Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Polymers (Basel) ; 15(18)2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37765665

ABSTRACT

Fluorinated polyimides incorporated with triptycene units have gained growing attention over the last decade since they present potentially interesting selectivities and a higher free volume with respect to their triptycene-free counterparts. This work examines the transport of single-gas and mixed-gas N2 and CH4 in the triptycene-based 6FDA-BAPT homopolyimide and in a block 15,000 g mol-1/15,000 g mol-1 6FDA-mPDA/BAPT copolyimide by using molecular dynamics (MD) simulations. The void-space analyses reveal that, while the free volume consists of small-to-medium holes in the 6FDA-BAPT homopolyimide, there are more medium-to-large holes in the 6FDA-mPDA/BAPT copolyimide. The single-gas sorption isotherms for N2 and CH4 over the 0-70 bar range at 338.5 K show that both gases are more soluble in the block copolyimide, with a higher affinity for methane. CH4 favours sites with the most favourable energetic interactions, while N2 probes more sites in the matrices. The volume swellings remain limited since neither N2 nor CH4 plasticise penetrants. The transport of a binary-gas 2:1 CH4/N2 mixture is also examined in both polyimides under operating conditions similar to those used in current natural gas processing, i.e., at 65.5 bar and 338.5 K. In the mixed-gas simulations, the solubility selectivities in favour of CH4 are enhanced similarly in both matrices. Although diffusion is higher in 6FDA-BAPT/6FDA-mPDA, the diffusion selectivities are also close. Both triptycene-based polyimides under study favour, to a similar extent, the transport of methane over that of nitrogen under the conditions studied.

2.
Phys Chem Chem Phys ; 24(31): 18667-18683, 2022 Aug 10.
Article in English | MEDLINE | ID: mdl-35894847

ABSTRACT

High-performance polymers with polybenzoxazole (PBO) structures, formed via thermal rearrangement (TR) of aromatic polyimide precursors, have been developed for gas separation applications. The present work compares the transport of N2 and CH4 in a 6FDA-bisAPAF polyimide precursor and in its TR-PBO derivative using molecular dynamics (MD) simulations. The modelling closely mimicked the experimental approach by transforming a 6FDA-bisAPAF atomistic model into its corresponding TR-PBO structure via a specific algorithm. The densities and void spaces of both precursor and TR polymers were found to compare well to experimental data. An iterative technique was used to obtain the single-gas sorption isotherms of N2 and CH4 at 338.5 K in both polymers over a range of feed pressures up to and exceeding 65 bar. CH4 was systematically found to be more soluble than N2. Solubilities in both matrices were quite similar with those in TR-PBO being slightly higher due to its larger fraction of significant volume. Volume dilation analyses confirmed a higher resistance to plasticization for TR-PBO. Extended single-gas N2 and CH4 simulations and 2 : 1 binary CH4/N2 mixed-gas simulations were then conducted in both matrices at 338.5 K and at a pressure of ∼65 bar corresponding to natural gas processing conditions. Mixed-gas sorption was modelled using a modification of the aforementioned iterative method, which fixed the pressure and iterated to convergence the number of molecules of each type of penetrant. The gas diffusion coefficients were estimated using the Trajectory-Extending Kinetic Monte Carlo (TEKMC) procedure. As found experimentally, significantly higher diffusivities and permeabilities were observed in the TR polymer, which led to a slightly lower ideal N2/CH4 permselectivity for TR-PBO (∼2.6) when compared to its 6FDA-bisAPAF precursor (∼3.8). However, both models showed a reduced N2/CH4 separation efficiency under 2 : 1 binary CH4/N2 mixed-gas conditions bordering on the loss of selectivity. For 6FDA-bisAPAF, both permeabilities decreased in the mixed-gas case, but more for N2 than for CH4. For TR-PBO, the permeability of the faster N2 decreased while the permeability of the slower CH4 increased under mixed-gas conditions. This confirms that single-gas simulations are not sufficient for the prediction of the actual mixed-gas permselectivity behaviour in such polymers.

