10 research outputs found

    p53 contributes to DOX-induced cardiac injury.

    No full text
    <p>A. Representative electron micrographs demonstrating cardiomyocyte injury and morphometric quantification of subcellular injury in cardiomyocytes in WT and p53<sup>(−/−)</sup> mice. WT (a) and p53<sup>(−/−)</sup> (b) mice treated with saline demonstrated normal ultrastructure of heart muscle with cardiomyocytes showing numerous mitochondria (M), prominent myofilaments (Myo) and lipids (Lip). WT (c) and p53<sup>(−/−)</sup> (d) mice treated with DOX demonstrated the following pathologic changes: lysosomal degradation of mitochondria (asterisk), mitochondria with loss of cristae (arrow), peri-mitochondrial swelling (double arrow), disruption of mitochondrial membranes (arrowhead), and mitochondrial degeneration (d); scale bar, 1 µm. B. Pathologic changes were quantified for saline and DOX treated mice in the mitochondria and total cellular area (excluding nuclei). Quantification of damage for each specific compartment is expressed in the ratio of damaged area versus the total area. All graphs represent the Mean ± SEM for each group. *p<0.05 when compared with saline treated mice of the same genotype, and <sup>#</sup>p<0.05 when compared with WT mice treated with DOX. n = 6 or 7. C & D. Left ventricular function, assessed by percentage of ejection fraction (LV%EF) (C) and fractional shortening (LV%FS) (D), is expressed in percentage of change from basal levels caused by saline and DOX three days after injections for WT and p53<sup>(−/−)</sup> mice. All bar graphs represent the Mean ± SEM for each group. <sup>#</sup>p<0.05 when compared to all three other groups. n = 6–11.</p

    DOX induces HNE adduction with pJNK and Bcl-2 phosphorylation, and triggers autophagic response.

    No full text
    <p>A. PhosphoJNK was detected by immunoprecipitation using a polyclonal goat anti-HNE antibody followed by Western blot analysis with a rabbit polyclonal Thr183/Tyr185 phosphorylation specific JNK antibody. Quantitative analysis represents the Mean ± SEM (n = 5) in each group; *p<0.001 as compared to other groups. B. Western blot analysis of p-Bcl2 in cardiac mitochondria from WT and p53<sup>(−/−)</sup> mice treated with saline or DOX and quantitative analysis represents the Mean ± SEM n = 5 in each group; *p<0.001 as compared to other groups. C. Western blot analysis of Beclin1 in cardiac mitochondria from WT and p53<sup>(−/−)</sup> mice treated with saline or DOX and quantitative analysis represents the Mean ± SEM n = 5 in each group; *p<0.001 as compared to other groups. D. Co-immunoprecipitation of Beclin1 with Bcl-2 in cardiac mitochondria from WT and p53<sup>(−/−)</sup> mice treated with saline or DOX and quantitative analysis represents the Mean ± SEM n = 5 in each group; *p<0.01 as compared to other groups.</p

    Absence of p53 selectively reduces DOX-induced oxidative damage in the mitochondria.

    No full text
    <p>A. Representative immunogold electron micrographs using antibody against 4HNE protein adducts in cardiomyocytes. Electron dense beads indicate positive staining for 4HNE protein adducts (arrow). Left ventricular tissues from WT (a) and p53<sup>(−/−)</sup> (b) mice treated with saline demonstrate low labeling of 4HNE in mitochondria (M) and myofilaments (Myo) (a and b). Significant increase in labeling of 4HNE protein adducts was observed in both mitochondria and myofilaments of the WT mice hearts (c) but not in the p53<sup>(−/−)</sup> mice (d) treated with DOX. Scale bar, 1 µm. B, C, and D. 4HNE-immunoreactive protein adducts were quantified in cardiomyocyte cytoplasm (B), mitochondria (C) and nuclei (D) for both WT and p53<sup>(−/−)</sup> mice treated with saline and DOX. Labeling density is expressed in gold beads/µm<sup>2</sup>. All graphs represent the Mean ± SEM for each group. *p<0.05 when compared with saline treated mice of the same genotype, <sup>#</sup> p<0.05 when compared with WT mice similarly treated.</p

    p53 triggers autophagy and cell death.

    No full text
    <p>A. Biochemical marker for cardiac injury by autophagy. Western blot analysis of autophagy marker (LC3) expression in heart tissue homogenates from WT and p53<sup>(−/−)</sup> mice treated with saline or DOX. Quantitative analysis represents the Mean ± SEM n = 5 in each group; <sup>#</sup>p<0.001 as compared to other groups. B. Western blot analysis of gamma H2AX marker for cell death in heart tissue nuclear extracts from WT and p53<sup>(−/−)</sup> mice treated with saline or DOX. Quantitative analysis represents the Mean ± SEM n = 5 in each group. <sup>#</sup>p<0.001 as compared to other groups.</p

    p53 enhances DOX-induced cardiac injury, in part, via enhancement of oxidative stress in mitochondria.

    No full text
    <p>The absence of p53 selectively prevents DOX-mediated increase in oxidative stress indicators, including 4HNE-adducted proteins in mitochondria but not in the nucleus. DOX-induced p53-dependent increased oxidative stress in mitochondria is associated with sustained activation of JNK1 and subsequent phosphorylation of Bcl-2 and release of beclin from the Bcl-2-beclin complex resulting in detrimental effect of autophagy (cellular injury).</p

    Plasma TNF-α and Soluble TNF Receptor Levels after Doxorubicin with or without Co-Administration of Mesna—A Randomized, Cross-Over Clinical Study

    Get PDF
    <div><p>Purpose</p><p>Chemotherapy-induced cognitive impairment (CICI) is a common sequelae of cancer therapy. Recent preclinical observations have suggested that CICI can be mediated by chemotherapy-induced plasma protein oxidation, which triggers TNF-α mediated CNS damage. This study evaluated sodium-2-mercaptoethane sulfonate (Mesna) co-administration with doxorubicin to reduce doxorubicin-induced plasma protein oxidation and resultant cascade of TNF-α, soluble TNF receptor levels and related cytokines.</p><p>Methods</p><p>Thirty-two evaluable patients were randomized using a crossover design to receive mesna or saline in either the first or second cycle of doxorubicin in the context of a standard chemotherapy regimen for either non-Hodgkin lymphoma or breast cancer. Mesna (360 mg/m<sup>2</sup>) or saline administration occurred 15 minutes prior and three hours post doxorubicin. Pre-treatment and post-treatment measurements of oxidative stress, TNF-α and related cytokines were evaluated during the two experimental cycles of chemotherapy.</p><p>Results</p><p>Co-administration of mesna with chemotherapy reduced post-treatment levels of TNF-related cytokines and TNF-receptor 1 (TNFR1) and TNF-receptor 2 (TNFR2) (p = 0.05 and p = 0.002, respectively). Patients with the highest pre-treatment levels of each cytokine and its receptors were the most likely to benefit from mesna co-administration.</p><p>Conclusions</p><p>The extracellular anti-oxidant mesna, when co-administered during a single cycle of doxorubicin, reduced levels of TNF-α and its receptors after that cycle of therapy, demonstrating for the first time a clinical interaction between mesna and doxorubicin, drugs often coincidentally co-administered in multi-agent regimens. These findings support further investigation to determine whether rationally-timed mesna co-administration with redox active chemotherapy may prevent or reduce the cascade of events that lead to CICI.</p><p>Trial Registration</p><p>clinicaltrials.gov <a href="https://www.clinicaltrials.gov/ct2/show/NCT01205503?term=NCT01205503&rank=1" target="_blank">NCT01205503</a>.</p></div
    corecore