25 research outputs found

    Operational Techniques for Dealing with Long Eclipses During the MMS Extended Mission

    Get PDF
    Launch window design for the Magnetospheric Multiscale (MMS) mission ensured that no excessive eclipses would be encountered during the prime mission. However, no orbit solutions exist that satisfy the eclipse constraints indefinitely: most extended mission years contain 1-3 eclipses long enough to potentially damage either the spacecraft or its scientific instruments. Two steps were taken to improve the situation. Firstly, raising apogee radius from 25 to 29.34 Earth radii altered the Sun-Earth-MMS phasing, so efficiently achieving reductions in the long eclipse durations. These maneuvers were performed early this year, in preparation for the first pair of long eclipses in August 2019. Secondly, a set of operational steps were taken around the time of the eclipses to help maintain spacecraft and instrument temperatures while preventing power load shedding. These operational steps included raising key onboard temperatures through adjusting the spacecraft attitude to tilt the instrument deck towards the Sun, and engaging select heaters prior to going into eclipses. In addition, all scientific instruments were turned off, as well as high-power, non-critical spacecraft systems, to conserve energy.These steps each came with trade-offs which will be discussed in the paper. Finally, the results that were obtained when the spacecraft experienced the first extremely long eclipses will be discussed, as will lessons learned for future long eclipses

    Retrospective evaluation of whole exome and genome mutation calls in 746 cancer samples

    No full text
    Funder: NCI U24CA211006Abstract: The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) curated consensus somatic mutation calls using whole exome sequencing (WES) and whole genome sequencing (WGS), respectively. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2,658 cancers across 38 tumour types, we compare WES and WGS side-by-side from 746 TCGA samples, finding that ~80% of mutations overlap in covered exonic regions. We estimate that low variant allele fraction (VAF < 15%) and clonal heterogeneity contribute up to 68% of private WGS mutations and 71% of private WES mutations. We observe that ~30% of private WGS mutations trace to mutations identified by a single variant caller in WES consensus efforts. WGS captures both ~50% more variation in exonic regions and un-observed mutations in loci with variable GC-content. Together, our analysis highlights technological divergences between two reproducible somatic variant detection efforts

    Preventing Alcohol-Exposed Pregnancies among Hispanic Women

    No full text
    Project Healthy CHOICES, a self-administered, mail-based prevention intervention, was developed for women at risk of an alcohol-exposed pregnancy (AEP). Participants were sent their assessment and study materials through the United States Postal Service. This article uses data from a larger study (N = 354) and focuses on the 89 women who identified as Hispanic. Potential participants who called in response to English and Spanish ads and who said they could read and write Spanish were given a choice of receiving the intervention materials in English or Spanish. The main objective of the present study was to evaluate differences in outcomes as a function of (a) the language in which the intervention materials were received, and (b) the participants’ acculturation levels. Prior to the study, all women were at risk of an AEP. At the 6-month follow-up, two thirds (66%) of all Hispanic women had reduced their overall risk of an AEP, primarily by practicing effective birth control. These outcomes are similar to those reported for previous Project CHOICES studies. Significantly more women who requested the intervention materials in English (75%) compared to Spanish (41%) reduced their overall risk of an AEP. Women with high English cultural domain scores were at significantly less risk of an AEP due to effective contraception and a reduced overall risk of an AEP. Compared to other Project CHOICES studies, Project Healthy CHOICES is less intensive; it is self-administered, freely available, and can be completed without visiting a health care practitioner or clinic

    An N-ethyl-N-nitrosourea (ENU)-induced dominant negative mutation in the JAK3 kinase protects against cerebral malaria.

