Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 703, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121738

ABSTRACT

Rho family mechano-signaling through the actin cytoskeleton positively regulates physiological TEAD/YAP transcription, while the evolutionarily conserved Hippo tumor suppressor pathway antagonizes this transcription through YAP cytoplasmic localization/degradation. The mechanisms responsible for oncogenic dysregulation of these pathways, their prevalence in tumors, as well as how such dysregulation can be therapeutically targeted are not resolved. We demonstrate that p53 DNA contact mutants in human tumors, indirectly hyperactivate RhoA/ROCK1/actomyosin signaling, which is both necessary and sufficient to drive oncogenic TEAD/YAP transcription. Moreover, we demonstrate that recurrent lesions in the Hippo pathway depend on physiological levels of ROCK1/actomyosin signaling for oncogenic TEAD/YAP transcription. Finally, we show that ROCK inhibitors selectively antagonize proliferation and motility of human tumors with either mechanism. Thus, we identify a cancer driver paradigm and a precision medicine approach for selective targeting of human malignancies driven by TEAD/YAP transcription through mechanisms that either upregulate or depend on homeostatic RhoA mechano-signaling.


Subject(s)
Cell Cycle Proteins/genetics , Neoplasms/genetics , Signal Transduction/genetics , TEA Domain Transcription Factors/genetics , Transcription Factors/genetics , rho-Associated Kinases/genetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/genetics , Humans , Mice, SCID , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , TEA Domain Transcription Factors/metabolism , Transcription Factors/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays/methods , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
2.
Oral Dis ; 28(4): 1094-1103, 2022 May.
Article in English | MEDLINE | ID: mdl-33615634

ABSTRACT

OBJECTIVE: Oral submucous fibrosis (OSF) is a debilitating potentially malignant condition of the buccal cavity characterized by extensive extracellular matrix deposition resulting in stiffness and trismus. As OSF is a progressive disease, we hypothesized that there would be extensive epigenetic changes in OSF tissues. MATERIALS AND METHODS: Using the Infinium HumanMethylation450 BeadChip Array, we analyzed gross DNA methylation changes in seven OSF tissues compared to five controls. Comparison with transcriptomic data and pathway analyses was conducted to find commonly regulated genes. RESULTS: A total of 3,294 differentially methylated regions mapping to 857 genes were identified. Comparison with transcriptome data revealed 38 downregulated-hypermethylated genes and 55 hypomethylated-upregulated genes. Using methylation-specific and qRT-PCR, aberrant hypomethylation and increased expression of FGF13, RPS6KA3, and ACSL4 genes were confirmed. Pathways involved in insulin signaling, ubiquitin-mediated proteolysis, nicotine addiction, and RAS/MAPK pathways were dysregulated, among others. Intriguingly, numerous genes located on the X chromosome were dysregulated in OSF tissues as the transcript for XIST gene was downregulated due to hypermethylation of the XIST promoter. CONCLUSIONS: This study highlights global epigenetic dysregulation of tissues of the oral cavity in OSF patients and hints at possible X chromosomal dysregulation, previously not implicated in the pathogenesis of OSF.


Subject(s)
DNA Methylation , Oral Submucous Fibrosis , Areca , Epigenesis, Genetic , Humans , Mouth Mucosa/pathology , Oral Submucous Fibrosis/genetics , Oral Submucous Fibrosis/pathology , Promoter Regions, Genetic/genetics
3.
Nat Commun ; 12(1): 6889, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824220

ABSTRACT

Inflammatory Breast Cancer (IBC) is a highly aggressive malignancy with distinct clinical and histopathological features whose molecular basis is unresolved. Here we describe a human IBC cell line, A3250, that recapitulates key IBC features in a mouse xenograft model, including skin erythema, diffuse tumor growth, dermal lymphatic invasion, and extensive metastases. A3250 cells express very high levels of the CCL2 chemokine and induce tumors enriched in macrophages. CCL2 knockdown leads to a striking reduction in macrophage densities, tumor proliferation, skin erythema, and metastasis. These results establish IBC-derived CCL2 as a key factor driving macrophage expansion, and indirectly tumor growth, with transcriptomic analysis demonstrating the activation of multiple inflammatory pathways. Finally, primary human IBCs exhibit macrophage infiltration and an enriched macrophage RNA signature. Thus, this human IBC model provides insight into the distinctive biology of IBC, and highlights potential therapeutic approaches to this deadly disease.


