carcinomas of the ovary (15.3%). Twenty other epithelial or germ cell neoplasms showed - a usually weak - CK13 positivity in less than 15% of the cases. In bladder cancer, reduced CK13 expression was linked to high grade and advanced stage (p < 0.0001 each). In squamous cell carcinoma of the cervix, reduced CK13 immunostaining was related to high grade (p = 0.0295) and shortened recurrence-free (p = 0.0094) and overall survival (p = 0.0274). In a combined analysis of 1,151 squamous cell carcinomas from 11 different sites of origin, reduced CK13 staining was linked to high grade (p = 0.0050). Our data provide a comprehensive overview on CK13 expression in normal and neoplastic human tissues. CK13 expression predominates in urothelial neoplasms and in squamous cell carcinomas of different organs, and a loss of CK13 expression is associated with aggressive disease in these tumors.


Subject(s)
Carcinoma, Squamous Cell , Keratin-13 , Urinary Bladder Neoplasms , Female , Humans , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Keratin-13/analysis , Keratin-13/genetics , Staining and Labeling , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
4.
Breast Cancer Res ; 24(1): 7, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078507

ABSTRACT

BACKGROUND: Keratins (KRTs) are intermediate filament proteins that interact with multiple regulatory proteins to initiate signaling cascades. Keratin 13 (KRT13) plays an important role in breast cancer progression and metastasis. The objective of this study is to elucidate the mechanism by which KRT13 promotes breast cancer growth and metastasis. METHODS: The function and mechanisms of KRT13 in breast cancer progression and metastasis were assessed by overexpression and knockdown followed by examination of altered behaviors in breast cancer cells and in xenograft tumor formation in mouse mammary fat pad. Human breast cancer specimens were examined by immunohistochemistry and multiplexed quantum dot labeling analysis to correlate KRT13 expression to breast cancer progression and metastasis. RESULTS: KRT13-overexpressing MCF7 cells displayed increased proliferation, invasion, migration and in vivo tumor growth and metastasis to bone and lung. Conversely, KRT13 knockdown inhibited the aggressive behaviors of HCC1954 cells. At the molecular level, KRT13 directly interacted with plakoglobin (PG, γ-catenin) to form complexes with desmoplakin (DSP). This complex interfered with PG expression and nuclear translocation and abrogated PG-mediated suppression of c-Myc expression, while the KRT13/PG/c-Myc signaling pathway increased epithelial to mesenchymal transition and stem cell-like phenotype. KRT13 expression in 58 human breast cancer tissues was up-regulated especially at the invasive front and in metastatic specimens (12/18) (p < 0.05). KRT13 up-regulation in primary breast cancer was associated with decreased overall patient survival. CONCLUSIONS: This study reveals that KRT13 promotes breast cancer cell growth and metastasis via a plakoglobin/c-Myc pathway. Our findings reveal a potential novel pathway for therapeutic targeting of breast cancer progression and metastasis.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Keratin-13/genetics , Keratin-13/metabolism , Mice , Neoplasm Metastasis , Proto-Oncogene Proteins c-myc , Signal Transduction , gamma Catenin/genetics , gamma Catenin/metabolism
5.
Head Neck ; 43(11): 3374-3385, 2021 11.
Article in English | MEDLINE | ID: mdl-34338386

ABSTRACT

BACKGROUND: This study aimed to evaluate the prognostic significance of expression levels of involucrin (IVL), cytokeratin (CK)-10 and -13 at different intratumor sites (tumor center and invading area) of oral tongue squamous cell carcinoma (OTSCC). METHODS: IVL, CK13 and CK10 expression levels were examined in a multicenter cohort of 146 OTSCCs using immunohistochemistry. External mRNA datasets were used for expression analysis and/or to validate survival associations. RESULTS: External transcriptomic datasets showed downregulation of IVL and KRT13 in oral malignancies including OTSCC as compared to normal controls. The combined loss of IVL and CK13 expression at the invading core but not at the center core was significantly associated with poor differentiation and reduced 5-year overall survival. Multivariate Cox analysis confirmed the loss of CK13 and IVL expression to be an independent prognostic factor. Transcriptomic dataset corroborated immunohistochemistry results. CONCLUSIONS: Combined expression levlels of IVL and CK13 might be useful as prognostic biomarkers in OTSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Keratin-13 , Protein Precursors , Squamous Cell Carcinoma of Head and Neck , Tongue Neoplasms , Carcinoma, Squamous Cell/genetics , Humans , Keratin-13/genetics , Prognosis , Tongue Neoplasms/genetics
6.
In Vitro Cell Dev Biol Anim ; 57(3): 300-314, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33537930

