18 research outputs found

    Human Papilloma Virus-Type 16 (HPV-16) & Human Herpes Virus-Type 8 (HHV-8) Infections Were Found to be Co-Existing Major Cancer-Contributing Factors. Individualized, Safe, Effective Treatment of Hopelessly Advanced Cancer Patients with Metastasis by Combining 4 Methods of Effective Treatment: 1) Optimal Dose of Vitamin D3 Using its 10 Unique Beneficial Effects, 2) Selective Drug Uptake Enhancement Method, 3) Stimulation of Newly Discovered Thymus Gland Representation Areas on the Back of Each Hand, & 4) Identification & Removal of Co-Existing Cancer-Contributing Factors

    No full text
    The article presents a study that described how human papillomavirus type 16 (HPV-16) and human herpes virus-type 8 infections are non-invasively detected and how to reduce the infections by the use of optimal dose of vitamin D3 to safe range. A method of determining individualized optimal dose of vitamin D3 is presented. The significance of co-existing the infections with single-cell parasite toxoplasma gondii is discussed. Example of the eggs with HPV-16 infection is shown

    p53-Independent Cell Cycle and Erythroid Differentiation Defects in Murine Embryonic Stem Cells Haploinsufficient for Diamond Blackfan Anemia-Proteins: <i>RPS19</i> versus <i>RPL5</i>

    Get PDF
    <div><p>Diamond Blackfan anemia (DBA) is a rare inherited bone marrow failure syndrome caused by ribosomal protein haploinsufficiency. DBA exhibits marked phenotypic variability, commonly presenting with erythroid hypoplasia, less consistently with non-erythroid features. The p53 pathway, activated by abortive ribosome assembly, is hypothesized to contribute to the erythroid failure of DBA. We studied murine embryonic stem (ES) cell lines harboring a gene trap mutation in a ribosomal protein gene, either <i>Rps19</i> or <i>Rpl5</i>. Both mutants exhibited ribosomal protein haploinsufficiency and polysome defects. <i>Rps19</i> mutant ES cells showed significant increase in p53 protein expression, however, there was no similar increase in the <i>Rpl5</i> mutant cells. Embryoid body formation was diminished in both mutants but nonspecifically rescued by knockdown of p53. When embryoid bodies were further differentiated to primitive erythroid colonies, both mutants exhibited a marked reduction in colony formation, which was again nonspecifically rescued by p53 inhibition. Cell cycle analyses were normal in Rps19 mutant ES cells, but there was a significant delay in the G2/M phase in the Rpl5 mutant cells, which was unaffected by p53 knockdown. Concordantly, <i>Rpl5</i> mutant ES cells had a more pronounced growth defect in liquid culture compared to the <i>Rps19</i> mutant cells. We conclude that the defects in our RPS19 and RPL5 haploinsufficient mouse ES cells are not adequately explained by p53 stabilization, as p53 knockdown appears to increase the growth and differentiation potential of both parental and mutant cells. Our studies demonstrate that gene trap mouse ES cells are useful tools to study the pathogenesis of DBA.</p></div

    The differentiation defects observed in <i>Rps19</i> and <i>Rpl5</i> mutants are nonspecifically rescued by p53 inhibition.

    No full text
    <p>(A) Western blot analyses were performed from mutant ES cells with antibodies against p53, using β-Actin as a loading control. ES cells from the <i>Rps19</i> mutant cells showed an increase in p53 expression. In contrast, the <i>Rpl5</i> mutant expressed no increase in p53, compared with the parental line. Image J quantification of western blots from 3 independent experiments demonstrated that the <i>Rps19</i> mutant ES cells had approximately a 4-fold increase in p53 protein compared to the wild type cells. (B) qRT-PCR performed on these ES cells showed an increase in p21 mRNA only in the <i>Rps19</i> mutant ES cells (3 independent experiments) while there was no similar increase in the <i>Rpl5</i> mutant ES cells (4 independent experiments). siRNA targeting p53 was used to transiently transfect ES cells 24 hours prior to primary differentiation, obtaining >90% p53 knockdown by qRT-PCR. Both mutants (C) showed a significant increase in EB formation with p53 knockdown (4 independent pooled experiments). This effect was nonspecific, as p53 knockdown of parental cells also increased EB formation (D). The primitive erythroid colony defect was partially compensated in the <i>Rps19</i> mutant after p53 inhibition and overcompensated in the <i>Rpl5</i> mutant (E) (3 independent pooled experiments). This augmentation of colony formation was again nonspecific, as there was an increase in primitive colony formation with p53 knockdown in both parental ES cells when compared with the control siRNA (3 independent pooled experiments for <i>Rpl5</i> parent and 4 independent experiments for <i>Rps19</i> parent) (F).</p
    corecore