Subject(s)
Chemokine CCL2/metabolism , Inflammatory Breast Neoplasms/metabolism , Inflammatory Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Inflammation , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/immunology , Mice , Mice, SCID , Myeloid Cells/metabolism , Neoplasm Metastasis , Receptors, CCR2/metabolism , Transplantation, Heterologous , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
4.
Periodontol 2000 ; 80(1): 213-224, 2019 06.
Article in English | MEDLINE | ID: mdl-31090136

ABSTRACT

Many oral mucosal lesions are due to substance abuse, such as tobacco and areca nut, amongst others. There is considerable evidence that oral lesions/disorders such as some leukoplakias, most erythroplakias, and submucous fibrosis have malignant potential, with a conversion rate of 5%-10% over a 10-year period. There have been several reports on possible biomarkers that predict malignant conversion of the oral lesions associated with these disorders. Management of these is mostly surgical removal of the lesion followed by observation, and in some cases treatment by antioxidants and anti-inflammatory agents. Oral submucous fibrosis is due to excessive deposition of extracellular matrix in the connective tissue plus, particularly, collagens. The deposition of collagen leads to stiffness of the affected regions and results in difficulty in mouth opening. Areca nut chewing is proposed as the most probable etiological factor in the manifestation of oral submucous fibrosis. Several studies suggest involvement of proinflammatory cytokines, dysregulated by areca nut, in the development of the disease. Amongst these, transforming growth factor-ß is in the forefront, which is also shown to be involved in fibrosis of other organs. This review addresses the molecular mechanisms involved in oral submucous fibrosis development and provides a model for the regulation of transforming growth factor-ß by areca nut. It provides an exemplar of the role of modern molecular techniques in the study of oral disease.


Subject(s)
Mouth Diseases , Oral Submucous Fibrosis , Areca , Humans , Mastication , Nuts
5.
J Inorg Biochem ; 174: 45-54, 2017 09.
Article in English | MEDLINE | ID: mdl-28601723

ABSTRACT

Oxovanadium(IV) complexes [VO(L1/L2)Cl2]n+ (1,2) of (anthracenyl)terpyridine (An-tpy as L1 in 1, n=0) and triphenylphosphonium-appended (anthracenyl)terpyridine (An-tpy-TPP+ as L2 in 2, n=1) were synthesized, characterized and their DNA crosslinking ability, photocytotoxicity in visible light and cellular localization in cancer cells studied. The bromide derivative of 2, viz. [VO(An-tpy-TPP)Br2]Br (3) is structurally characterized. The structure showed trans disposition of two halides in the coordination sphere and the TPP+ unit is a pendant to the terpyridyl ligand. The DNA melting and comet assay studies on the complexes suggest the formation of DNA crosslinks. Complexes 1 and 2 displayed ~10 fold increase in cytotoxicity on exposure to visible light (400-700nm) when compared to those in dark in HeLa and MCF-7 cells. FACScan (Fluorescence Associated Cell Sorter Scan) analysis showed cellular apoptosis when treated with the complex in visible light in comparison to their dark controls. Fluorescence microscopic studies using complex 2 revealed its mitochondrial localization within the cancer cells.


Subject(s)
Anthracenes , Cross-Linking Reagents , DNA, Neoplasm/metabolism , Neoplasms/drug therapy , Phototherapy , Vanadates , Anthracenes/chemical synthesis , Anthracenes/chemistry , Anthracenes/pharmacology , Apoptosis/drug effects , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondria/pathology , Neoplasms/metabolism , Neoplasms/pathology , Vanadates/chemical synthesis , Vanadates/chemistry , Vanadates/pharmacology
6.
Sci Rep ; 6: 34314, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708346

ABSTRACT

Oral submucous fibrosis (OSF) is potentially premalignant with progressive and irreversible extracellular matrix deposition accompanied by epithelial atrophy and like other fibrotic disorders, is primarily a TGF-ß driven disease. OSF is caused by prolonged chewing of areca nut. Our previous studies reported a pivotal role for TGF-ß activation and its effects contributing to OSF. However, the mechanism for activation of TGF-ß signaling in OSF is still unknown. In this study we demonstrate activation of TGF-ß signaling with sub-cytotoxic dose of areca nut in epithelial cells and discovered a key role for pJNK in this process. In good correlation; pJNK was detected in OSF tissues but not in normal tissues. Moreover, activation of JNK was found to be dependent on muscarinic acid receptor induced Ca2+/CAMKII as well as ROS. JNK dependent phosphorylation of ATF2/c-Jun transcription factors resulted in TGF-ß transcription and its signaling. pATF2/p-c-Jun were enriched on TGF-ß promoter and co-localized in nuclei of epithelial cells upon areca nut treatment. In corroboration, OSF tissue sections also had nuclear pATF2 and p-c-Jun. Our results provide comprehensive mechanistic details of TGF-ß signaling induced by etiological agent areca nut in the manifestation of fibrosis which can lead to new therapeutic modalities for OSF.