3.
Life (Basel) ; 12(3)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35330094

ABSTRACT

Human oral cancer is the single largest group of malignancies in the Indian subcontinent and the sixth largest group of malignancies worldwide. Squamous cell carcinomas (SCC) are the most common epithelial malignancy of the oral cavity, constituting over 90% of oral cancers. About 90% of OSCCs arise from pre-existing, potentially malignant lesions. According to WHO, OSCC has a 5-year survival rate of 45-60%. Late diagnosis, recurrence, and regional or lymph nodal metastases could be the main causes of the high mortality rates. Biomarkers may help categorize and predict premalignant lesions as high risk of developing malignancy, local recurrence, and lymph nodal metastasis. However, at present, there is a dearth of such markers, and this is an area of ongoing research. Keratins (K) or cytokeratins are a group of intermediate filament proteins that show paired and differentiation dependent expression. Our laboratory and others have shown consistent alterations in the expression patterns of keratins in both oral precancerous lesions and tumors. The correlation of these changes with clinicopathological parameters has also been demonstrated. Furthermore, the functional significance of aberrant keratins 8/18 expression in the malignant transformation and progression of oral tumors has also been documented. This article reviews the literature that emphasizes the value of keratins as biomarkers for the prognostication of human oral precancers and cancers.

4.
Life (Basel) ; 12(2)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35207438

ABSTRACT

Oral carcinogenesis is a multistep process. As much as 5% to 85% of oral tumors can develop from potentially malignant disorders (PMD). Although the oral cavity is accessible for visual examination, the ability of current clinical or histological methods to predict the lesions that can progress to malignancy is limited. Thus, developing biological markers that will serve as an adjunct to histodiagnosis has become essential. Our previous studies comprehensively demonstrated that aberrant vimentin expression in oral premalignant lesions correlates to the degree of malignancy. Likewise, overwhelming research from various groups show a substantial contribution of vimentin in oral cancer progression. In this review, we have described studies on vimentin in oral cancers, to make a compelling case for vimentin as a prognostic biomarker.

5.
Cell Biol Int ; 45(8): 1720-1732, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33847415

ABSTRACT

Keratin 8/18, the predominant keratin pair of simple epithelia, is known to be aberrantly expressed in several squamous cell carcinomas (SCCs), where its expression is often correlated with increased invasion, neoplastic progression, and poor prognosis. The majority of keratin 8/18 structural and regulatory functions are governed by posttranslational modifications, particularly phosphorylation. Apart from filament reorganization, cellular processes including cell cycle, cell growth, cellular stress, and apoptosis are known to be orchestrated by K8 phosphorylation at specific residues in the head and tail domains. Even though deregulation of K8 phosphorylation at two significant sites (Serine73 /Serine431 ) has been implicated in neoplastic progression of SCCs by various in vitro studies, including ours, it is reported to be highly context-dependent. Therefore, to delineate the precise role of Kereatin 8 phosphorylation in cancer initiation and progression, we have developed the tissue-specific transgenic mouse model expressing Keratin 8 wild type and phosphodead mutants under Keratin 14 promoter. Subjecting these mice to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-mediated skin carcinogenesis revealed that Keratin 8 phosphorylation may lead to an early onset of tumors compared to Keratin 8 wild-type expressing mice. Conclusively, the transgenic mouse model developed in the present study ascertained a positive impact of Keratin 8 phosphorylation on the neoplastic transformation of skin-squamous cells.


Subject(s)
Carcinogenesis/metabolism , Keratin-8/metabolism , Mutation/physiology , Skin Neoplasms/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Electroporation/methods , HEK293 Cells , Humans , Keratin-8/genetics , Male , Mice , Mice, Transgenic , Phosphorylation/physiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
6.
J Biosci ; 44(2)2019 Jun.
Article in English | MEDLINE | ID: mdl-31180046

ABSTRACT

Keratins, the epithelial-predominant members of the intermediate filament superfamily, are expressed in a pairwise, tissuespecific and differentiation-dependent manner. There are 28 type I and 26 type II keratins, which share a common structure comprising a central coiled coil α-helical rod domain flanked by two nonhelical head and tail domains. These domains harbor sites for major posttranslational modifications like phosphorylation and glycosylation, which govern keratin function and dynamics. Apart from providing structural support, keratins regulate various signaling machinery involved in cell growth, motility, apoptosis etc. However, tissue-specific functions of keratins in relation to cell proliferation and differentiation are still emerging. Altered keratin expression pattern during and after malignant transformation is reported to modulate different signaling pathways involved in tumor progression in a context-dependent fashion. The current review focuses on the literature related to the role of keratins in the regulation of cell proliferation, differentiation and transformation in different types of epithelia.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Keratins/genetics , Neoplasms/genetics , Protein Processing, Post-Translational , Acetylation , Animals , Apoptosis/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Glycosylation , Humans , Keratins/chemistry , Keratins/classification , Keratins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Protein Structure, Secondary , Signal Transduction
7.
Microsc Microanal ; 25(6): 1367-1375, 2019 12.
Article in English | MEDLINE | ID: mdl-30867083

