Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Cell Rep ; 42(7): 112823, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37463106

ABSTRACT

Cancers often display immune escape, but the mechanisms are incompletely understood. Herein, we identify SMYD3 as a mediator of immune escape in human papilloma virus (HPV)-negative head and neck squamous cell carcinoma (HNSCC), an aggressive disease with poor response to immunotherapy with pembrolizumab. SMYD3 depletion induces upregulation of multiple type I interferon (IFN) response and antigen presentation machinery genes in HNSCC cells. Mechanistically, SMYD3 binds to and regulates the transcription of UHRF1, encoding for a reader of H3K9me3, which binds to H3K9me3-enriched promoters of key immune-related genes, recruits DNMT1, and silences their expression. SMYD3 further maintains the repression of immune-related genes through intragenic deposition of H4K20me3. In vivo, Smyd3 depletion induces influx of CD8+ T cells and increases sensitivity to anti-programmed death 1 (PD-1) therapy. SMYD3 overexpression is associated with decreased CD8 T cell infiltration and poor response to neoadjuvant pembrolizumab. These data support combining SMYD3 depletion strategies with checkpoint blockade to overcome anti-PD-1 resistance in HPV-negative HNSCC.


Subject(s)
Head and Neck Neoplasms , Histone-Lysine N-Methyltransferase , Interferon Type I , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Humans , CCAAT-Enhancer-Binding Proteins , CD8-Positive T-Lymphocytes , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Histone-Lysine N-Methyltransferase/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Ubiquitin-Protein Ligases
3.
Cell Rep ; 42(6): 112587, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37294637

ABSTRACT

Embryonic expression of DNMT3B is critical for establishing de novo DNA methylation. This study uncovers the mechanism through which the promoter-associated long non-coding RNA (lncRNA) Dnmt3bas controls the induction and alternative splicing of Dnmt3b during embryonic stem cell (ESC) differentiation. Dnmt3bas recruits the PRC2 (polycomb repressive complex 2) at cis-regulatory elements of the Dnmt3b gene expressed at a basal level. Correspondingly, Dnmt3bas knockdown enhances Dnmt3b transcriptional induction, whereas overexpression of Dnmt3bas dampens it. Dnmt3b induction coincides with exon inclusion, switching the predominant isoform from the inactive Dnmt3b6 to the active Dnmt3b1. Intriguingly, overexpressing Dnmt3bas further enhances the Dnmt3b1:Dnmt3b6 ratio, attributed to its interaction with hnRNPL (heterogeneous nuclear ribonucleoprotein L), a splicing factor that promotes exon inclusion. Our data suggest that Dnmt3bas coordinates alternative splicing and transcriptional induction of Dnmt3b by facilitating the hnRNPL and RNA polymerase II (RNA Pol II) interaction at the Dnmt3b promoter. This dual mechanism precisely regulates the expression of catalytically active DNMT3B, ensuring fidelity and specificity of de novo DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methylation , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Embryonic Stem Cells/metabolism , Exons/genetics , Polycomb Repressive Complex 2/metabolism , Mice , DNA Methyltransferase 3B , Animals
4.
ACS Omega ; 7(23): 19687-19693, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35721984

ABSTRACT

Density functional theory calculations within the framework of generalized gradient approximation (GGA), meta-GGA, and local functionals were carried out to investigate the reactivity and catalytic activity of Ag n (n = 15-20) clusters. Our results reveal that all the Ag n clusters in this size range, except Ag20, adsorb O2 preferably in the bridged mode with enhanced binding energy as compared to the atop mode. The O2 binding energies range from 0.77 to 0.29 in the bridged mode and from 0.36 to 0.15 eV in the atop mode of O2 adsorption. The strong binding in the case of the bridged mode of O2 adsorption is also reflected in the increase in O-O bond distance. Natural bond orbital charge analysis and vibrational frequency calculations reveal that enhanced charge transfer occurs to the O2 molecule and there is significant red shift in the stretching frequency of O-O bond in the case of the bridged mode of O2 adsorption on the clusters, thereby confirming the above results. Moreover, the simulated CO oxidation reaction pathways show that the oxidation of the CO molecule is highly facile on Ag16 and Ag18 clusters involving small kinetic barriers and higher heats toward CO2 formation.