ABSTRACT

Cancer metastasis and recurrence are potentially lethal. A small number of cancer cell groups called cancer stem cells (CSCs) have both stem cell capacity and cancer-forming ability and are reported to play important roles in cancer metastasis and recurrence. These CSCs are considered to be radiation-resistant (RR). Therefore, understanding the biological effects of radiation on squamous cell carcinoma (SCC) cell lines in vitro and in vivo might be worthwhile to circumvent radiation resistance. Currently, there are no reports on the establishment of RR-SCC cells in serum-free defined culture, which mimics biological mechanisms and prevents instability of using serum in the culture medium. We isolated radiation-resistant strains, designated A431-LDR and A431-HDR, from A431 cells derived from vulval SCC and irradiated them with a total dose of 60 Gy at a low-dose rate (2.2 Gy/d) (RM1000) and a high-dose rate (5 Gy/5.75min) in serum-free defined culture. These cells exhibited high sphere-forming and migration ability in vitro and high tumor-forming ability in nude mice xenografts. Overexpression of KRT13 in A431-RR cells might play a role in its radiation-resistant characteristics. These cells might be useful not only to study cancer stem cells but also to study the circumvention of radiation resistance by novel cancer treatment modalities.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Keratin-13/genetics , Radiation Tolerance/genetics , Vulvar Neoplasms/genetics , Animals , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Culture Media, Serum-Free , Dose-Response Relationship, Radiation , Female , Humans , Keratin-13/metabolism , Mice, Nude , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , RNA, Small Interfering/metabolism , Tumor Stem Cell Assay , Vulvar Neoplasms/pathology
7.
Ned Tijdschr Geneeskd ; 1642020 09 15.
Article in Dutch | MEDLINE | ID: mdl-32940983

ABSTRACT

White sponge naevus (WSN) is a rare, autosomal dominant disorder that causes various complaints WSN is most commonly found on the buccal mucosa. Clinically, the white, slightly elevated lesions of WSN may be confused with other disorders on oral mucosa. We report a case of WSN in a 14-year-old boy who had complaints for a considerable period of time. WSN is caused by mutations in KRT4 and KRT13.


Subject(s)
Keratin-13/genetics , Keratin-4/genetics , Leukokeratosis, Hereditary Mucosal/genetics , Adolescent , Humans , Leukokeratosis, Hereditary Mucosal/pathology , Male , Mouth Mucosa/pathology , Mutation
8.
Dev Biol ; 468(1-2): 146-153, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32758484

