100 research outputs found

    Significance of CD44 and CD24 as Cancer Stem Cell Markers: An Enduring Ambiguity

    Get PDF
    Cancer stem cell population is a subset of cells capable of dictating invasion, metastasis, heterogeneity, and therapeutic resistance in tumours. Eradication of this rare population is a new insight in cancer treatment. However, prospective identification, characterization, and isolation of these CSCs have been a major challenge. Many studies were performed on surface markers for potential identification and isolation of CSCs. Lack of universal expression of surface markers limits their usage and no best combination of markers has yet been confirmed to identify CSCs capable of initiating and metastasizing tumours. CD44, a hyaluronic acid receptor, is one of the most commonly studied surface markers, which is expressed by almost every tumour cell. CD24, a heat stable antigen, is another surface marker expressed in many tumour types. However, their expression and prognostic value in isolating CSCs are still an enduring ambiguity. In this critical review, we assess the role of CD44 and CD24 in tumour initiation, development, and metastasis. We mainly focus on analysing the significance of CD44 and CD24 as CSC surface markers in combination or with other putative markers in different types of cancer

    Advanced Glycation End-Products Can Activate or Block Bitter Taste Receptors

    No full text
    Bitter taste receptors (T2Rs) are expressed in several tissues of the body and are involved in a variety of roles apart from bitter taste perception. Advanced glycation end-products (AGEs) are produced by glycation of amino acids in proteins. There are varying sources of AGEs, including dietary food products, as well as endogenous reactions within our body. Whether these AGEs are T2R ligands remains to be characterized. In this study, we selected two AGEs, namely, glyoxal-derived lysine dimer (GOLD) and carboxymethyllysine (CML), based on their predicted interaction with the well-studied T2R4, and its physiochemical properties. Results showed predicted binding affinities (Kd) for GOLD and CML towards T2R4 in the nM and μM range, respectively. Calcium mobilization assays showed that GOLD inhibited quinine activation of T2R4 with IC50 10.52 ± 4.7 μM, whilst CML was less effective with IC50 32.62 ± 9.5 μM. To characterize whether this antagonism was specific to quinine activated T2R4 or applicable to other T2Rs, we selected T2R14 and T2R20, which are expressed at significant levels in different human tissues. A similar effect of GOLD was observed with T2R14; and in contrast, GOLD and CML activated T2R20 with an EC50 of 79.35 ± 29.16 μM and 65.31 ± 17.79 μM, respectively. In this study, we identified AGEs as novel T2R ligands that caused either activation or inhibition of different T2Rs

    Reply: A morpho-molecular prognostic model for hepatocellular carcinoma

    Get PDF
    10.1038/bjc.2012.515British Journal of Cancer1083741-BJCA

    The Pharmacochaperone Activity of Quinine on Bitter Taste Receptors.

    No full text
    Bitter taste is one of the five basic taste sensations which is mediated by 25 bitter taste receptors (T2Rs) in humans. The mechanism of bitter taste signal transduction is not yet elucidated. The cellular processes underlying T2R desensitization including receptor internalization, trafficking and degradation are yet to be studied. Here, using a combination of molecular and pharmacological techniques we show that T2R4 is not internalized upon agonist treatment. Pretreatment with bitter agonist quinine led to a reduction in subsequent quinine-mediated calcium responses to 35 ± 5% compared to the control untreated cells. Interestingly, treatment with different bitter agonists did not cause internalization of T2R4. Instead, quinine treatment led to a 2-fold increase in T2R4 cell surface expression which was sensitive to Brefeldin A, suggesting a novel pharmacochaperone activity of quinine. This phenomenon of chaperone activity of quinine was also observed for T2R7, T2R10, T2R39 and T2R46. Our results suggest that the observed action of quinine for these T2Rs is independent of its agonist activity. This study provides novel insights into the pharmacochaperone activity of quinine and possible mechanism of T2R desensitization, which is of fundamental importance in understanding the mechanism of bitter taste signal transduction

    Abscisic acid acts as a blocker of the bitter taste G protein-coupled receptor T2R4