5.
J Cell Sci ; 135(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35686490

ABSTRACT

IGF1R is pursued as a therapeutic target because of its abnormal expression in various cancers. Recently, we reported the presence of a putative allosteric inhibitor binding pocket in IGF1R that could be exploited for developing novel anti-cancer agents. In this study, we examined the role of nine highly conserved residues surrounding this binding pocket, with the aim of screening compound libraries in order to develop small-molecule allosteric inhibitors of IGF1R. We generated GFP fusion constructs of these mutants to analyze their impact on subcellular localization, kinase activity and downstream signaling of IGF1R. K1055H and E1056G were seen to completely abrogate the kinase activity of IGF1R, whereas R1064K and L1065A were seen to significantly reduce IGF1R kinase activity. During molecular dynamics analysis, various structural and conformational changes were observed in different conserved regions of mutant proteins, particularly in the activation loop, compromising the kinase activity of IGF1R. These results show that a stretch of four discontinuous residues within this newly identified binding pocket is critical for the kinase activity and structural integrity of IGF1R. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Amino Acids , Receptor, IGF Type 1 , Amino Acids/metabolism , Cell Line, Tumor , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction
6.
Sci Rep ; 12(1): 7713, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35562179

ABSTRACT

The role of ER Ca2+ release via ryanodine receptors (RyR) in pancreatic ß-cell function is not well defined. Deletion of RyR2 from the rat insulinoma INS-1 (RyR2KO) enhanced IP3 receptor activity stimulated by 7.5 mM glucose, coincident with reduced levels of the protein IP3 Receptor Binding protein released with Inositol 1,4,5 Trisphosphate (IRBIT). Insulin content, basal (2.5 mM glucose) and 7.5 mM glucose-stimulated insulin secretion were reduced in RyR2KO and IRBITKO cells compared to controls. INS2 mRNA levels were reduced in both RyR2KO and IRBITKO cells, but INS1 mRNA levels were specifically decreased in RyR2KO cells. Nuclear localization of S-adenosylhomocysteinase (AHCY) was increased in RyR2KO and IRBITKO cells. DNA methylation of the INS1 and INS2 gene promotor regions was very low, and not different among RyR2KO, IRBITKO, and controls, but exon 2 of the INS1 and INS2 genes was more extensively methylated in RyR2KO and IRBITKO cells. Exploratory proteomic analysis revealed that deletion of RyR2 or IRBIT resulted in differential regulation of 314 and 137 proteins, respectively, with 41 in common. These results suggest that RyR2 regulates IRBIT levels and activity in INS-1 cells, and together maintain insulin content and secretion, and regulate the proteome, perhaps via DNA methylation.


Subject(s)
Insulinoma , Pancreatic Neoplasms , Animals , Cell Line , Glucose , Insulin/metabolism , Insulinoma/genetics , Pancreatic Neoplasms/genetics , Proteomics , RNA, Messenger , Rats , Ryanodine Receptor Calcium Release Channel/metabolism
7.
Protoplasma ; 258(5): 991-1007, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33629144

ABSTRACT

Squalene epoxidase (SQE) is a crucial regulatory enzyme for the biosynthesis of several important classes of compounds including sterols and triterpenoids. The present paper identified and characterised five SQE genes (GgSQE1 to GgSQE5) from Glycyrrhiza glabra through transcriptome data mining and homology-based cloning, for the first time. The phylogenetic analysis implied their functional divergence. The ORF corresponding to one of the five SQEs, namely, GgSQE1, was cloned and studied for its function in a heterologous system, following transient and stable expressions. The transient expression followed by GgSQE1 encoding protein purification suggested approximately 58.0-kDa protein following the predicted molecular mass of the deduced protein. The gene expression profiling based on qRT-PCR indicated its highest expression (6.4-folds) in the 10-month-old roots. Furthermore, ABA (12.4-folds) and GA3 (2.47) treatments upregulated the expression of GgSQE1 in the shoots after 10 and 12 hours, respectively, which was also reflected in glycyrrhizin accumulation. The inductive effects of ABA and GA3 over GgSQE1 expression were also confirmed through functional analysis of GgSQE1 promoters using GUS fusion construct. Stable constitutive expression of GgSQE1 in Nicotiana tabacum modulated the sterol contents. The study could pave the way for understanding the metabolic flux regulation concerning biosynthesis of related sterols and triterpenoids.