ABSTRACT

Early detection of altered epithelium can help in controlling the further progression by timely intervention. Alterations in cellular adhesion are one of the hallmarks of cancer progression, which can be detected at the intracellular level using high-resolution electron microscopy. This study aimed to evaluate the role of electron microscopy in the establishment of ultrastructural markers for early detection of altered epithelium using tissues from 4-Nitroquinoline-1-Oxide (4NQO) induced rat tongue carcinogenesis. Our previous study using light microscopy displayed no histopathological alterations in 4NQO treated tissues until 40 days of treatment, while dysplasia, papilloma and carcinoma were detected at 80/120, 160 and 200 days, respectively. However, electron microscopy detected alterations such as detachment of desmosomes from cell membranes and their clustering in the cytoplasm, increased tonofilaments, keratohyaline granules and thickened corneum in 40 days treated corresponding tissues. These alterations are apparent with hyperkeratosis/hyperplasia but remained undetected using light microscopy. Further, in dysplasia, papilloma and carcinoma, gradual and significant loss of desmosomes, leading to the significant widening of intercellular spaces, was observed using iTEM software. These parameters may serve as indicators for progression of oral cancer. Our results highlight the importance of electron microscopy in the early detection of subcellular changes in the altered epithelium.


Subject(s)
Carcinoma/diagnosis , Epithelium/pathology , Microscopy, Electron/methods , Mouth Mucosa/pathology , Mouth Neoplasms/diagnosis , 4-Nitroquinoline-1-oxide/administration & dosage , Animals , Carcinogens/administration & dosage , Disease Models, Animal , Early Diagnosis , Rats
8.
Exp Mol Pathol ; 105(3): 243-251, 2018 12.
Article in English | MEDLINE | ID: mdl-30189187

ABSTRACT

OBJECTIVE: We have previously reported the aberrant expression of vimentin in human oral premalignant lesions and a 4-Nitroquinoline 1-oxide (4NQO) model of rat lingual carcinogenesis. Hence, we wanted to understand whether the expression of vimentin in early stage contributes to the process of transformation. STUDY DESIGN: Vimentin was stably expressed in oral premalignant lesion derived cells (vimentin negative) and various transformation related phenotypic assays were performed. Since vimentin alone failed to transform the cells, an additional carcinogenic stimulus benzo[a]pyrene (BP) was used. Concomitantly, immunohistochemistry (IHC) was performed on oral leukoplakia and tumor tissues for studying the expression of vimentin and E-cadherin. RESULTS: Exogenous expression of vimentin led to the appearance of EMT and stemness-related signatures. Further, upon BP treatment, vimentin expressing clones showed an increase in vitro and in vivo transformation efficiency. Importantly, high vimentin-low E-cadherin expression significantly correlated with the grade of dysplasia, as also with the lymph node metastasis in oral tumors. CONCLUSION: Our study suggests that the expression of vimentin in early stages may be beneficial, although not sufficient to achieve transformation. Further, high vimentin-low E-cadherin expression, if validated in more number of early oral lesions, may prove useful in the identification of high risk human premalignant lesions.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition/physiology , Mouth Neoplasms/pathology , Precancerous Conditions/pathology , Vimentin/metabolism , Animals , Cell Transformation, Neoplastic/pathology , Heterografts , Humans , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Precancerous Conditions/metabolism
9.
Eur J Oral Sci ; 126(4): 251-262, 2018 08.
Article in English | MEDLINE | ID: mdl-29905981

ABSTRACT

With the aim of developing early diagnostic/prognostic markers for oral cancer, desmosomal adhesion in sequentially progressive grades of tissues from oral normal/disorders (normal, hyperplastic, dysplastic, non-metastatic/metastatic tumours, and metastatic nodes) was investigated at protein and ultrastructural levels using immunohistochemistry and transmission electron microscopy, respectively. The expression of desmosomal proteins was higher in hyperplastic tissues than in normal tissues but was significantly decreased in subsequent progressive stages of the disease. Altered expression of desmosomal proteins was significantly correlated with local recurrence and disease-free survival. Ultrastructural analysis in the corresponding tissues revealed cytoplasmic clustering of desmosomes in hyperplasia; in more advanced disease stages, a significantly lower number of desmosomes and widened intercellular spaces were observed. Altered protein expression resulting in structural changes was confirmed by knocking down desmoplakin expression in non-transformed cells, which failed to form normal desmosome structures and induced a cell-transformation phenotype. Our data suggest that alterations in desmosomal assembly initiate at an early hyperplastic grade and, with more advanced disease stages, the severity of the alterations gradually becomes higher. Alterations in desmosomal adhesion can be useful for early detection of high-risk premalignant lesions, as well as for identification of invasive characteristics of primary non-metastatic tumours. Early detection will help to control further progression of disease by timely intervention.