ABSTRACT

White sponge nevus (WSN) is a benign autosomal dominant disorder characterized by the formation of white spongy plaques in the oral mucosa. Keratin (KRT) 13 is highly expressed in the mucosa, and mutations in this gene have been commonly associated with WSN patients. However, it remains unknown whether there is a causal relationship between KRT13 mutations and WSN and what the underlying mechanisms might be. Here, we use mouse genetic models to demonstrate that Krt13 is crucial for the maintenance of epithelial integrity. Krt13 knockout mice show a WSN-like phenotype in several tissues, including the tongue, buccal mucosa, and esophagus. Transcriptome analyses uncover that Krt13 regulates a cohort of gene networks in tongue epithelial cells, including epithelial differentiation, immune responses, stress-activated kinase signaling, and metabolic processes. We also provide evidence that epithelial cells without Krt13 are susceptible to mechanical stresses experienced during postnatal life, resulting in unbalanced cell proliferation and differentiation. These data demonstrate that Krt13 is essential for maintaining epithelial homeostasis and loss of Krt13 causes the WSN-like phenotype in mice.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells , Keratin-13/genetics , Leukokeratosis, Hereditary Mucosal , Mouth Mucosa , Mutation , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Keratin-13/metabolism , Leukokeratosis, Hereditary Mucosal/embryology , Leukokeratosis, Hereditary Mucosal/genetics , Leukokeratosis, Hereditary Mucosal/pathology , Mice , Mice, Knockout , Mouth Mucosa/embryology , Mouth Mucosa/pathology
9.
Biotechnol Lett ; 40(5): 765-772, 2018 May.
Article in English | MEDLINE | ID: mdl-29605938

ABSTRACT

OBJECTIVE: To establish a dermal sheath cell line, a dermal papilla cell line and a outer root sheath cell line from Cashmere goat and clarify the similarities and differences among them. RESULTS: We established a dermal sheath cell line, a dermal papilla cell line and a outer root sheath cell line from the pelage skin hair follicles of Cashmere goat. The growth rate of dermal sheath cells was intermediate between that of dermal papilla cells and outer root sheath cells. Immunofluorescence experiments and reverse transcription-polymerase chain reaction analysis showed that at both the transcriptional and translational levels, the dermal sheath cells were alpha-smooth muscle actin (α-SMA)+/cytokeratin 13+, while the dermal papilla cells were α-SMA+/cytokeratin 13- and the outer root sheath cells were α-SMA-/cytokeratin 13+. Patterns of cytokeratin 13 expression could distinguish the dermal sheath cells from the dermal papilla cells. CONCLUSIONS: These results suggest that cytokeratin 13 could serve as a novel biomarker for dermal sheath cells of Cashmere goat, and should prove useful for researchers investigating dermal stem cells or interaction of different types of cells during hair cycle.


Subject(s)
Dermis/cytology , Epidermal Cells , Keratin-13/genetics , Keratin-13/metabolism , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Line , Cell Proliferation , Dermis/metabolism , Epidermis/metabolism , Goats , Hair Follicle/cytology , Hair Follicle/metabolism , Organ Specificity
10.
Med Oral Patol Oral Cir Bucal ; 23(2): e144-e150, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29476668

ABSTRACT

BACKGROUND: Oral white sponge nevus (WSN) is a rare autosomal dominant benign condition, characterized by asymptomatic spongy white plaques. Mutations in Keratin 4 (KRT4) and 13 (KRT13) have been shown to cause WSN. Familial cases are uncommon due to irregular penetrance. Thus, the aim of the study was: a) to demonstrate the clinical and histopathological features of a three-generation Turkish family with oral WSN b) to determine whether KRT4 or KRT13 gene mutation was the molecular basis of WSN. MATERIAL AND METHODS: Out of twenty members of the family ten were available for assessment. Venous blood samples from six affected and five unaffected members and 48 healthy controls were obtained for genetic mutational analysis. Polymerase chain reaction was used to amplify all exons within KRT4 and KRT13 genes. These products were sequenced and the data was examined for mutations and polymorphisms. RESULTS: Varying presentation and severity of clinical features were observed. Analysis of the KRT13 gene revealed the sequence variant Y118D as the disease-causing mutation. One patient revealed several previously unreported polymorphisms including a novel mutation in exon 1 of the KRT13 gene and a heterozygous deletion in exon 1 of KRT4. This deletion in the KRT4 gene was found to be a common polymorphism reflecting a high allele frequency of 31.25% in the Turkish population. CONCLUSIONS: Oral WSN may manifest variable clinical features. The novel mutation found in the KRT13 gene is believed to add evidence for a mutational hotspot in the mucosal keratins. Molecular genetic analysis is required to establish correct diagnosis and appropriate genetic consultation.