Subject(s)
Activating Transcription Factor 2/metabolism , Areca/chemistry , MAP Kinase Kinase 4/metabolism , Mouth Mucosa , Mouth Neoplasms , Nuts/chemistry , Oncogene Protein p65(gag-jun)/metabolism , Plant Extracts/pharmacology , Precancerous Conditions , Transforming Growth Factor beta/metabolism , Cell Line, Transformed , Female , Fibrosis , Humans , Male , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Plant Extracts/chemistry , Precancerous Conditions/drug therapy , Precancerous Conditions/metabolism , Precancerous Conditions/pathology
7.
Inorg Chem ; 55(12): 6027-35, 2016 Jun 20.
Article in English | MEDLINE | ID: mdl-27244480

ABSTRACT

Ternary cobalt(III) complexes of curcumin (Hcur) and mitocurcumin [Hmitocur, a dicationic bis(triphenylphosphonium) derivative of curcumin] having a tetradentate phenolate-based ligand (H2L), namely, [Co(cur)(L)] (1) and [Co(mitocur)(L)]Cl2 (2), were prepared and structurally characterized, and their photoinduced cytotoxicity was studied. The diamagnetic cobalt(III) complexes show an irreversible Co(III)-Co(II) redox response and a quasireversible curcuminoid-based reduction near -1.45 and -1.74 V SCE, respectively, in DMF/0.1 M [(n)Bu4N](ClO4). The complexes exhibit a curcumin/mitocurcumin-based absorption band near 420 nm. Complex 1 was structurally characterized by X-ray crystallography. The structure contains the metal in a CoN2O4 distorted octahedral coordination arrangement with curcumin binding to the metal in its enolic form. Binding to cobalt(III) increases the hydrolytic stability of curcumin. Complex 2, having a dicationic curcuminoid, shows significant cellular uptake and photoinduced cytotoxicity compared to its curcumin analogue 1. The dicationic cobalt(III) complex 2 has significantly better cellular uptake and bioactivity than the neutral species 1. Complex 2 with mitochondrial localization releases the mitocurcumin dye upon exposure to visible light (400-700 nm) in human breast cancer MCF-7 cells through photoreduction of cobalt(III) to cobalt(II). Complex 2 displays a remarkable photodynamic therapy (PDT) effect, giving an IC50 value of ∼3.9 µM in visible light (400-700 nm) in MCF-7 cells while being much less toxic in the dark (>50 µM). The released mitocurcumin acts as a phototoxin, generating intracellular reactive oxygen species (ROSs). The overall process leads to light-controlled delivery of a curcuminoid (mitocur) into the tumor cells while the dye alone suffers from hydrolytic instability and poor bioavailability.


Subject(s)
Apoptosis/drug effects , Cobalt/chemistry , Coordination Complexes/chemistry , Curcumin/analogs & derivatives , Light , Photochemotherapy , Coordination Complexes/pharmacology , Curcumin/administration & dosage , Curcumin/pharmacology , Humans , MCF-7 Cells
8.
J Cell Mol Med ; 19(10): 2397-412, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26248978