    No full text
    Bitter taste receptors (T2Rs) belong to the G protein-coupled receptor superfamily. In humans, 25 T2Rs mediate bitter taste sensation. In addition to the oral cavity, T2Rs are expressed in many extraoral tissues, including the central nervous system, respiratory system, and reproductive system. To understand the mechanistic roles of the T2Rs in oral and extraoral tissues, novel blockers or antagonists are urgently needed. Recently, we elucidated the binding pocket of T2R4 for its agonist quinine, and an antagonist and inhibitory neurotransmitter, \u3b3-aminobutyric acid. This structure 12function information about T2R4 led us to screen the plant hormone abscisic acid (ABA), its precursor (xanthoxin), and catabolite phaseic acid for their ability to bind and activate or inhibit T2R4. Molecular docking studies followed by functional assays involving calcium imaging confirmed that ABA is an antagonist with an IC50 value of 34.4 \ub1 1.1 \u3bcM. However, ABA precursor xanthoxin acts as an agonist on T2R4. Interestingly, molecular model-guided site-directed mutagenesis suggests that the T2R4 residues involved in quinine binding are also predominantly involved in binding to the novel antagonist, ABA. The antagonist ability of ABA was tested using another T2R4 agonist, yohimbine. Our results suggest that ABA does not inhibit yohimbineinduced T2R4 activity. The discovery of natural bitter blockers has immense nutraceutical and physiological significance and will help in dissecting the T2R molecular pathways in various tissues.Peer reviewed: YesNRC publication: Ye

    Characterization of the Binding Sites for Bacterial Acyl Homoserine Lactones (AHLs) on Human Bitter Taste Receptors (T2Rs)

    No full text
    The 25 bitter taste receptors (T2Rs) in humans are novel players in mediating host–pathogen responses in the airways and innate immunity. The chemosensory T2Rs are expressed in different extraoral tissues and perform diverse pathophysiological roles from mediating bronchodilation to detecting bacterial infection in the airways. T2Rs were suggested to be activated by multiple bacterial quorum sensing molecules (QSMs). However, whether bacterial QSMs bind to T2Rs and the structural features on T2Rs has not yet been characterized. Here, we analyzed the taste sensory profiles of QSMs including acyl homoserine lactones (C4-AHL, C8-AHL, and 3-oxo-C12-AHL) and hydro­xy­quino­lones (HHQ and NHQ) predominantly secreted by Gram-negative bacteria and characterized the candidate T2Rs interacting with different QSMs using structure–function approaches. The potency of the above QSMs for T2Rs significantly expressed in the airways, namely T2R4, T2R14, and T2R20, was characterized. 3-Oxo-C12-AHL activated T2R4, T2R14, and T2R20, while C8-AHL activated T2R4 and T2R14 with strong potency. The T2R amino acid residues involved in the interactions were characterized by molecular-model-guided site-directed mutagenesis. AHLs bind to a similar orthosteric site present on the extracellular surface in all three T2Rs with significant contributions from residues in extracellular loop 2. Our results reveal the mode of binding of AHLs for different T2Rs and provide biochemical insights into their interactions. This study will facilitate mechanistic studies aimed at understanding the role of these T2Rs as “sensors” of bacteria and in host–pathogen interactions

    Agonist promoted desensitization of T2R4 expressed in HEK293T cells.

    No full text
    <p><b>[A]</b>. Representative calcium traces of T2R4-expressing HEK293T cells pretreated with assay buffer, quinine or yohimbine. <b>[B]</b>. Representative calcium traces of T2R4-expressing HEK293T cells pretreated with assay buffer or yohimbine. Fluo-4 NW loaded cells were pretreated with buffer alone or 1 mM each of quinine or yohimbine for 15 min, washed with assay buffer, and exposed to same concentrations of the agonist by the Flexstation 3 microplate reader. Changes in intracellular calcium were measured in terms of relative fluorescence units (RFU) at 525 nm following excitation at 494 nm. The arrow at 20 sec indicates the addition of agonist by Flexstation 3 microplate reader. <b>[C]</b>. Bar plot representation of the measured intracellular calcium. Buffer pretreated T2R4-expressing cells stimulated with quinine were represented as 100%, and calcium responses of other treatments were normalized to it. Data is representative of four independent experiments and expressed as mean ± SEM. Significance was checked by one-way ANOVA using <i>Dunnette’s</i> post hoc test, *p<0.05, ***p<0.001.</p
    corecore