29 research outputs found

    Functional analysis of genes dysregulated as a result of <i>hPNPase<sup>old-35</sup></i> overexpression.

    No full text
    <p>(A) The biological functions and states associated with genes differentially expressed when <i>hPNPase<sup>old-35</sup></i> is overexpressed in human melanoma cells. (B) Toxicologically related functionalities and pathways associated with genes dysregulated (proportions shown in graphs) after <i>hPNPase<sup>old-35</sup></i> overexpression in melanoma cells, as identified by IPA Toxicogenomic Analysis.</p

    Identification of Genes Potentially Regulated by Human Polynucleotide Phosphorylase (<i>hPNPase<sup>old-35</sup></i>) Using Melanoma as a Model

    Get PDF
    <div><p>Human Polynucleotide Phosphorylase (<i>hPNPase<sup>old-35</sup></i> or <i>PNPT1</i>) is an evolutionarily conserved 3′→5′ exoribonuclease implicated in the regulation of numerous physiological processes including maintenance of mitochondrial homeostasis, mtRNA import and aging-associated inflammation. From an RNase perspective, little is known about the RNA or miRNA species it targets for degradation or whose expression it regulates; except for <i>c-myc</i> and miR-221. To further elucidate the functional implications of <i>hPNPase<sup>old-35</sup></i> in cellular physiology, we knocked-down and overexpressed <i>hPNPase<sup>old-35</sup></i> in human melanoma cells and performed gene expression analyses to identify differentially expressed transcripts. Ingenuity Pathway Analysis indicated that knockdown of <i>hPNPase<sup>old-35</sup></i> resulted in significant gene expression changes associated with mitochondrial dysfunction and cholesterol biosynthesis; whereas overexpression of <i>hPNPase<sup>old-35</sup></i> caused global changes in cell-cycle related functions. Additionally, comparative gene expression analyses between our <i>hPNPase<sup>old-35</sup></i> knockdown and overexpression datasets allowed us to identify 77 potential “<i>direct”</i> and 61 potential “<i>indirect”</i> targets of <i>hPNPase<sup>old-35</sup></i> which formed correlated networks enriched for cell-cycle and wound healing functional association, respectively. These results provide a comprehensive database of genes responsive to <i>hPNPase<sup>old-35</sup></i> expression levels; along with the identification new potential candidate genes offering fresh insight into cellular pathways regulated by PNPT1 and which may be used in the future for possible therapeutic intervention in mitochondrial- or inflammation-associated disease phenotypes.</p></div

    Functional analysis of <i>hPNPase<sup>old-35</sup></i>-putative “<i>indirectly</i>” regulated genes.

    No full text
    <p>(A) The biological functions and states associated with <i>hPNPase<sup>old-35</sup></i>-putative “<i>indirectly</i>” regulated genes in human melanoma cells. (B) Toxicologically related functionalities and pathways associated with <i>hPNPase<sup>old-35</sup></i>-putative “<i>indirectly</i>” regulated genes, as identified by IPA Toxicogenomic Analysis.</p

    Real time qRT-PCR validation of microarray findings.

    No full text
    <p>(A) qRT-PCR verification of <i>hPNPase<sup>old-35</sup></i>-putative “<i>directly</i>” regulated genes identified by microarray analyses in response to <i>hPNPase<sup>old-35</sup></i> (i) knockdown or (ii) overexpression in HO-1 melanoma cells. (B) qRT-PCR verification of <i>hPNPase<sup>old-35</sup></i>-putative “<i>indirectly</i>” regulated genes identified by microarray analyses in response to <i>hPNPase<sup>old-35</sup></i> (i) knockdown or (ii) overexpression in HO-1 melanoma cells. Error bars represent mean ± S.E. error of three replicate experiments.</p

    Generation of a melanoma cell culture model for hPNPase<sup>old-35</sup> expression.

