10 research outputs found

    Computerized clinical decision support systems for acute care management: A decision-maker-researcher partnership systematic review of effects on process of care and patient outcomes

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Acute medical care often demands timely, accurate decisions in complex situations. Computerized clinical decision support systems (CCDSSs) have many features that could help. However, as for any medical intervention, claims that CCDSSs improve care processes and patient outcomes need to be rigorously assessed. The objective of this review was to systematically review the effects of CCDSSs on process of care and patient outcomes for acute medical care.</p> <p>Methods</p> <p>We conducted a decision-maker-researcher partnership systematic review. MEDLINE, EMBASE, Evidence-Based Medicine Reviews databases (Cochrane Database of Systematic Reviews, DARE, ACP Journal Club, and others), and the Inspec bibliographic database were searched to January 2010, in all languages, for randomized controlled trials (RCTs) of CCDSSs in all clinical areas. We included RCTs that evaluated the effect on process of care or patient outcomes of a CCDSS used for acute medical care compared with care provided without a CCDSS. A study was considered to have a positive effect (<it>i.e.</it>, CCDSS showed improvement) if at least 50% of the relevant study outcomes were statistically significantly positive.</p> <p>Results</p> <p>Thirty-six studies met our inclusion criteria for acute medical care. The CCDSS improved process of care in 63% (22/35) of studies, including 64% (9/14) of medication dosing assistants, 82% (9/11) of management assistants using alerts/reminders, 38% (3/8) of management assistants using guidelines/algorithms, and 67% (2/3) of diagnostic assistants. Twenty studies evaluated patient outcomes, of which three (15%) reported improvements, all of which were medication dosing assistants.</p> <p>Conclusion</p> <p>The majority of CCDSSs demonstrated improvements in process of care, but patient outcomes were less likely to be evaluated and far less likely to show positive results.</p

    Nek5 promotes centrosome integrity in interphase and loss of centrosome cohesion in mitosis

    No full text
    Nek5 is a poorly characterized member of the NIMA-related kinase family, other members of which play roles in cell cycle progression and primary cilia function. Here, we show that Nek5, similar to Nek2, localizes to the proximal ends of centrioles. Depletion of Nek5 or overexpression of kinase-inactive Nek5 caused unscheduled separation of centrosomes in interphase, a phenotype also observed upon overexpression of active Nek2. However, separated centrosomes that resulted from Nek5 depletion remained relatively close together, exhibited excess recruitment of the centrosome linker protein rootletin, and had reduced levels of Nek2. In addition, Nek5 depletion led to loss of PCM components, including gamma-tubulin, pericentrin, and Cdk5Rap2, with centrosomes exhibiting reduced microtubule nucleation. Upon mitotic entry, Nek5-depleted cells inappropriately retained centrosome linker components and exhibited delayed centrosome separation and defective chromosome segregation. Hence, Nek5 is required for the loss of centrosome linker proteins and enhanced microtubule nucleation that lead to timely centrosome separation and bipolar spindle formation in mitosis

    Nek11S is required for G2/M checkpoint arrest and cell survival.

    No full text
    <p><b>A-L.</b> Using the protocols described in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0140975#pone.0140975.g001" target="_blank">Fig 1A</a> for irradiation and <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0140975#pone.0140975.g003" target="_blank">Fig 3A</a> for irinotecan treatment, HCT116 WT (A-C, G-I) and HCT116 p53-null (D-F, J-L) cells were transfected with siRNA oligonucleotides to co-deplete Nek11L and Nek11D (L/D), or deplete Nek11S or luciferase (siGL2). Histograms show the percentage of cycling cells at G2/M (A-F) and of total cells with sub-2n DNA (G-L). <i>p</i> values are relative to siGL2 for each treatment.</p

    Nek5 promotes centrosome integrity in interphase and loss of centrosome cohesion in mitosis

    Full text link
    Nek5 is a poorly characterized member of the NIMA-related kinase family, other members of which play roles in cell cycle progression and primary cilia function. Here, we show that Nek5, similar to Nek2, localizes to the proximal ends of centrioles. Depletion of Nek5 or overexpression of kinase-inactive Nek5 caused unscheduled separation of centrosomes in interphase, a phenotype also observed upon overexpression of active Nek2. However, separated centrosomes that resulted from Nek5 depletion remained relatively close together, exhibited excess recruitment of the centrosome linker protein rootletin, and had reduced levels of Nek2. In addition, Nek5 depletion led to loss of PCM components, including γ-tubulin, pericentrin, and Cdk5Rap2, with centrosomes exhibiting reduced microtubule nucleation. Upon mitotic entry, Nek5-depleted cells inappropriately retained centrosome linker components and exhibited delayed centrosome separation and defective chromosome segregation. Hence, Nek5 is required for the loss of centrosome linker proteins and enhanced microtubule nucleation that lead to timely centrosome separation and bipolar spindle formation in mitosis

    Mapping of regions in Nek11 required for nuclear import and export.