ABSTRACT

Exposure of oral cavity to areca nut is associated with several pathological conditions including oral submucous fibrosis (OSF). Histopathologically OSF is characterized by epithelial atrophy, chronic inflammation, juxtaepithelial hyalinization, leading to fibrosis of submucosal tissue and affects 0.5% of the population in the Indian subcontinent. As the molecular mechanisms leading to atrophied epithelium and fibrosis are poorly understood, we studied areca nut actions on human keratinocyte and gingival fibroblast cells. Areca nut water extract (ANW) was cytotoxic to epithelial cells and had a pro-proliferative effect on fibroblasts. This opposite effect of ANW on epithelial and fibroblast cells was intriguing but reflects the OSF histopathology such as epithelial atrophy and proliferation of fibroblasts. We demonstrate that the pro-proliferative effects of ANW on fibroblasts are dependent on insulin-like growth factor signalling while the cytotoxic effects on keratinocytes are dependent on the generation of reactive oxygen species. Treatment of keratinocytes with arecoline which is a component of ANW along with copper resulted in enhanced cytotoxicity which becomes comparable to IC(50) of ANW. Furthermore, studies using cyclic voltammetry, mass spectrometry and plasmid cleavage assay suggested that the presence of arecoline increases oxidation reduction potential of copper leading to enhanced cleavage of DNA which could generate an apoptotic response. Terminal deoxynucleotidyl transferase dUTP Nick End Labeling assay and Ki-67 index of OSF tissue sections suggested epithelial apoptosis, which could be responsible for the atrophy of OSF epithelium.


Subject(s)
Areca/chemistry , Arecoline/toxicity , Copper/toxicity , Epithelium/pathology , Nuts/chemistry , Oral Submucous Fibrosis/pathology , Apoptosis/drug effects , Atrophy , Catalase/metabolism , Cell Proliferation/drug effects , Cells, Cultured , DNA Cleavage/drug effects , Epithelium/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , In Situ Nick-End Labeling , Keratinocytes/cytology , Keratinocytes/drug effects , Ki-67 Antigen/metabolism , Oxidation-Reduction/drug effects , Plant Extracts/toxicity , Receptor, IGF Type 1/metabolism , Superoxides/metabolism
9.
PLoS One ; 10(6): e0129252, 2015.
Article in English | MEDLINE | ID: mdl-26107172

ABSTRACT

Areca nut consumption has been implicated in the progression of Oral Submucous fibrosis (OSF); an inflammatory precancerous fibrotic condition. Our previous studies have demonstrated the activation of TGF-ß signaling in epithelial cells by areca nut components and also propose a role for epithelial expressed TGF-ß in the pathogenesis of OSF. Although the importance of epithelial cells in the manifestation of OSF has been proposed, the actual effectors are fibroblast cells. However, the role of areca nut and TGF-ß in the context of fibroblast response has not been elucidated. Therefore, to understand their role in the context of fibroblast response in OSF pathogenesis, human gingival fibroblasts (hGF) were treated with areca nut and/or TGF-ß followed by transcriptome profiling. The gene expression profile obtained was compared with the previously published transcriptome profiles of OSF tissues and areca nut treated epithelial cells. The analysis revealed regulation of 4666 and 1214 genes by areca nut and TGF-ß treatment respectively. The expression of 413 genes in hGF cells was potentiated by areca nut and TGF-ß together. Further, the differentially expressed genes of OSF tissues compared to normal tissues overlapped significantly with areca nut and TGF-ß induced genes in epithelial and hGF cells. Several positively enriched pathways were found to be common between OSF tissues and areca nut +TGF-ß treated hGF cells. In concordance, areca nut along with TGF-ß enhanced fibroblast activation as demonstrated by potentiation of αSMA, γSMA and collagen gel contraction by hGF cells. Furthermore, TGF-ß secreted by areca nut treated epithelial cells influenced fibroblast activation and other genes implicated in fibrosis. These data establish a role for areca nut influenced epithelial cells in OSF progression by activation of fibroblasts and emphasizes the importance of epithelial-mesenchymal interaction in OSF.


Subject(s)
Areca/chemistry , Epithelial-Mesenchymal Transition , Oral Submucous Fibrosis/chemically induced , Oral Submucous Fibrosis/pathology , Transforming Growth Factor beta/metabolism , Cluster Analysis , Collagen/chemistry , Disease Progression , Epithelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Genome, Human , Gingiva/drug effects , Humans , Inflammation , Nuts/chemistry , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/pathology , Signal Transduction , Transcriptome
10.
Inorg Chem ; 54(8): 3748-58, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25849848