Subject(s)
Keratin-13/genetics , Keratin-4/genetics , Leukokeratosis, Hereditary Mucosal/diagnosis , Leukokeratosis, Hereditary Mucosal/genetics , Adolescent , Adult , Case-Control Studies , Child , Cytogenetic Analysis , Humans , Male , Middle Aged , Mutation , Pedigree , Turkey , Young Adult
11.
Biochem Biophys Res Commun ; 496(2): 381-386, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29326042

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a biological event in which epithelial cells lose their polarity and cell-cell adhesions and concomitantly acquire mesenchymal traits, and is thought to play an important role in pathological processes such as wound healing and cancer progression. In this study, we evaluated transforming growth factor (TGF)-ß1-treated human keratinocyte HaCaT cells as an in vitro model of EMT. HaCaT cells were changed into an elongated fibroblast-like morphology, which is indicative of EMT in response to TGF-ß1. Phalloidin staining demonstrated the formation of actin stress fibers in TGF-ß1-treated cells. Quantitative RT-PCR analysis revealed that TGF-ß1 increased the mRNA levels of EMT transcription factors (SNAI2, TWIST1, and ZEB1) and mesenchymal markers (CDH2, VIM, and FN1), while it decreased the transcripts of epithelial phenotypic genes (CLDN1, OCLN, KRT5, KRT15, KRT13, and TGM1). Furthermore, we found that KRT13 was drastically suppressed through the reduction of RNA polymerase II occupancy of its promoter, which was accompanied by a decrease in active histone marks (H3K4me3 and H3K27ac) and an increase in a repressive mark (H3K27me3) during EMT. These findings indicate that the TGF-ß1-induced EMT program regulates a subset of epithelial and mesenchymal marker genes, and that KRT13 is transcriptionally suppressed through the modulation of the chromatin state at the KRT13 promoter in HaCaT cells.


Subject(s)
Epigenesis, Genetic , Epithelial-Mesenchymal Transition/drug effects , Keratin-13/genetics , Keratinocytes/drug effects , Transforming Growth Factor beta1/pharmacology , Actins/genetics , Actins/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line, Transformed , Cytokines/genetics , Cytokines/metabolism , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibronectins , Histones/genetics , Histones/metabolism , Humans , Keratin-13/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
12.
J Oral Pathol Med ; 47(2): 152-157, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29047160

ABSTRACT

BACKGROUND: White sponge nevus is a rare autosomal dominant disorder that affects the non-keratinised stratified squamous epithelium. Mutations in the genes that encode mucosa-specific keratin-4 and keratin-13 are strongly linked to the manifestation of white sponge nevus. This study involved mutational analysis of the genes encoding keratin-4 and keratin-13 in two Swedish families with white sponge nevus. METHODS: The diagnosis of white sponge nevus was based on disease history, clinical characteristics of the lesions and, in the majority of the cases, histopathological examination. Samples were collected from the affected buccal mucosa using buccal swabs. DNA was subsequently extracted and amplified using touchdown-PCR. The keratin-4 and keratin-13 genes were sequenced, and a genetic analysis was performed. RESULTS: A novel heterozygous missense mutation was identified in exon 1A of the keratin-4 gene in Family 2. In addition, previously reported heterozygous missense mutations were identified in the keratin-4 (E449K, A72V, Q156R, R208H) and keratin-13 (L115P) genes in both families. CONCLUSION: We describe a novel heterozygous missense mutation in the keratin-4 gene of a Swedish family with white sponge nevus. Our results support the notion that mutations in keratin-4 and keratin-13 are the underlying cause of white sponge nevus.