Subject(s)
Carcinogenesis/pathology , Desmosomes/ultrastructure , Mouth Neoplasms/pathology , Blotting, Western , Cell Adhesion , Cell Movement , Cells, Cultured , Desmoglein 2/metabolism , Desmoplakins/metabolism , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Microscopy, Electron, Transmission , Neoplasm Grading , Stem Cells , Survival Rate , gamma Catenin/metabolism
10.
Oncol Rep ; 39(5): 2393-2401, 2018 May.
Article in English | MEDLINE | ID: mdl-29512781

ABSTRACT

Keratins 5/14 (K5/14) are intermediate filament proteins expressed in the basal layer of stratified epithelial cells and are known targets of p63. Previous research in our laboratory showed that upon K5/14 downregulation in oral squamous cell carcinoma (OSCC)­derived cells, there was an increase in intracellular Notch­1 levels and differentiation markers such as involucrin, keratin 1 and a decrease in tumorigenic potential in vivo. However, the molecules involved in the K14 regulated cell differentiation and transformation are not known to date. In order to understand the possible role of TAp63, we downregulated TAp63 in a K14­knockdown background. We observed that there was a decrease in the expression of Notch­1. Expression levels of differentiation markers such as involucrin, K1, loricrin and filaggrin were also decreased. Furthermore, TAp63 downregulation led to an increase in invasion, migration and in vivo tumorigenic potential of these cells. We observed a decrease in ß­catenin signaling in K14­downregulated cells. Notably, when TAp63 was downregulated in K14­knockdown cells, there was increase in non­phospho ß­catenin levels. Hence, this study indicates that TAp63 plays an important role in K14­downregulated cells possibly by regulating the Notch­1 expression. K14 regulates the expression of TAp63 which in turn regulates expression of Notch­1. The present study is a step forward in our quest to understand the functional significance of molecules that regulate the process of differentiation and tumorigenesis in stratified epithelial cells.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Keratin-14/metabolism , Keratin-5/metabolism , Mouth Neoplasms/metabolism , Receptor, Notch1/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Down-Regulation , Filaggrin Proteins , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation
11.
FEBS J ; 285(7): 1251-1276, 2018 04.
Article in English | MEDLINE | ID: mdl-29427328

ABSTRACT

Keratin 8/18, the predominant keratin pair of simple epithelia, is often aberrantly expressed in various squamous cell carcinomas (SCCs) including skin SCC. Its aberrant expression is correlated with increased invasiveness and poor prognosis of the same, although the underlying mechanism is still unclear. A previous report from our laboratory has shown K8-mediated regulation of α6ß4 integrin signaling and thereby tumorigenic potential of oral SCC-derived cells. Another study on transgenic mouse model has shown that during skin carcinogenesis, K8 favors conversion of papillomas toward malignancy. In order to understand the role of K8 and allied mechanism in skin SCC, K8 was stably knocked down in a skin epidermoid carcinoma-derived A431 cells. K8 downregulation significantly reduced the tumorigenic potential of these cells. In agreement with our phenotypic data, differential quantitative proteomics followed by IPA analysis showed altered expression of many proteins associated with biological functions including 'Cancer', 'Cellular movement', 'Cell death and survival', and 'Cellular morphology'. Some of these proteins were TMS1, MARCKSL1, RanBP1, 14-3-3γ, Rho-GDI2, etc. Furthermore, to our surprise, there was a significant reduction in K17 protein stability upon loss of K8, probably due to its caspase-mediated degradation. This was supported by altered TMS1-NF-κB signaling, leading to increased apoptotic sensitivity of A431 cells which in turn affected 'Cell death and survival'. Moreover, MARCKSL1-Paxillin1-Rac axis was found to be deregulated bestowing a possible mechanism behind altered 'Cellular movement' pathway. Altogether our study unravels a much broader regulatory role of K8, governing multiple signaling pathways and consequently regulating oncogenic potential of skin SCC-derived cells. DATABASE: Proteome Xchange Consortium via PRIDE database (dataset identifier PXD007206).