ABSTRACT

Iron(III) complexes of pyridoxal (vitamin B6, VB6) or salicylaldehyde Schiff bases and modified dipicolylamines, namely, [Fe(B)(L)](NO3) (1-5), where B is phenyl-N,N-bis((pyridin-2-yl)methyl)methanamine (phbpa in 1), (anthracen-9-yl)-N,N-bis((pyridin-2-yl)methyl)methanamine (anbpa in 2, 4) and (pyren-1-yl)-N,N-bis((pyridin-2-yl)methyl)methanamine (pybpa in 3, 5) (H2L(1) is 3-hydroxy-5-(hydroxymethyl)-4-(((2-hydroxyphenyl)imino)methyl)-2-methylpyridine (1-3) and H2L(2) is 2-[(2-hydroxyphenyl-imino)methyl]phenol), were prepared and their uptake in cancer cells and photocytotoxicity were studied. Complexes 4 and 5, having a non-pyridoxal Schiff base, were prepared to probe the role of the pyridoxal group in tumor targeting and cellular uptake. The PF6 salt (1a) of complex 1 is structurally characterized. The complexes have a distorted six-coordinate FeN4O2 core where the metal is in the +3 oxidation state with five unpaired electrons. The complexes display a ligand to metal charge transfer band near 520 and 420 nm from phenolate to the iron(III) center. The photophysical properties of the complexes are explained from the time dependent density functional theory calculations. The redox active complexes show a quasi-reversible Fe(III)/Fe(II) response near -0.3 V vs saturated calomel electrode. Complexes 2 and 3 exhibit remarkable photocytotoxicity in various cancer cells with IC50 values ranging from 0.4 to 5 µM with 10-fold lower dark toxicity. The cell death proceeded by the apoptotic pathway due to generation of reactive oxygen species upon light exposure. The nonvitamin complexes 4 and 5 display 3-fold lower photocytotoxicity compared to their VB6 analogues, possibly due to preferential and faster uptake of the vitamin complexes in the cancer cells. Complexes 2 and 3 show significant uptake in the endoplasmic reticulum, while complexes 4 and 5 are distributed throughout the cells without any specific localization pattern.


Subject(s)
Ferric Compounds/metabolism , Ferric Compounds/pharmacology , Photosensitizing Agents/metabolism , Photosensitizing Agents/pharmacology , Pyridoxal/metabolism , Schiff Bases/metabolism , Schiff Bases/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Ferric Compounds/chemical synthesis , Ferric Compounds/chemistry , HeLa Cells , Humans , Light , MCF-7 Cells , Models, Molecular , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Pyridoxal/chemical synthesis , Pyridoxal/chemistry , Pyridoxal/pharmacology , Quantum Theory , Reactive Oxygen Species/metabolism , Schiff Bases/chemical synthesis , Schiff Bases/chemistry , Structure-Activity Relationship
11.
Dalton Trans ; 44(9): 4108-22, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25623080

ABSTRACT

Oxovanadium(IV) complexes of polypyridyl and curcumin-based ligands, viz. [VO(cur)(L)Cl] (1, 2) and [VO(scur)(L)Cl] (3, 4), where L is 1,10-phenanthroline (phen in 1 and 3), dipyrido[3,2-a:2',3'-c]phenazine (dppz in 2 and 4), Hcur is curcumin and Hscur is diglucosylcurcumin, were synthesized and characterized and their cellular uptake, photocytotoxicity, intracellular localization, DNA binding, and DNA photo-cleavage activity studied. Complex [VO(cur)(phen)Cl] (1) has V(IV)N2O3Cl distorted octahedral geometry as evidenced from its crystal structure. The sugar appended complexes show significantly higher uptake into the cancer cells compared to their normal analogues. The complexes are remarkably photocytotoxic in visible light (400-700 nm) giving an IC50 value of <5 µM in HeLa, HaCaT and MCF-7 cells with no significant dark toxicity. The green emission of the complexes was used for cellular imaging. Predominant cytosolic localization of the complexes 1-4 to a lesser extent into the nucleus was evidenced from confocal imaging. The complexes as strong binders of calf thymus DNA displayed photocleavage of supercoiled pUC19 DNA in red light by generating ˙OH radicals as the ROS. The cell death is via an apoptotic pathway involving the ROS. Binding to the VO(2+) moiety has resulted in stability against any hydrolytic degradation of curcumin along with an enhancement of its photocytotoxicity.