Subject(s)
Glycyrrhiza , Triterpenes , Glycyrrhiza/genetics , Phylogeny , Squalene Monooxygenase/genetics , Transcriptome/genetics
8.
Cell Rep ; 30(5): 1478-1490.e6, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023463

ABSTRACT

An aberrant increase in pluripotency gene (PpG) expression due to enhancer reactivation could induce stemness and enhance the tumorigenicity of cancer stem cells. Silencing of PpG enhancers (PpGe) during embryonic stem cell differentiation involves Lsd1-mediated H3K4me1 demethylation and DNA methylation. Here, we observed retention of H3K4me1 and DNA hypomethylation at PpGe associated with a partial repression of PpGs in F9 embryonal carcinoma cells (ECCs) post-differentiation. H3K4me1 demethylation in F9 ECCs could not be rescued by Lsd1 overexpression. Given our observation that H3K4me1 demethylation is accompanied by strong Oct4 repression in P19 ECCs, we tested if Oct4 interaction with Lsd1 affects its catalytic activity. Our data show a dose-dependent inhibition of Lsd1 activity by Oct4 and retention of H3K4me1 at PpGe in Oct4-overexpressing P19 ECCs. These data suggest that Lsd1-Oct4 interaction in cancer stem cells could establish a "primed" enhancer state that is susceptible to reactivation, leading to aberrant PpG expression.


Subject(s)
Enhancer Elements, Genetic , Histone Demethylases/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Biocatalysis , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin/metabolism , DNA Methylation/genetics , Epigenesis, Genetic , Histones/metabolism , Humans , Male , Models, Biological , Pluripotent Stem Cells/cytology
9.
Plant Physiol Biochem ; 147: 242-250, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31881433

ABSTRACT

Plant viruses infect various economically important crops and cause a serious threat to agriculture. As of now, conventional strategies employed are inadequate to circumvent the proliferation of rapidly evolving plant viruses. In this regard, recent advancement in genome-editing approach looks promising to produce plants resistant to DNA/RNA virus infections. Clustered regularly interspaced palindromic repeats (CRISPR) system has been emerged as a promising genome-editing tool that has received special interest because of its ease, competence and reproducibility. Recent studies have demonstrated that CRISPR/Cas9 system has great potential to confer plant immunity by either directly targeting or cleaving the viral genome in both RNA and DNA viruses. Similarly, the approach can be used for targeting the host susceptibility genes more particularly in case of RNA viruses. In the present review, different approaches and strategies being used to improve plant resistance against devastating viruses are discussed in view of recent advances in CRISPR systems. This review also describes the major pitfalls of CRISPR/Cas9 system that utilizes highly efficient and novel platforms to engineer interference to single and multiple plant RNA viruses.


Subject(s)
CRISPR-Cas Systems , Crops, Agricultural , Gene Editing , Genome, Plant , Plant Viruses , Crops, Agricultural/virology , Gene Editing/methods , Plant Viruses/physiology , Reproducibility of Results
10.
Cancer Chemother Pharmacol ; 84(3): 551-559, 2019 09.
Article in English | MEDLINE | ID: mdl-31129716

ABSTRACT

PURPOSE: Despite the fact that hyper-activation of Wnt/ß-catenin signaling pathway has been seen in many cancers, including liver, colorectal and lung carcinoma, no small molecule inhibitors are available that specifically target this pathway. In this study, we analyzed the impact of dinactin (DA), an antibiotic ionophore produced by Streptomyces species, as an effective small molecule targeting Wnt/ß-catenin signaling pathway in cancer cells. METHODS: We performed MTT assays to investigate cell viability and proliferation after exposure to small molecules. Protein expression analysis was carried out by western blotting. Top-Flash reporter assays were used to score for ß-catenin signaling and cell cycle analysis was carried out by flow cytometry. RESULTS: In the first set of experiments, DA was seen to selectively inhibit the proliferation of HCT-116 and HepG2 cancer cells, unlike HEK-293 cells (a low tumorigenic cell line), in apoptosis-independent manner. Further, DA was seen to block the G1/S progression and decrease the expression of cyclin D1 in cancer cells. Since cyclin D1 is the downstream target gene of Wnt/ß-catenin signaling, we examined the impact of DA on TCF-dependent ß-catenin activity using Top-Flash reporter assay. Interestingly, DA significantly decreased Top-Flash activity at lower nano-molar concentrations when compared with salinomycin in HCT-116 and HepG2 cells. CONCLUSION: We report the identification of dinactin as a natural product-based small molecule that effectively blocks the Wnt/ß-catenin signaling pathway in cancer cells at nano-molar concentration. We anticipate that DA could be developed as a novel drug for anti-cancer therapy and for the management of neuropathic pain.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Macrolides/pharmacology , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Wnt1 Protein/metabolism , beta Catenin/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , HCT116 Cells , Hep G2 Cells , Humans , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Wnt1 Protein/genetics , beta Catenin/genetics
11.
Genes (Basel) ; 10(5)2019 05 14.
Article in English | MEDLINE | ID: mdl-31091831