Subject(s)
Keratin-18 , Keratin-8 , Signal Transduction/genetics , Carcinogenesis/genetics , Cell Survival/genetics , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , Immunohistochemistry , Keratin-18/genetics , Keratin-18/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Proteomics
12.
Exp Cell Res ; 360(2): 125-137, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28867478

ABSTRACT

BPAG1e and Plectin are hemidesmosomal linker proteins which anchor intermediate filament proteins to the cell surface through ß4 integrin. Recent reports indicate that these proteins play a role in various cellular processes apart from their known anchoring function. However, the available literature is inconsistent. Further, the previous study from our laboratory suggested that Keratin8/18 pair promotes cell motility and tumor progression by deregulating ß4 integrin signaling in oral squamous cell carcinoma (OSCC) derived cells. Based on these findings, we hypothesized that linker proteins may have a role in neoplastic progression of OSCC. Downregulation of hemidesmosomal linker proteins in OSCC derived cells resulted in reduced cell migration accompanied by alterations in actin organization. Further, decreased MMP9 activity led to reduced cell invasion in linker proteins knockdown cells. Moreover, loss of these proteins resulted in reduced tumorigenic potential. SWATH analysis demonstrated upregulation of N-Myc downstream regulated gene 1 (NDRG1) in linker proteins downregulated cells as compared to vector control cells. Further, the defects in phenotype upon linker proteins ablation were rescued upon loss of NDRG1 in linker proteins knockdown background. These data together indicate that hemidesmosomal linker proteins regulate cell motility, invasion and tumorigenicity possibly through NDRG1 in OSCC derived cells.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/pathology , Cell Movement/genetics , Cytoskeletal Proteins/physiology , Hemidesmosomes/physiology , Mouth Neoplasms/pathology , Animals , Carcinogenesis/pathology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Dystonin/physiology , HEK293 Cells , Hemidesmosomes/genetics , Hemidesmosomes/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mouth Neoplasms/genetics , Neoplasm Invasiveness , Plectin/genetics , Plectin/physiology
13.
PLoS One ; 12(2): e0172559, 2017.
Article in English | MEDLINE | ID: mdl-28225793

ABSTRACT

Vimentin is an intermediate filament protein, predominantly expressed in cells of mesenchymal origin, although its aberrant expression is seen in many carcinomas during epithelial mesenchymal transition. In cancer, vimentin expression is associated with the transition from a more differentiated epithelial phenotype to a dedifferentiated state. In view of the perceived role of keratins (Ks) as regulators of differentiation in epithelia, it was important to understand whether vimentin modulates differentiation through the reprogramming of keratins, in transformed cells. To address this, vimentin was stably downregulated in oral cancer derived cells. Further, global keratin profiling was performed after high salt keratin extraction. K5/K14 pair was found to be significantly downregulated, both at protein and mRNA levels upon vimentin downregulation. The previous study from our laboratory has shown a role of the K5/K14 pair in proliferation and differentiation of squamous epithelial cells. Vimentin depleted cells showed an increase in the differentiation state, marked by an increase in the levels of differentiation specific markers K1, involucrin, filaggrin and loricrin while its proliferation status remained unchanged. Rescue experiments with the K5/K14 pair overexpressed in vimentin knockdown background resulted in decreased differentiation state. ΔNp63 emerged as one of the indirect targets of vimentin, through which it modulates the expression levels of K5/K14. Further, immunohistochemistry showed a significant correlation between high vimentin-K14 expression and recurrence/poor survival in oral cancer patients. Thus, in conclusion, vimentin regulates the differentiation switch via modulation of K5/K14 expression. Moreover, vimentin-K14 together may prove to be the novel markers for the prognostication of human oral cancer.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic , Keratin-14/genetics , Mouth Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Vimentin/genetics , Animals , Cell Line, Tumor , Down-Regulation , Female , Filaggrin Proteins , Humans , Keratin-14/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Male , Mice , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Prognosis , Receptors, Notch/genetics , Receptors, Notch/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vimentin/metabolism
14.
Proteomics ; 17(7)2017 04.
Article in English | MEDLINE | ID: mdl-28176443