Subject(s)
Keratin-13/genetics , Keratin-4/genetics , Leukokeratosis, Hereditary Mucosal/genetics , Mouth Neoplasms/genetics , Mutation, Missense , Adult , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , DNA Mutational Analysis , Epithelium/pathology , Exons/genetics , Female , Heterozygote , Humans , Leukokeratosis, Hereditary Mucosal/pathology , Middle Aged , Mouth Mucosa/pathology , Mouth Neoplasms/pathology , Multigene Family , Pedigree , Sequence Analysis, Protein , Sweden , Young Adult
13.
Clin Cancer Res ; 23(14): 3769-3780, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28174235

ABSTRACT

Purpose: Head and neck squamous cell carcinomas (HNSCCs) cause more than 300,000 deaths worldwide each year. Locoregional and distant recurrences represent worse prognostic events and accepted surrogate markers of patients' overall survival. No valid biomarker and salvage therapy exist to identify and treat patients at high-risk of recurrence. We aimed to verify if selected miRNAs could be used as biomarkers of recurrence in HNSCC.Experimental Design: A NanoString array was used to identify miRNAs associated with locoregional recurrence in 44 patients with HNSCC. Bioinformatic approaches validated the signature and identified potential miRNA targets. Validation experiments were performed using an independent cohort of primary HNSCC samples and a panel of HNSCC cell lines. In vivo experiments validated the in vitro results.Results: Our data identified a four-miRNA signature that classified HNSCC patients at high- or low-risk of recurrence. These miRNAs collectively impinge on the epithelial-mesenchymal transition process. In silico and wet lab approaches showed that miR-9, expressed at high levels in recurrent HNSCC, targets SASH1 and KRT13, whereas miR-1, miR-133, and miR-150, expressed at low levels in recurrent HNSCC, collectively target SP1 and TGFß pathways. A six-gene signature comprising these targets identified patients at high risk of recurrences, as well. Combined pharmacological inhibition of SP1 and TGFß pathways induced HNSCC cell death and, when timely administered, prevented recurrence formation in a preclinical model of HNSCC recurrence.Conclusions: By integrating different experimental approaches and competences, we identified critical mediators of recurrence formation in HNSCC that may merit to be considered for future clinical development. Clin Cancer Res; 23(14); 3769-80. ©2017 AACR.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Keratin-13/genetics , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/pathology , Signal Transduction , Sp1 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck , Transforming Growth Factor beta/genetics , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(51): 84645-84657, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27835867

ABSTRACT

Lethal progression of prostate cancer metastasis can be improved by developing animal models that recapitulate the clinical conditions. We report here that cytokeratin 13 (KRT13), an intermediate filament protein, plays a directive role in prostate cancer bone, brain, and soft tissue metastases. KRT13 expression was elevated in bone, brain, and soft tissue metastatic prostate cancer cell lines and in primary and metastatic clinical prostate, lung, and breast cancer specimens. When KRT13 expression was determined at a single cell level in primary tumor tissues of 44 prostate cancer cases, KRT13 level predicted bone metastasis and the overall survival of prostate cancer patients. Genetically enforced KRT13 expression in human prostate cancer cell lines drove metastases toward mouse bone, brain and soft tissues through a RANKL-independent mechanism, as KRT13 altered the expression of genes associated with EMT, stemness, neuroendocrine/neuromimicry, osteomimicry, development, and extracellular matrices, but not receptor activator NF-κB ligand (RANKL) signaling networks in prostate cancer cells. Our results suggest new inhibitors targeting RANKL-independent pathways should be developed for the treatment of prostate cancer bone and soft tissue metastases.


Subject(s)
Adenocarcinoma/metabolism , Bone Neoplasms/metabolism , Brain Neoplasms/metabolism , Keratin-13/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/secondary , Animals , Bone Neoplasms/secondary , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Movement , Cellular Reprogramming , Gene Expression Regulation, Neoplastic , Humans , Keratin-13/genetics , Male , Mice , Mice, SCID , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , RANK Ligand/metabolism , Survival Analysis , Transcriptome , Up-Regulation , Xenograft Model Antitumor Assays
15.
PLoS One ; 11(10): e0163232, 2016.
Article in English | MEDLINE | ID: mdl-27711225