    Get PDF
    Cerebral malaria (CM) is a lethal neurological complication of malaria. We implemented a genome-wide screen in mutagenized mice to identify host proteins involved in CM pathogenesis and whose inhibition may be of therapeutic value. One pedigree (P48) segregated a resistance trait whose CM-protective effect was fully penetrant, mapped to chromosome 8, and identified by genome sequencing as homozygosity for a mis-sense mutation (W81R) in the FERM domain of Janus-associated kinase 3 (Jak3). The causative effect of Jak3(W81R) was verified by complementation testing in Jak3(W81R/-) double heterozygotes that were fully protected against CM. Jak3(W81R) homozygotes showed defects in thymic development with depletion of CD8(+) T cell, B cell, and NK cell compartments, and defective T cell-dependent production of IFN-γ. Adoptive transfer of normal splenocytes abrogates CM resistance in Jak3(W81R) homozygotes, an effect attributed to the CD8(+) T cells. Jak3(W81R) behaves as a dominant negative variant, with significant CM resistance of Jak3(W81R/+) heterozygotes, compared to CM-susceptible Jak3(+/+) and Jak3(+/-) controls. CM resistance in Jak3(W81R/+) heterozygotes occurs in presence of normal T, B and NK cell numbers. These findings highlight the pathological role of CD8(+) T cells and Jak3-dependent IFN-γ-mediated Th1 responses in CM pathogenesis

    Resistance to cerebral malaria in pedigree 48 maps to the central portion of chromosome 8.

    No full text
    <p>Genome-wide linkage analysis of the CM-resistance trait (survival) was conducted in 44 G3 (15 resistant, 29 susceptible) mice from pedigree 48, using polymorphic markers informative for the B6 and B10 progenitors. (<b>A</b>) LOD score traces identifying significant linkage to chromosome 8 (p = 0.05, genome-wide significance shown as dotted line); the position of informative markers is shown, including rs33080067 and rs32729089 (LOD score ∼5.8). (<b>B</b>) Haplotype analysis of the central portion of chromosome 8 in CM-resistant and CM-susceptible G3 animals from pedigree 48 (A, B6 homozygotes; H, B6/B10 heterozygotes; B, B10 homozygotes) showing exclusion of homozygote B6 haplotypes from the CM-susceptible group. The positions of the markers (in Mb) from the centromere (shown as a grey dot) are shown. (<b>C</b>) The G1 male from pedigree 48 was out-crossed to 129S1/SvlmJ to generate an F2 population (n = 211) that was phenotyped for response to <i>P. berghei</i> ANKA infection. Survival of F2 mice as well as parental 129S1/SvlmJ and C57BL/10J controls is shown.</p

    The <i>Jak3<sup>W81R</sup></i> mutation confers susceptibility to infection with different bacterial pathogens.

    No full text
    <p>(<b>A</b>) Control (C57BL/6J) and <i>Jak3<sup>W81R</sup></i> homozygote mutants were infected with 5×10<sup>4</sup> colony-forming units (CFUs) of <i>Mycobacterium bovis</i> (BCG), and 6 weeks later, mice were sacrificed and the degree of infection was assessed by determination of spleen CFUs (left) and splenomegaly (spleen index; right). (<b>B</b>) <i>Jak3<sup>W81R</sup></i> homozygotes and B6 controls were infected by aerosol inoculation of 50 CFUs/lung of <i>Mycobacterium tuberculosis</i> H37Rv, and monitored for survival. (<b>C</b>) <i>Jak3<sup>W81R</sup></i> homozygotes, <i>Jak3<sup>W81R/+</sup></i> heterozygotes and B6 controls were infected by oral gavage with 3×10<sup>8</sup> CFUs of <i>Citrobacter rodentium</i> strain DBS100. Mice were monitored for survival for 30 days post infection. Data represent two independent experiments.</p

    Phenotypic expression of resistance to cerebral malaria in mice from pedigree 48.

    No full text
    <p>G3 and F2 mice homozygote (P48/P48) or heterozygote (P48/+) for the B6-derived mutant central chromosome 8 were identified by genotyping, and were subjected to several analyses, along with parental C57BL/6J, C57BL/10J and 129S1/SvlmJ controls. (<b>A</b>) Macroscopic examination of thymus from control and mutants showing severely atrophied thymus in homozygote mutants (representative of 5 mice per group). (<b>B</b>) FACS density plots of different cell populations in thymus (top), spleen (middle) and bone marrow (bottom) stained for CD4, CD8, CD19, and CD117. The position of the different cell lineages in the scatter plots are identified at the extreme right panel and their numbers are expressed as a percentage (± SE; <i>n</i> = 5 mice per group) of total cells in this tissue. (<b>C</b>) Flow cytometric analysis of immune cell lineage composition expressed as the absolute number (mean ± SD; <i>n</i> = 4–6 mice per group) of CD4<sup>+</sup> and CD8<sup>+</sup> single positive (SP), CD4<sup>+</sup>CD8<sup>+</sup> double positive (DP), B cells (CD19<sup>+</sup>), granulocytes (GR; Gr1<sup>+</sup>), hematopoietic stem cells (HSC; lineage<sup>−</sup>CD117<sup>+</sup>) and NK cells from 10<sup>5</sup> cells from spleen, thymus and bone marrow. Asterisks (one-way Anova test with Bonferroni post-test) identify significant differences between experimental animals and the C57BL/6J controls: *<i>p</i><0.05; ** <i>p</i><0.01; *** <i>p</i><0.001. Data are representative of two independent experiments.</p