ABSTRACT

Keratin 8/18, a simple epithelia specific keratin pair, is often aberrantly expressed in squamous cell carcinomas (SCC) where its expression is correlated with increased invasion and poor prognosis. Majority of Keratin 8 (K8) functions are governed by its phosphorylation at Serine73 (head-domain) and Serine431 (tail-domain) residues. Although, deregulation of K8 phosphorylation is associated with progression of different carcinomas, its role in skin-SCC and the underlying mechanism is obscure. In this direction, we performed tandem mass tag-based quantitative phosphoproteomics by expressing K8 wild type, phosphodead, and phosphomimetic mutants in K8-deficient A431 cells. Further analysis of our phosphoproteomics data showed a significant proportion of total phosphoproteome associated with migratory, proliferative, and invasive potential of these cells to be differentially phosphorylated. Differential phosphorylation of CDK1T14,Y15 , EIF4EBP1T46,T50 , EIF4BS422 , AKT1S1T246,S247 , CTTN1T401,S405,Y421 , and CAP1S307/309 in K8-S73A/D mutant and CTTN1T401,S405,Y421 , BUB1BS1043 , and CARHSP1S30,S32 in K8-S431A/D mutants as well as some anonymous phosphosites including MYCS176 , ZYXS344 , and PNNS692 could be potential candidates associated with K8 phosphorylation mediated tumorigenicity. Biochemical validation followed by phenotypic analysis further confirmed our quantitative phosphoproteomics data. In conclusion, our study provides the first global picture of K8 site-specific phosphorylation function in neoplastic progression of A431 cells and suggests various potential starting points for further mechanistic studies.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Keratin-8/genetics , Phosphoproteins/genetics , Proteomics/methods , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , CDC2 Protein Kinase , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cortactin/genetics , Cortactin/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/pathology , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Humans , Keratin-8/metabolism , Mutation , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Skin/metabolism , Skin/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Biophotonics ; 10(10): 1377-1384, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28067994

ABSTRACT

Accurate understanding of cellular processes and responses to stimuli is of paramount importance in biomedical research and diagnosis. Raman spectroscopy (RS), a label-free and nondestructive spectroscopic method has the potential to serve as a novel 'theranostics' tool. Both fiber-optic and micro-Raman studies have demonstrated efficacy in diagnostics and therapeutic response monitoring. In the present study, we have evaluated the potential of micro-Raman spectroscopic maps in identifying changes induced by loss of K8/18 proteins in a tongue cancer cell line. Furthermore, we also evaluated the efficacy of less expensive and commercially available fiber probes to identify K8/18 wild and knock-down cell pellets, in view of the utility of cell pellet-based studies. The findings suggest that major differences in the cellular morphology and biochemical composition can be objectively identified and can be utilized for classification using both micro-Raman and fiber-probe-based RS. These findings highlight the potential of fiber-optic probe-based RS in noninvasive cellular phenotyping for diagnosis and therapeutic response monitoring, especially in low-resource settings.


Subject(s)
Gene Knockdown Techniques , Keratin-18/deficiency , Keratin-18/genetics , Keratin-8/deficiency , Keratin-8/genetics , Spectrum Analysis, Raman , Cell Line, Tumor , Humans , Mouth Neoplasms/pathology
16.
PLoS One ; 11(8): e0160615, 2016.
Article in English | MEDLINE | ID: mdl-27501241

ABSTRACT

To study multistep tumorigenesis process, there is a need of in-vitro 3D model simulating in-vivo tissue. Present study aimed to reconstitute in-vitro tissue models comprising various stages of neoplastic progression of tongue tumorigenesis and to evaluate the utility of these models to investigate the role of stromal fibroblasts in maintenance of desmosomal anchoring junctions using transmission electron microscopy. We reconstituted in-vitro models representing normal, dysplastic, and malignant tissues by seeding primary keratinocytes on either fibroblast embedded in collagen matrix or plain collagen matrix in growth factor-free medium. The findings of histomorphometry, immunohistochemistry, and electron microscopy analyses of the three types of 3D cultures showed that the stratified growth, cell proliferation, and differentiation were comparable between co-cultures and their respective native tissues; however, they largely differed in cultures grown without fibroblasts. The immunostaining intensity of proteins, viz., desmoplakin, desmoglein, and plakoglobin, was reduced as the disease stage increased in all co-cultures as observed in respective native tissues. Desmosome-like structures were identified using immunogold labeling in these cultures. Moreover, electron microscopic observations revealed that the desmosome number and their length were significantly reduced and intercellular spaces were increased in cultures grown without fibroblasts when compared with their co-culture counterparts. Our results showed that the major steps of tongue tumorigenesis can be reproduced in-vitro. Stromal fibroblasts play a role in regulation of epithelial thickness, cell proliferation, differentiation, and maintenance of desmosomalanchoring junctions in in-vitro grown tissues. The reconstituted co-culture models could help to answer various biological questions especially related to tongue tumorigenesis.