ABSTRACT

Despite a large body of evidence supporting the role of aberrant DNA methylation in etiology of several human diseases, the fundamental mechanisms that regulate the activity of mammalian DNA methyltransferases (DNMTs) are not fully understood. Recent advances in whole genome association studies have helped identify mutations and genetic alterations of DNMTs in various diseases that have a potential to affect the biological function and activity of these enzymes. Several of these mutations are germline-transmitted and associated with a number of hereditary disorders, which are potentially caused by aberrant DNA methylation patterns in the regulatory compartments of the genome. These hereditary disorders usually cause neurological dysfunction, growth defects, and inherited cancers. Biochemical and biological characterization of DNMT variants can reveal the molecular mechanism of these enzymes and give insights on their specific functions. In this review, we introduce roles and regulation of DNA methylation and DNMTs. We discuss DNMT mutations that are associated with rare diseases, the characterized effects of these mutations on enzyme activity and provide insights on their potential effects based on the known crystal structure of these proteins.


Subject(s)
DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Germ-Line Mutation/genetics , Animals , Cerebellar Ataxia/genetics , DNA , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Genome , Germ-Line Mutation/physiology , Hearing Loss, Sensorineural/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Humans , Mutation , Narcolepsy/genetics , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 29(9): 1043-1046, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30871771

ABSTRACT

Neuropathic pain is a debilitating form of treatment-resistant chronic pain caused by damage to the nervous system. Cannabinoids have been known for suppressing neuropathic pain by modulating the endo cannabinoid system. Since the canonical Wnt/ß-catenin signaling has recently been implicated in pain sensation, we investigated the impact of major cannabinoids (1-6) from the leaves of Cannabis sativa and an epoxy derivative of compound 2, here upon referred to as 2a, on modulating Wnt/ß-catenin signaling pathway. The results presented in this study show that compound 1, 2 and 2a exhibited potent inhibitory activity against Wnt/ß-catenin pathway in a dose-dependent manner. Compound 2a was seen to inhibit this pathway at slightly lower concentrations than its parent molecule 2, under similar conditions. Taken together, compound 1, 2 and 2a, by virtue of their inhibition of Wnt/ß-catenin signaling pathway, could be developed as effective neuroprotective agents for the management of neuropathic pain.


Subject(s)
Cannabinoids/chemistry , Wnt Signaling Pathway/drug effects , Animals , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cannabis/chemistry , Cannabis/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Neuralgia/drug therapy , Neuralgia/pathology , Plant Leaves/chemistry , Plant Leaves/metabolism
13.
Biochim Biophys Acta Mol Cell Res ; 1865(6): 920-931, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29621572

ABSTRACT

IGF1R is a ubiquitous receptor tyrosine kinase that plays critical roles in cell proliferation, growth and survival. Clinical studies have demonstrated upregulation of IGF1R mediated signaling in a number of malignancies including colon, breast, and lung cancers. Overexpression of the IGF1R in these malignancies is associated with a poor prognosis and overall survival. IGF1R specific kinase inhibitors have failed in multiple clinical trials partly because of the complex nature of IGF1R signaling. Thus identifying new binding partners and allosteric sites on IGF1R are emerging areas of research. More recently, IGF1R has been shown to translocate into the nucleus and perform many functions. In this study, we generated a library of IGF1R deletion and point mutants to examine IGF1R subcellular localization and activation of downstream signaling pathways. We show that the nuclear localization of IGF1R is primarily defined by its cytoplasmic domain. We identified a cross-talk between IGF1R and Wnt/ß-catenin signaling pathways and showed, for the first time, that IGF1R is associated with upregulation of TCF-mediated ß-catenin transcriptional activity. Using loss-of-function mutants, deletion analysis and IGF1R specific inhibitor(s), we show that cytoplasmic and nuclear activities are two independent functions of IGF1R. Furthermore, we identified a unique loss-of-function mutation in IGF1R. This unique loss-of-function mutant retains only nuclear functions and sits in a pocket, outside ATP and substrate binding region, that is suited for designing allosteric inhibitors of IGF1R.