    ENU-induced mutation that protects mice against <i>P. berghei</i> ANKA-induced cerebral malaria.

    No full text
    <p>(<b>A</b>) Breeding scheme for the production and identification of ENU-induced recessive mutations that convey protection against cerebral malaria (CM). Details of the breeding strategy are described in “<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0031012#s4" target="_blank">Materials and Methods</a>”. G3 and F2 pedigrees were phenotyped for the presence of animals resistant to <i>P. berghei</i>-induced CM. (<b>B</b>) Mice were infected with <i>P. berghei</i> ANKA (10<sup>6 </sup><i>P. berghei</i> ANKA-parasitized red blood cells, i.v.) and survival was monitored over time for individual pedigree P48 G3s (green and blue lines) derived from independent G2 females and a G1 male, and for susceptible C57BL/6J (B6, red line), and resistant mutant mouse strains bearing loss of function mutations in either the IFN-γ gene KO (IFN-γ KO, black line) or IRF8 (BXH2, brown line). Mice surviving past day 13 post infection were considered to be CM-resistant.</p

    Resistance to cerebral malaria in <i>Jak3<sup>W81R/−</sup></i> compound heterozygotes.

    No full text
    <p><i>Jak3<sup>W81R</sup></i> homozygotes were crossed to a mouse line bearing a null <i>Jak3</i> allele (<i>Jak3<sup>−/−</sup></i>) to create the <i>Jak3<sup>W81R/+</sup></i> compound heterozygotes. Single heterozygotes (<i>Jak3<sup>W81R/+</sup></i>, <i>Jak3<sup>+/−</sup></i>; <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0031012#pone-0031012-g005" target="_blank">Fig. 5A</a>), homozygotes (<i>Jak3<sup>W81R</sup></i>, <i>Jak3<sup>−/−</sup></i>; <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0031012#pone-0031012-g005" target="_blank">Fig. 5B</a>), compound heterozygotes (<i>Jak3<sup>W81R/−</sup></i>, gray line; <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0031012#pone-0031012-g005" target="_blank">Fig. 5A</a>) and C57BL/10J controls were infected with 10<sup>6 </sup><i>P. berghei</i> ANKA-parasitized RBCs and monitored for survival. All surviving mice were sacrificed on day 15 post-infection (experimental end-point). The data are representative of 2 independent experiments.</p

    Cell transfer experiments to induce susceptibility to cerebral malaria in <i>Jak3<sup>W81R</sup></i> mice.

    No full text
    <p>Wild type C57BL/10J (B10) mice, <i>Jak3<sup>W81R</sup></i> homozygote mutants (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0031012#pone-0031012-g006" target="_blank">Fig. 6A</a>), or <i>Jak3<sup>W81R</sup></i> homozygote mutants having received the indicated cell populations (20 million total splenocytes or 5 million each of the indicated cell population; 2 hrs prior to infection); (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0031012#pone-0031012-g006" target="_blank">Fig. 6A, 6B, 6C</a>) were infected i.v. with 10<sup>6 </sup><i>P. berghei</i> ANKA-parasitized RBCs, and survival from infection was monitored. Untreated wild type B10 and <i>Jak3<sup>W81R</sup></i> mutants were used as susceptible and resistant controls, respectively. All surviving mice were sacrificed on day 15 post-infection (experimental end-point). Data represent 2 independent experiments.</p
    corecore