Subject(s)
Coculture Techniques/methods , Tongue Neoplasms/pathology , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , Desmogleins/metabolism , Desmoplakins/metabolism , Desmosomes/metabolism , Desmosomes/ultrastructure , Fibroblasts/pathology , Humans , Keratinocytes/pathology , Tongue/pathology , gamma Catenin
17.
J Biol Chem ; 291(23): 12003-13, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27059955

ABSTRACT

Keratins 8/18 (K8/18) are phosphoglycoproteins and form the major intermediate filament network of simple epithelia. The three O-GlcNAcylation (Ser(29), Ser(30), and Ser(48)) and two phosphorylation (Ser(33) and Ser(52)) serine sites on K18 are well characterized. Both of these modifications have been reported to increase K18 solubility and regulate its filament organization. In this report, we investigated the site-specific interplay between these two modifications in regulating the functional properties of K18, like solubility, stability, and filament organization. An immortalized hepatocyte cell line (HHL-17) stably expressing site-specific single, double, and triple O-GlcNAc and phosphomutants of K18 were used to identify the site(s) critical for regulating these functions. Keratin 18 mutants where O-GlcNAcylation at Ser(30) was abolished (K18-S30A) exhibited reduced phosphorylation induced solubility, increased stability, defective filament architecture, and slower migration. Interestingly, K18-S30A mutants also showed loss of phosphorylation at Ser(33), a modification known to regulate the solubility of K18. Further to this, the K18 phosphomutant (K18-S33A) mimicked K18-S30A in its stability, filament organization, and cell migration. These results indicate that O-GlcNAcylation at Ser(30) promotes phosphorylation at Ser(33) to regulate the functional properties of K18 and also impact cellular processes like migration. O-GlcNAcylation and phosphorylation on the same or adjacent sites on most proteins antagonize each other in regulating protein functions. Here we report a novel, positive interplay between O-GlcNAcylation and phosphorylation at adjacent sites on K18 to regulate its fundamental properties.


Subject(s)
Acetylglucosamine/metabolism , Keratin-18/metabolism , Serine/metabolism , Acylation , Binding Sites/genetics , Cell Line , Cell Movement/genetics , Fibronectins/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Immunoblotting , Keratin-18/genetics , Microscopy, Confocal , Mutation, Missense , Phosphorylation , Serine/genetics
18.
Int J Biochem Cell Biol ; 70: 161-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26646105

ABSTRACT

Vimentin expression correlates well with migratory and invasive potential of the carcinoma cells. The molecular mechanism by which vimentin regulates cell motility is not yet clear. Here, we addressed this issue by depleting vimentin in oral squamous cell carcinoma derived cell line. Vimentin knockdown cells showed enhanced adhesion and spreading to laminin-5. However, we found that they were less invasive as compared to the vector control cells. In addition, signaling associated with adhesion behavior of the cell was increased in vimentin knockdown clones. These findings suggest that the normal function of ß4 integrin as mechanical adhesive device is enhanced upon vimentin downregulation. As a proof of principle, the compromised invasive potential of vimentin depleted cells could be rescued upon blocking with ß4 integrin adhesion-blocking (ASC-8) antibody or downregulation of ß4 integrin in vimentin knockdown background. Interestingly, plectin which associates with α6ß4 integrin in the hemidesmosomes, was also found to be upregulated in vimentin knockdown clones. Furthermore, experiments on lysosome and proteasome inhibition revealed that perhaps vimentin regulates the turnover of ß4 integrin and plectin. Moreover, an inverse association was observed between vimentin expression and ß4 integrin in oral squamous cell carcinoma (OSCC). Collectively, our results show a novel role of vimentin in modulating cell motility by destabilizing ß4 integrin-mediated adhesive interactions. Further, vimentin-ß4 integrin together may prove to be useful markers for prognostication of human oral cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Integrin beta4/genetics , Mouth Neoplasms/genetics , Vimentin/genetics , Antibodies, Neutralizing/pharmacology , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Female , Hemidesmosomes/drug effects , Hemidesmosomes/metabolism , Hemidesmosomes/ultrastructure , Humans , Integrin beta4/metabolism , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Male , Middle Aged , Mouth Neoplasms/diagnosis , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Neoplasm Invasiveness , Neoplasm Staging , Plectin/genetics , Plectin/metabolism , Primary Cell Culture , Prognosis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Vimentin/antagonists & inhibitors , Vimentin/metabolism , Kalinin
19.
Clin Oral Investig ; 20(1): 43-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25914047