ABSTRACT

BACKGROUND: Benign human prostate tubule-initiating cells (TIC) and aggressive prostate cancer display common traits, including tolerance of low androgen levels, resistance to apoptosis, and microenvironment interactions that drive epithelial budding and outgrowth. TIC can be distinguished from epithelial and stromal cells that comprise prostate tissue via cell sorting based upon Epcam, CD44, and CD49f antigenic profiles. Fetal prostate epithelial cells (FC) possess a similar antigenic profile to adult TIC and are capable of inducing tubule formation. To identify the TIC niche in human prostate tissue, differential keratin (KRT) expression was evaluated. RESULTS: Gene expression data generated from Affymetrix Gene Chip human U133 Plus 2.0 array of sorted adult and fetal epithelial cells revealed KRT13 to be significantly enriched in FC and TIC compared to basal cells (BC) and luminal cells (LC) (p<0.001). Enriched KRT13 expression was confirmed by RT-PCR and cytospin immunostaining. Immunohistochemical analysis of KRT13 expression revealed rare KRT13+ epithelia throughout prostatic ducts/acini in adult tissue specimens and differentiated tubules in 24-week recombinant grafts, In contrast, abundant KRT13 expression was observed in developing ducts/acini in fetal prostate and cord-like structures composing 8-week recombinant grafts. Immunostaining of a prostate tissue microarray revealed KRT13+ tumor foci in approximately 9% of cases, and this subset displayed significantly shorter time to recurrence (p = 0.031), metastases (p = 0.032), and decreased overall survival (p = 0.004). Diagnostic prostate needle biopsies (PNBX) from untreated patients with concurrent bone metastases (clinical stage M1) displayed KRT13+ tumor foci, as did bone metastatic foci. CONCLUSIONS: The expression profile of KRT13 in benign fetal and adult prostate tissue and in recombinant grafts, as well as the frequency of KRT13 expression in primary and metastatic prostate cancer indicates that it may be a marker of a stem/progenitor-like cell state that is co-opted in aggressive tumor cells. KRT13 is enriched in benign stem-like cells that display androgen-resistance, apoptosis-resistance, and branching morphogenesis properties. Collectively our data demonstrate that KRT13 expression is associated with poor prognosis at multiple stages of disease progression and may represent an important biomarker of adverse outcome in patients with prostate cancer.


Subject(s)
Bone Neoplasms/secondary , Keratin-13/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , Adult , Animals , Biopsy, Needle , Cell Differentiation , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Keratin-13/genetics , Male , Mice , Neoplastic Stem Cells/pathology , Prostate/metabolism , Prostate/surgery , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery , Survival Analysis
16.
Hum Mol Genet ; 25(16): 3600-3612, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27378695

ABSTRACT

Rare and low frequency variants are not well covered in most germline genotyping arrays and are understudied in relation to epithelial ovarian cancer (EOC) risk. To address this gap, we used genotyping arrays targeting rarer protein-coding variation in 8,165 EOC cases and 11,619 controls from the international Ovarian Cancer Association Consortium (OCAC). Pooled association analyses were conducted at the variant and gene level for 98,543 variants directly genotyped through two exome genotyping projects. Only common variants that represent or are in strong linkage disequilibrium (LD) with previously-identified signals at established loci reached traditional thresholds for exome-wide significance (P < 5.0 × 10 - 7). One of the most significant signals (Pall histologies = 1.01 × 10 - 13;Pserous = 3.54 × 10 - 14) occurred at 3q25.31 for rs62273959, a missense variant mapping to the LEKR1 gene that is in LD (r2 = 0.90) with a previously identified 'best hit' (rs7651446) mapping to an intron of TIPARP. Suggestive associations (5.0 × 10 - 5 > P≥5.0 ×10 - 7) were detected for rare and low-frequency variants at 16 novel loci. Four rare missense variants were identified (ACTBL2 rs73757391 (5q11.2), BTD rs200337373 (3p25.1), KRT13 rs150321809 (17q21.2) and MC2R rs104894658 (18p11.21)), but only MC2R rs104894668 had a large effect size (OR = 9.66). Genes most strongly associated with EOC risk included ACTBL2 (PAML = 3.23 × 10 - 5; PSKAT-o = 9.23 × 10 - 4) and KRT13 (PAML = 1.67 × 10 - 4; PSKAT-o = 1.07 × 10 - 5), reaffirming variant-level analysis. In summary, this large study identified several rare and low-frequency variants and genes that may contribute to EOC susceptibility, albeit with possible small effects. Future studies that integrate epidemiology, sequencing, and functional assays are needed to further unravel the unexplained heritability and biology of this disease.