Subject(s)
Coordination Complexes , Curcumin , Photosensitizing Agents , Vanadates , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/radiation effects , Curcumin/chemistry , Curcumin/pharmacology , Curcumin/radiation effects , DNA/chemistry , Glucose/pharmacology , Humans , Hydrolysis , Light , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/radiation effects , Reactive Oxygen Species/metabolism , Vanadates/chemistry , Vanadates/pharmacology , Vanadates/radiation effects
12.
Chem Asian J ; 9(9): 2494-504, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25044734

ABSTRACT

Iron(III) complexes [Fe(L)(L')(NO3)]--in which L is phenyl-N,N-bis[(pyridin-2-yl)methyl]methanamine (1), (anthracen-9-yl)-N,N-bis[(pyridin-2-yl)methyl]methanamine (2), (pyreny-1-yl)-N,N-bis[(pyridin-2-yl)methyl]methanamine (3-5), and L' is catecholate (1-3), 4-tert-butyl catecholate (4), and 4-(2-aminoethyl)-benzene-1,2-diolate (5)--were synthesized and their photocytotoxic properties examined. The five electron-paramagnetic complexes displayed a Fe(III)/Fe(II) redox couple near -0.4 V versus a saturated calomel electrode (SCE) in DMF/0.1 M tetrabutylammonium perchlorate (TBAP). They showed unprecedented photocytotoxicity in red light (600-720 nm) to give IC50≈15 µM in various cell lines by means of apoptosis to generate reactive oxygen species. They were ingested in the nucleus of HeLa and HaCaT cells in 4 h, thereby interacting favorably with calf thymus (ct)-DNA and photocleaving pUC19 DNA in red light of 785 nm to form hydroxyl radicals.


Subject(s)
Catechols/chemistry , Color , DNA/chemistry , DNA/radiation effects , Ferrous Compounds/chemistry , Light , Molecular Imaging/methods , Photosensitizing Agents/chemistry , Animals , Apoptosis/drug effects , Cattle , Crystallography, X-Ray , DNA/drug effects , DNA Cleavage , Ferrous Compounds/chemical synthesis , Ferrous Compounds/pharmacology , HeLa Cells , Humans , Models, Molecular , Molecular Structure , Photochemical Processes , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacology , Plasmids/chemistry , Plasmids/drug effects , Plasmids/radiation effects , Quantum Theory , Reactive Oxygen Species/metabolism
13.
PLoS One ; 7(12): e51806, 2012.
Article in English | MEDLINE | ID: mdl-23284772

ABSTRACT

Oral submucous fibrosis (OSF) is a chronic inflammatory disease characterized by the accumulation of excess collagen, and areca nut chewing has been proposed as an important etiological factor for disease manifestation. Activation of transforming growth factor-ß signaling has been postulated as the main causative event for increased collagen production in OSF. Oral epithelium plays important roles in OSF, and arecoline has been shown to induce TGF-ß in epithelial cells. In an attempt to understand the role of areca nut constituents in the manifestation of OSF, we studied the global gene expression profile in epithelial cells (HaCaT) following treatment with areca nut water extract or TGF-ß. Interestingly, 64% of the differentially regulated genes by areca nut water extract matches with the TGF-ß induced gene expression profile. Out of these, expression of 57% of genes was compromised in the presence of ALK5 (TßRI) inhibitor and 7% were independently induced by areca nut, highlighting the importance of TGF-ß in areca nut actions. Areca nut water extract treatment induced p-SMAD2 and TGF-ß downstream targets in HaCaT cells but not in human gingival fibroblast cells (hGF), suggesting epithelial cells could be the source of TGF-ß in promoting OSF. Water extract of areca nut consists of polyphenols and alkaloids. Both polyphenol and alkaloid fractions of areca nut were able to induce TGF-ß signaling and its downstream targets. Also, SMAD-2 was phosphorylated following treatment of HaCaT cells by Catechin, Tannin and alkaloids namely Arecoline, Arecaidine and Guvacine. Moreover, both polyphenols and alkaloids induced TGF-ß2 and THBS1 (activator of latent TGF-ß) in HaCaT cells suggesting areca nut mediated activation of p-SMAD2 involves up-regulation and activation of TGF-ß. These data suggest a major causative role for TGF-ß that is induced by areca nut in OSF progression.


Subject(s)
Areca/adverse effects , Arecoline/analogs & derivatives , Biomarkers/metabolism , Nuts/adverse effects , Oral Submucous Fibrosis/etiology , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Arecoline/pharmacology , Cells, Cultured , Collagen/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gingiva/cytology , Gingiva/drug effects , Gingiva/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mastication , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Plant Extracts/pharmacology , Plants, Toxic/adverse effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/genetics , Transforming Growth Factor beta/genetics , Up-Regulation
14.
Indian J Microbiol ; 51(2): 242-3, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22654174
SELECTION OF CITATIONS
SEARCH DETAIL
...