ABSTRACT

OBJECTIVE: In the present study, we have investigated the prognostic value of known stem cell-associated molecules such as Oct4, CD44 and c-Myc in patients with oral SCC who had received post-surgery radio- and/or chemotherapy. MATERIALS AND METHODS: Immunohistochemistry was performed to analyse the expression of Oct4, CD44 and c-Myc in 87 tumour tissues, and the expression profile obtained was correlated with clinicopathological parameters of the patients with oral cancer. Tumourigenic potential of these molecules was also evaluated by in vivo studies. RESULTS: Our results showed significant correlation of Oct4 (OS, p = 0.003; DFS, p = 0.001) and c-Myc (OS, p = 0.01; DFS, p = 0.03) with overall survival and disease-free survival independently. Furthermore, all the three markers in combinations of two markers each, i.e. Oct4 + CD44 (OS, p = 0.003; DFS, p = 0.001), Oct4 + c-Myc (OS, p = 0.0001; DFS, p = 0.0001), CD44 + c-Myc (OS, p = 0.008; DFS, p = 0.02) and in combinations of three markers each, i.e. Oct4 + CD44 + c-Myc (OS, p = 0.0001; DFS, p = 0.0001) also significantly correlated with overall survival and disease-free survival. Univariate and multivariate analyses further established the independent prognostic value of Oct4. Oct4-, CD44- and c-Myc-enriched populations independently induced sarcomatoid carcinomas whereas primary keratinocytes developed poorly differentiated carcinomas in immunodeficient mice. CONCLUSIONS: Oct4 and c-Myc independently as well as in combination with CD44 might be useful for the prediction of local recurrence and poor survival of patients with oral cancer which is the novel finding of this study. CLINICAL RELEVANCE: Oct4, c-Myc and CD44 can be used to predict local recurrence and the outcome of treatment in oral cancer patients. In addition, these molecules may find use as molecular targets for effective therapy.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , DNA-Binding Proteins/metabolism , Hyaluronan Receptors/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/therapy , Octamer Transcription Factor-3/metabolism , Salivary alpha-Amylases/metabolism , Transcription Factors/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Middle Aged , Mouth Neoplasms/pathology , Neoplasm Recurrence, Local , Prognosis , Survival Rate
20.
PLoS One ; 10(9): e0136542, 2015.
Article in English | MEDLINE | ID: mdl-26325577

ABSTRACT

Omega 3 (n3) and Omega 6 (n6) polyunsaturated fatty acids (PUFAs) have been reported to exhibit opposing roles in cancer progression. Our objective was to determine whether different ratios of n6/n3 (AA/EPA+DHA) FAs could modulate the cell viability, lipid peroxidation, total cellular fatty acid composition and expression of tumor regulatory Matrix Attachment Region binding proteins (MARBPs) in breast cancer cell lines and in non-cancerous, MCF10A cells. Low ratios of n6/n3 (1:2.5, 1:4, 1:5, 1:10) FA decreased the viability and growth of MDA-MB-231 and MCF7 significantly compared to the non-cancerous cells (MCF10A). Contrarily, higher n6/n3 FA (2.5:1, 4:1, 5:1, 10:1) decreased the survival of both the cancerous and non-cancerous cell types. Lower ratios of n6/n3 selectively induced LPO in the breast cancer cells whereas the higher ratios induced in both cancerous and non-cancerous cell types. Interestingly, compared to higher n6/n3 FA ratios, lower ratios increased the expression of tumor suppressor MARBP, SMAR1 and decreased the expression of tumor activator Cux/CDP in both breast cancer and non-cancerous, MCF10A cells. Low n6/n3 FAs significantly increased SMAR1 expression which resulted into activation of p21WAF1/CIP1 in MDA-MB-231 and MCF7, the increase being ratio dependent in MDA-MB-231. These results suggest that increased intake of n3 fatty acids in our diet could help both in the prevention as well as management of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/pharmacology , Lipid Peroxidation , MCF-7 Cells/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Blotting, Western , Breast Neoplasms/chemistry , Cell Line, Tumor/chemistry , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-6/analysis , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells/chemistry , MCF-7 Cells/drug effects , alpha-Linolenic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...