    No full text
    <p>(A) Phase contrast LM (top) and GFP fluorescent micrographs (bottom) of HO-1 melanoma cell lines following transduction with GFP expressing scrambled shRNA (HO-1 Csh) and <i>hPNPase<sup>old-35</sup></i> shRNA1 (shown in clone 4; cl4) and 2 (shown is clone 9; cl9) expressing lentiviruses and selection with puromycin. qRT-PCR expression of <i>hPNPase<sup>old-35</sup></i> (<i>hPNPase<sup>old-35</sup></i> knockdown) normalized to control (shScramble). Mean values normalized to a GAPDH internal reference; error bars represent mean ± S.E. of three replicate experiments. Anti-hPNPase<sup>old-35</sup> and EF1α loading control immunoblots. (B) qRT-PCR expression of <i>hPNPase<sup>old-35</sup></i> in HO-1 cells infected with Ad.<i>hPNPase<sup>old-35</sup></i> normalized to cells infected with Ad.<i>Vec</i> for 36 h. Immunoblot showing hPNPase<sup>old-35</sup> overexpression compared to Ad.<i>Vec</i> post 36 hour of infection. Error bars represent mean ± S.E of three replicate experiments. <i>* P<0.02</i>, <i>*** P<0.001</i>.</p

    Unravelling the Secrets of Mycobacterial Cidality through the Lens of Antisense

    No full text
    <div><p>One of the major impediments in anti-tubercular drug discovery is the lack of a robust grammar that governs the in-vitro to the in-vivo translation of efficacy. <i>Mycobacterium tuberculosis</i> (Mtb) is capable of growing both extracellular as well as intracellular; encountering various hostile conditions like acidic milieu, free radicals, starvation, oxygen deprivation, and immune effector mechanisms. Unique survival strategies of Mtb have prompted researchers to develop in-vitro equivalents to simulate in-vivo physiologies and exploited to find efficacious inhibitors against various phenotypes. Conventionally, the inhibitors are screened on Mtb under the conditions that are unrelated to the in-vivo disease environments. The present study was aimed to (1). Investigate cidality of Mtb targets using a non-chemical inhibitor antisense-RNA (AS-RNA) under in-vivo simulated in-vitro conditions.(2). Confirm the cidality of the targets under in-vivo in experimental tuberculosis. (3). Correlate in-vitro <i>vs</i>. in-vivo cidality data to identify the in-vitro condition that best predicts in-vivo cidality potential of the targets. Using cidality as a metric for efficacy, and AS-RNA as a target-specific inhibitor, we delineated the cidality potential of five target genes under six different physiological conditions (replicating, hypoxia, low pH, nutrient starvation, nitrogen depletion, and nitric oxide).In-vitro cidality confirmed in experimental tuberculosis in BALB/c mice using the AS-RNA allowed us to identify cidal targets in the rank order of <i>rpoB>aroK>ppk>rpoC>ilvB</i>. <i>RpoB</i> was used as the cidality control. In-vitro and in-vivo studies feature <i>aroK</i> (encoding shikimate kinase) as an in-vivo mycobactericidal target suitable for anti-TB drug discovery. In-vitro to in-vivo cidality correlations suggested the low pH (R = 0.9856) in-vitro model as best predictor of in-vivo cidality; however, similar correlation studies in pathologically relevant (Kramnik) mice are warranted. In the acute infection phase for the high fidelity translation, the compound efficacy may also be evaluated in the low pH, in addition to the standard replication condition.</p></div

    Functional analysis <i>hPNPase<sup>old-35</sup></i>-putative “<i>directly</i>” regulated genes.

    No full text
    <p>(A) The biological functions and states associated with <i>hPNPase<sup>old-35</sup></i>-putative “<i>directly</i>” regulated genes in human melanoma cells. (B) Toxicologically related functionalities and pathways associated with <i>hPNPase<sup>old-35</sup></i>-putative “<i>directly</i>” regulated genes, as identified by IPA Toxicogenomic Analysis.</p
    corecore