Subject(s)
Loss of Function Mutation , Receptors, Somatomedin , Up-Regulation/physiology , Wnt Signaling Pathway/physiology , HEK293 Cells , Hep G2 Cells , Humans , Protein Domains , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
Microbiol Res ; 207: 196-202, 2018 03.
Article in English | MEDLINE | ID: mdl-29458855

ABSTRACT

A highly active actinobacterial strain isolated from untapped areas of Northwestern Himalayas and characterised as Streptomyces puniceus strain AS13 by 16S rRNA gene sequencing was selected for production of bioactive metabolites. The bioassay-guided fractionation of microbial cultured ethyl acetate extract of the strain, led to isolation of macrotetrolide compound 1 (Dinactin) and compound 2 (1-(2,4-dihydroxy-6-methylphenyl)-ethanone). Structures of the isolated compounds were elucidated by [corrected] interpretation of NMR and other spectroscopic data including HR-ESI-MS, FT-IR. These compounds are reported for first time from Streptomyces Puniceus. Compound 1 exhibited strong anti-microbial activity against all tested bacterial pathogens including Mycobacterium tuberculosis. The MIC values of compound 1 against Gram negative and Gram positive bacterial pathogens ranged between 0.019 - 0.156µgml-1 and 1µgml-1 against Mycobacterium tuberculosis H37Rv. Dinactin exhibited marked anti-tumor potential with IC50 of 1.1- 9.7µM in various human cancerous cell lines and showed least cytotoxicity (IC50∼80µM) in normal cells (HEK-293). Dinactin inhabited cellular proliferation in cancer cells, reduced their clonogenic survival as validated by clonogenic assay and also inhabited cell migration and invasion characteristics in colon cancer (HCT-116) cells. Our results expressed the antimicrobial potential of dinactin and also spotted its prospective as an antitumor antibiotic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Macrolides/pharmacology , Mycobacterium tuberculosis/drug effects , Streptomyces/metabolism , Anti-Bacterial Agents/metabolism , Antineoplastic Agents/metabolism , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Humans , Macrolides/metabolism , Microbial Sensitivity Tests , RNA, Ribosomal, 16S/genetics , Streptomyces/classification , Streptomyces/genetics
15.
Mutat Res Rev Mutat Res ; 773: 122-133, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28927523

ABSTRACT

The Wnt/ß-catenin is a highly conserved signaling pathway involved in cell fate decisions during various stages of development. Dysregulation of canonical Wnt/ß-catenin signaling has been associated with various diseases including cancer. ß-Catenin, the central component of canonical Wnt signaling pathway, is a multi-functional protein playing both structural and signaling roles. ß-Catenin is composed of three distinct domains: N-terminal domain, C-terminal domain and a central armadillo repeat domain. N-terminal domain of ß-catenin harbours almost all of the cancer causing mutations, thus deciphering its critical structural and functional roles offers great potential in cancer detection and therapy. Here, in this review, we have collected information from pharmacological analysis, bio-physical and structural studies, molecular modeling, in-vivo and in-vitro assays, and transgenic animal experiments employing various N-terminal domain variants of ß-catenin to discuss the interaction of ß-catenin with its binding partners that specifically interact with this domain and the implications of these interactions on signaling, cell fate determination, and in tumorigenesis. A thorough understanding of interactions between ß-catenin and its binding partners will enable us to more effectively understand how ß-catenin switches between its multiple roles, and will lead to the development of specific assays for the identification of small molecules as chemotherapeutic agents to treat diseases, including cancer and neurological disorders, where Wnt/ß-catenin signaling is dysregulated.


Subject(s)
Neoplasms/genetics , beta Catenin/genetics , beta Catenin/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cell Line, Tumor , Humans , Models, Molecular , Mutation, Missense , Point Mutation , Protein Conformation , Wnt Signaling Pathway
16.
FEBS Lett ; 590(18): 3071-82, 2016 09.
Article in English | MEDLINE | ID: mdl-27552098