Subject(s)
Actins/genetics , Biotinidase/genetics , Keratin-13/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Receptor, Melanocortin, Type 2/genetics , Carcinoma, Ovarian Epithelial , Exome/genetics , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Neoplasm Proteins/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Polymorphism, Single Nucleotide
17.
Proc Natl Acad Sci U S A ; 112(49): E6770-9, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26598683

ABSTRACT

Avian integumentary organs include feathers, scales, claws, and beaks. They cover the body surface and play various functions to help adapt birds to diverse environments. These keratinized structures are mainly composed of corneous materials made of α-keratins, which exist in all vertebrates, and ß-keratins, which only exist in birds and reptiles. Here, members of the keratin gene families were used to study how gene family evolution contributes to novelty and adaptation, focusing on tissue morphogenesis. Using chicken as a model, we applied RNA-seq and in situ hybridization to map α- and ß-keratin genes in various skin appendages at embryonic developmental stages. The data demonstrate that temporal and spatial α- and ß-keratin expression is involved in establishing the diversity of skin appendage phenotypes. Embryonic feathers express a higher proportion of ß-keratin genes than other skin regions. In feather filament morphogenesis, ß-keratins show intricate complexity in diverse substructures of feather branches. To explore functional interactions, we used a retrovirus transgenic system to ectopically express mutant α- or antisense ß-keratin forms. α- and ß-keratins show mutual dependence and mutations in either keratin type results in disrupted keratin networks and failure to form proper feather branches. Our data suggest that combinations of α- and ß-keratin genes contribute to the morphological and structural diversity of different avian skin appendages, with feather-ß-keratins conferring more possible composites in building intrafeather architecture complexity, setting up a platform of morphological evolution of functional forms in feathers.


Subject(s)
Biological Evolution , Chromosome Mapping , Keratins/genetics , Skin/embryology , beta-Keratins/genetics , Animals , Chick Embryo , In Situ Hybridization , Keratin-13/genetics , RNA, Antisense/pharmacology , Skin/metabolism
18.
J Biol Chem ; 290(21): 13567-77, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25851906

ABSTRACT

Squamous cell differentiation requires the coordinated activation and repression of genes specific to the differentiation process; disruption of this program accompanies malignant transformation of epithelium. The exploration of genes that control epidermal proliferation and terminal differentiation is vital to better understand esophageal carcinogenesis. KLF4 is a member of the KLF family of transcription factors and is involved in both cellular proliferation and differentiation. This study using immunohistochemistry analysis of KLF4 in clinical specimens of esophageal squamous cell carcinoma (ESCC) demonstrated that decreased KLF4 was substantially associated with poor differentiation. Moreover, we determined that both KLF4 and KRT13 levels were undoubtedly augmented upon sodium butyrate-induced ESCC differentiation and G1 phase arrest. Conversely, silencing of KLF4 and KRT13 abrogated the inhibition of G1-S transition induced by sodium butyrate. Molecular investigation demonstrated that KLF4 transcriptionally regulated KRT13 and the expression of the two molecules appreciably correlated in ESCC tissues and cell lines. Collectively, these results suggest that KLF4 transcriptionally regulates KRT13 and is invovled in ESCC cell differentiation.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Cycle , Esophageal Neoplasms/pathology , Esophagus/metabolism , Gene Expression Regulation, Neoplastic , Keratin-13/genetics , Kruppel-Like Transcription Factors/metabolism , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Humans , Immunoenzyme Techniques , Keratin-13/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
19.
Asian Pac J Cancer Prev ; 16(3): 953-8, 2015.
Article in English | MEDLINE | ID: mdl-25735388