    No full text
    <p><b>A.</b> Schematic representation of GFP-Nek11L constructs used to examine subcellular localisation. Predominant localisation to cytoplasm (C), nucleus (N) or equal distribution (C/N) ±LMB treatment is indicated. <b>B.</b> Western blots with GFP and α-tubulin antibodies of lysates prepared from U2OS cells transiently transfected for 24 hours with the Nek11L constructs indicated. Kinase domain includes residues 1–287 and C-terminal domain includes residues 288–645. M. wts (kDa) are indicated on the left. <b>C</b>. U2OS cells were transfected with constructs indicated and, after 24 hours, treated ±LMB for 3 hours before fixation and staining with GFP antibodies. <b>D & E</b>. Western blots and immunofluorescence staining was performed as in B and C, respectively, with the constructs indicated. Scale bars, 5 μm.</p

    Irinotecan induces Nek11-dependent G2/M arrest in HCT116 cells.

    No full text
    <p><b>A.</b> Schematic representation of time-course for cell treatments. 24 hours after seeding, cells were transfected with siRNA oligonucleotides. 52 hours later, cells were either untreated or treated with 5 μM irinotecan. They were then collected and fixed for PI-based flow cytometry after a further 20 hours. <b>B & C</b>. Following the protocol described in A, HCT116 WT and p53-null cells were transfected with siRNAs as indicated and left untreated (B) or treated with irinotecan (C), before analysis by flow cytometry. Distribution of cells according to flow cytometry profile is indicated (2n, G1; 2n-4n, S; 4n, G2/M). <b>D-G.</b> Histograms represent percentage of cycling HCT116 WT (D, E) and p53-null (F, G) cells at G2/M. <b>H-K.</b> Histograms show the percentage of HCT116 WT (H, I) and p53-null (J, K) cells with sub-2n DNA. Histograms in D-K are based on data in B and C. <i>p</i> values are relative to siGL2.</p

    Nek11 depletion leads to apoptosis and loss of cell viability.

    No full text
    <p><b>A-D.</b> Following the protocol described in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0140975#pone.0140975.g001" target="_blank">Fig 1A</a>, HCT116 WT (A, B) and p53-null (C, D) cells were transfected with siRNAs as indicated, before irradiation and analysis by annexin V-based flow cytometry. Histograms represent the percentage of all cells in apoptosis. <i>p</i> values are relative to siGL2. <b>E.</b> HCT116 WT and p53-null cells were transfected with siRNAs as indicated and after 56 hours treated ±2 Gy IR. Cells were collected after a further 16 hours and plated for clonogenic assays (50 cells per plate for siGL2, 500 cells per plate for other treatments). Colonies were stained with crystal violet. <b>F.</b> Colonies from E were counted and % survival determined as described in Materials and Methods. <b>G.</b> HCT116 p53-null cells were transfected with either siGL2 or siNek11-2 and after 56 hours either left untreated or irradiated. Cells were fixed 48 hours post IR and stained with α-tubulin antibody (green). DNA was stained with Hoechst to observe nuclear morphology. Scale bars, 10 um. <b>H.</b> Histogram represents the percentage of cells exhibiting multiple nuclei following the treatments indicated as described in G. 500 cells were counted per sample and the average percentage of multinuclear cells was determined across two independent experiments.</p

    Identification by high-throughput screening of viridin analogs as biochemical and cell-based inhibitors of the cell cycle-regulated Nek2 kinase

    Full text link
    Nek2 is a serine/threonine protein kinase that localizes to the centrosome and is implicated in mitotic regulation. Overexpression of Nek2 induces premature centrosome separation and nuclear defects indicative of mitotic errors, whereas depletion of Nek2 interferes with cell growth. As Nek2 expression is upregulated in a range of cancer cell lines and primary human tumors, inhibitors of Nek2 may have therapeutic value in cancer treatment. The authors used a radiometric proximity assay in a high-throughput screen to identify small-molecule inhibitors of Nek2 kinase activity. The assay was based on the measurement of the radiolabeled phosphorylated product of the kinase reaction brought into contact with the surface of wells of solid scintillant-coated microplates. Seventy nonaggregating hits were identified from approximately 73,000 compounds screened and included a number of toxoflavins and a series of viridin/wortmannin-like compounds. The viridin-like compounds were >70-fold selective for Nek2 over Nek6 and Nek7 and inhibited the growth of human tumor cell lines at concentrations consistent with their biochemical potencies. An automated mechanism-based microscopy assay in which centrosomes were visualized using pericentrin antibodies confirmed that 2 of the viridin inhibitors reduced centrosome separation in a human tumor cell line. The data presented show that pharmacological inhibition of Nek2 kinase results in the expected phenotype of disruption to centrosome function associated with growth inhibition and further supports Nek2 as a target for cancer drug discovery
    corecore