ABSTRACT

Class-1 phosphatidylinositol-3-kinases (PI3Ks) are activated by a variety of extracellular stimuli and have been implicated in a wide range of cellular processes. p110α and p110ß are the two most studied isoforms of the class-1A PI3K signaling pathway. Although these two isoforms are ubiquitously expressed and play multiple redundant roles, they also have distinct functions within the cell. More recently, p110α and p110ß isoforms have been shown to translocate into the nucleus and play a role in DNA replication and repair, and in cell cycle progression. In the following Review article, we discuss the overlapping and unique roles of p110α and p110ß isoforms with a particular focus on their structure, expression analysis, subcellular localization, and signaling contributions in various cell types and model organisms.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Signal Transduction , Animals , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Humans , Phosphoinositide-3 Kinase Inhibitors , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport
17.
Biochim Biophys Acta ; 1863(9): 2345-57, 2016 09.
Article in English | MEDLINE | ID: mdl-27368802

ABSTRACT

ß-Catenin, the central molecule of canonical Wnt signaling pathway, has multiple binding partners and performs many roles in the cell. Apart from being a transcriptional activator, ß-catenin acts as a crucial effector component of cadherin/catenin complex to physically interact with actin cytoskeleton along with α-catenin and E-cadherin for regulating cell-cell adhesion. Here, we have generated a library of ß-catenin point and deletion mutants to delineate regions within ß-catenin that are important for α-catenin-ß-catenin interaction, nuclear localization, and transcriptional activity of ß-catenin. We observed a unique mechanism for nuclear localization of ß-catenin and its mutants and show that N-terminal exon-3 region and C-terminal domain of ß-catenin are critical for this activity of ß-catenin. Furthermore, we show HepG2 cells have high ß-catenin mediated transcriptional activity due to the presence of an interstitial deletion at the N-terminal region of ß-catenin. Due to this deletion mutant (hereupon called TM), GSK3ß and HDAC inhibitors failed to show any impact whereas curcumin significantly inhibited ß-catenin mediated transcriptional activity reiterating that TM is primarily responsible for the high transcriptional activity of HepG2 cells. Moreover, we show the recombinant TM does not physically interact with α-catenin, localizes predominantly in the nucleus, and has nearly two-fold higher transcriptional activity than the wildtype ß-catenin.


Subject(s)
Transcription, Genetic , beta Catenin/chemistry , beta Catenin/metabolism , Cell Adhesion/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Computer Simulation , Curcumin/pharmacology , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Models, Biological , Protein Binding/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Sequence Deletion , Structure-Activity Relationship , Subcellular Fractions/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , alpha Catenin/metabolism
18.
J Cell Biochem ; 117(12): 2864-2874, 2016 12.
Article in English | MEDLINE | ID: mdl-27167447

ABSTRACT

Cancer therapeutics is a hot subject and PI3K class 1A isoforms (p110α and p110ß) are pursued as major targets. Genetic analysis, biochemical approaches, and structural studies have demonstrated crucial roles for these isoforms in several physiological processes. p110α is critical for insulin signaling, whereas p110ß is essential for the growth and differs from p110α in many ways. Here, we have generated GFP-fusion clones of wildtype and mutant version of p110α and p110ß and expressed them in HEK293 and cancer cells to examine their subcellular localization and their impact on downstream signaling. In HEK293 cells, p110ß GFP-fusion protein is translocated into the nucleus, whereas p110α-GFP stays exclusively in the cytoplasm. This study demonstrates that p110α and p110ß oncogenecity, kinase activity, and interaction with p85 regulatory subunit does not have any impact on their subcellular localization. PI3K pathway specific inhibitor, LY294002, abrogated PI3K signaling by reducing pAkt levels, however, the subcellular localization of p110α and p110ß remained unchanged. Furthermore, we analyzed the expression of recombinant p110α and p110ß in a panel of human cancer cells and observed remarkable differences in their expression levels. The differential expression of recombinant p110α and p110ß was observed to be mainly regulated by the endogenous levels of pAkt. Unlike in HEK293, p110α showed nuclear localization in cancer cells in a similar fashion to p110ß. Moreover, we observed the PI3K signaling activities in low pAkt expressing cells are mediated by PDK1 and S6K proteins. Finally, p110α and p110ß were seen to play an essential role in promoting the cell cycle progression in MCF-7 and HCT-116 cells. J. Cell. Biochem. 117: 2864-2874, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Green Fluorescent Proteins/metabolism , Neoplasms/metabolism , Recombinant Fusion Proteins/metabolism , Apoptosis , Blotting, Western , Cell Cycle , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cells, Cultured , HEK293 Cells , Humans , Neoplasms/pathology , Phosphorylation , Protein Isoforms , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...