ABSTRACT

BACKGROUND: Expression of KRT13, FAIM2 and CYP2W1 appears to be influenced by risk habits, thus exploring the associations of these genes in oral squamous cell cancer (OSCC) with risk habits, clinico-pathological parameters and patient survival may be beneficial in identifying relevant biomarkers with different oncogenic pathways. MATERIALS AND METHODS: cDNAs from 41 OSCC samples with and without risk habits were included in this study. Quantitative real-time PCR was used to analyze KRT13, FAIM2 and CYP2W1 in OSCC. The housekeeping gene (GAPDH) was used as an endogenous control. RESULTS: Of the 41 OSCC samples, KRT13 was down-regulated in 40 samples (97.6%), while FAIM2 and CYP2W1 were down-regulated in 61.0% and 48.8%, respectively. Overall, there were no associations between KRT13, FAIM2 and CYP2W1 expression with risk habits, selected socio-demographic and clinico-pathological parameters and patient survival. CONCLUSIONS: Although this study was unable to show significance, there were some tendencies in the associations of KRT13, FAIM2 and CYP2W1 expression in OSCC with selected clinic-pathological parameters and survival.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinoma, Squamous Cell/genetics , Cytochrome P-450 Enzyme System/genetics , Keratin-13/genetics , Membrane Proteins/genetics , Mouth Neoplasms/genetics , RNA, Messenger/genetics , Adult , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cross-Sectional Studies , Cytochrome P450 Family 2 , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Neoplasm Staging , Prognosis , Real-Time Polymerase Chain Reaction , Risk Factors , Survival Rate
20.
BMC Cancer ; 14: 988, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25527207

ABSTRACT

BACKGROUND: Epigenetic modifications play important roles in the regulation of gene expression determining cellular phenotype as well as various pathologies such as cancer. Although the loss of keratin 13 (KRT13) is reportedly linked to malignant transformation of oral epithelial cells, the molecular mechanisms through which KRT13 is repressed in oral squamous cell carcinoma (OSCC) remain unclear. The aim of this study is to identify the epigenetic alterations of the KRT13 gene in OSCCs. METHODS: We investigated KRT13 expression levels and chromatin modifications of the KRT13 promoter in the three OSCC cell lines (HSC4, HSC3, and SAS). The expression levels of KRT13 protein and mRNA were analyzed by western blotting and quantitative reverse-transcription polymerase chain reaction, respectively, and the localization of KRT13 protein was detected by immunofluorescence. DNA methylation and histone modifications in the KRT13 promoter were determined by bisulfite sequencing and chromatin immunoprecipitation (ChIP), respectively. For the pharmacological depletion of Polycomb repressive complex 2 (PRC2), cells were treated with 3-deazaneplanocin A (DZNep). RESULTS: KRT13 expression was transcriptionally silenced in the HSC3 and SAS cells and post-transcriptionally repressed in the HSC4 cells, while the KRT13 promoter was hypermethylated in all of the three OSCC cell lines. ChIP analysis revealed that PRC2-mediated trimethylation of Lys 27 on histone H3 (H3K27me3) was increased in the KRT13 promoter in the HSC3 and SAS cells. Finally, we demonstrated that the treatment of SAS cells with DZNep reactivated the transcription of KRT13 gene. CONCLUSIONS: Our data provide mechanistic insights into the epigenetic silencing of KRT13 genes in OSCC cells and might be useful for the development of diagnostic markers and novel therapeutic approaches against OSCCs.


Subject(s)
Carcinoma, Squamous Cell/genetics , Epigenesis, Genetic , Keratin-13/genetics , Mouth Neoplasms/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , CpG Islands , DNA Methylation , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Keratin-13/metabolism , Methylation , Mouth Neoplasms/metabolism , Promoter Regions, Genetic , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL